Next Article in Journal
Himalayan Ficus palmata L. Fruit Extract Showed In Vivo Central and Peripheral Analgesic Activity Involving COX-2 and Mu Opioid Receptors
Previous Article in Journal
SIRT1 Activation Enhancing 8,3′-Neolignans from the Twigs of Corylopsis coreana Uyeki
Previous Article in Special Issue
Removal of Methylene Blue and Congo Red Using Adsorptive Membrane Impregnated with Dried Ulva fasciata and Sargassum dentifolium
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advanced Applications for Protein and Compounds from Microalgae

by
Daniela Castiglia
1,†,
Simone Landi
2,*,† and
Sergio Esposito
2
1
Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
2
Department of Biology, University of Naples “Federico II”, Via Cinthia, 80126 Napoli, Italy
*
Author to whom correspondence should be addressed.
These authors have contributed equally to this work and share first authorship.
Plants 2021, 10(8), 1686; https://doi.org/10.3390/plants10081686
Submission received: 23 June 2021 / Revised: 10 August 2021 / Accepted: 13 August 2021 / Published: 16 August 2021

Abstract

:
Algal species still show unrevealed and unexplored potentiality for the identification of new compounds. Photosynthetic organisms represent a valuable resource to exploit and sustain the urgent need of sustainable and green technologies. Particularly, unconventional organisms from extreme environments could hide properties to be employed in a wide range of biotechnology applications, due to their peculiar alleles, proteins, and molecules. In this review we report a detailed dissection about the latest and advanced applications of protein derived from algae. Furthermore, the innovative use of modified algae as bio-reactors to generate proteins or bioactive compounds was discussed. The latest progress about pharmaceutical applications, including the possibility to obtain drugs to counteract virus (as SARS-CoV-2) were also examined. The last paragraph will survey recent cases of the utilization of extremophiles as bio-factories for specific protein and molecule production.

1. Introduction

In recent years, public opinion, research community, and commercial stakeholders paid great attention to green and sustainable biotechnologies; these are not anymore considered a luxury, but a necessity in all countries. Actually, the current world population is estimated in 7.8 billion people, and an increase is expected of about 2 billions in the next 30 years (UN; www.un.org; accessed on 1 July 2021); therefore, it is essential to develop sustainable biotechnologies and strategies to face the increased food demand with a reduced impact on natural environment. Plants and algae possess still untouched possibilities; thus, these organisms are emerging as formidable sustainable tools with a number of advantages over the conventional biotechnologies [1,2,3,4].
Since 1980s, plants emerged as one of the most promising production platforms for recombinant proteins and for the valuable production of bio-compounds [5,6]. The agricultural scale cultivation represents a cost-effective method to produce recombinant proteins as well as an innovative approach to reduce risks of product contamination [2]. Encouraging results in the production of recombinant proteins and molecules for pharmaceutical applications were preliminarily obtained using higher plants [7]. On the other hand, the intensive exploitation of cropland represents a severe constraint influencing the environment utilization, CO2 emission, chemical pollution, and water quality and availability [8].
At the same time, microalgae overcome a number of limitations of plants, emerging as effective biotechnological platforms and giving added value by the exploitation of bioactive compounds obtained from these biomasses [9,10]. Microalgae are characterized for being unicellular and their versatile metabolism, representing efficient and economic platforms to gather organics compounds such as proteins, lipids, pigments, sterols, and carbohydrates for a number of commercial applications such as nutraceutical, pharmaceutical and for biofuels [11,12]. This group includes photosynthetic prokaryotes (e.g., cyanobacteria) or eukaryotes, which are able to live in diversified environments [9]. As consequence, a number of microalgae species are currently cultivated for commercial and industrial aims, and more than 75% of this production is related to healthier supplements for human consumption [10].
Furthermore, these organisms can be utilized for advanced synthetic biology applications. This represents a new experimental field, connecting molecular biology with bioengineering [13]. Recent papers reported the ability of microalgae for the management of metabolism by synthetic biology approaches [13,14]. These studies further strengthen the idea of the utilization of microalgae as versatile systems for sustainable and effective biotechnological applications.
In this review, we analyze some recent examples about the utilization of photosynthetic organisms from marine environments, as bio-factories. Furthermore, we reported a survey of the latest and advanced applications of proteins and bioactive compounds from microalgae.

2. Microalgae Engineering to Obtain Platforms for Biocompounds Productions

In recent years macroalgae and microalgae highlighted a renewed commercial interest about the production of natural metabolites for nutraceutical, pharmaceutical, energetic, and food industries purposes [11,12,15,16,17].
Microalgae are considered promising and sustainable platforms to produce recombinant proteins; these aquatic organisms show several advantages with respect to higher plants, such as safety, low-cost production and the ability to be grown rapidly using freshwater, seawater and wastewaters, thus avoiding arable land [18].
Microalgae cultivations represent an alternative and renewable source to produce biofuels [19,20]. In fact, microalgae produce high levels of lipids, carbohydrates and proteins, which make them a feasible candidate for ecofriendly third-generation biofuels production [11,12]. Unfortunately, the actual costs of microalgal biofuels are still considered higher than conventional fossil oils, therefore, an experimental progress is still necessary to improve and modulate the bio-compounds content. In this context, genetic engineering represents an effective approach in order to obtain a better performance from microalgae [21] (Figure 1).
Thus, a number of strategies have been used to hyperaccumulate fatty acids and/or triacylglycerol (TAG) content (Table 1). Phaeodactylum tricornutum and Thalassiosira pseudonana engineered strains were obtained modifying TAG biosynthetic pathways. In this context, critical enzymes are glycerol-3-phosphate acyltransferase (GPAT), lysophosphatidic acid acyl- transferase (LPAAT), and acyl-CoA:diacylglycerol acyltransferase (DGAT). These enzymes regulate fatty acid and TAG biosynthetic pathways in plants and microalgae [22,23,24]. The overexpression of GPAT is effective to obtain suitable strains for biofuel production (Table 1). Both isoforms GPAT1 and GPAT2 were overexpressed in P. tricornutum resulting in a 2.9- and 2.3-fold change increased lipid content, respectively [19,20]. Interestingly, the overexpression of GPAT2 regulated the natural expression of LPAT and DGAT, causing an enhanced effect on total lipid content [20]. Interestingly, this strain was also reported as better tolerant to abiotic stress, namely salinity and chilling [20]. A similar approach was recently used in Neochloris oleoabundans where the plastidial NeoLPAAT1 and endoplasmic reticulum-located NeoDGAT2 were co-overexpressed. This strategy increased the TAG content of about 2-fold compared to wild type; furthermore, engineered strain reporting no effects on algal biomass [25]. Significant results were obtained using heterologous expression, mutagenesis in situ and knockdown strategies of genes related to the lipid biosynthetic pathway. Trentecoste et al. [26] and Barka et al. [27], developed specific lipase knockout strains of T. pseudonana and P. tricornutum, respectively. The suppression of TAG lipase in these two diatoms significantly increased the lipid content. Interestingly both did not show effects on growth [12,26,27]. TALEN-based genome editing technique was used in P. tricornutum strain [28] reporting modification in PtTES1, a hotdog-fold thioesterase involved in acyl-CoA hydrolysis. This modification lead to a 1.7-fold change increase of TAG content [28]. Successful editing results were obtained by using CRISP/CAS9 approach on P. tricornutum and T. pseudonana. These preliminary results opened new perspectives to obtain improved microalgae strains using this technique [29,30,31].
An increase in lipids content has been obtained by modifying different microalgal pathways. LEAFY COTYLEDON1 from Arabidopsis thalian (AtLEC1) represents a key transcription factor involved in plants lipid metabolism [32]. Liu et al. [33] demonstrated that the endogenous expression of AtLEC1 improves fatty acids and total lipids content in Chlorella ellipsoidea both under mixotrophic and autotrophic culture conditions. These differences between wild type and the engineered algal strain were related to an enhanced regulation of key enzymes namely ACCase, GPDH, PDAT1, and DGAT1 [33]. Another interesting example of the transfer of genes from A. thaliana in microalgae showed the heterologous overexpression of 1-deoxy-D-xylulose 5-phosphate synthase from A. thaliana in Nannochloropsis oceanica. This strain reported an improved CO2 fixation, thus increasing biomass, lipids, and carbohydrates productions upon different nutritional and stressed conditions [34]. Particularly, Nannochloropsis represented a further model used as platform for biofuel production from microalgae [34,35,36,37]. In the last year a number of papers have been published about the genetic manipulation of Nannochloropsis, particularly focused on the increase on lipid and fatty acids content. Different approaches, namely overexpression [36], endogenous expression [37], and insertional mutagenesis [38] were successfully used in Nannochloropsis. For example, Jeon et al. [36] overexpressed the NADP-dependent malic enzyme (NsME1) in Nannochloropsis salina. This enzyme is an important NADPH supplier, playing the central role in the C4-like cycle of microalgae. The overexpressed strain showed an increased fatty acid and lipids contents compared with wild type, up to 53% and 38%, respectively [36].
Recently, several papers reported the role of glucose-6-phosphate dehydrogenase (G6PDH) in lipogenesis regulation [38,39]. This enzyme catalyzes the first and rate-limiting reaction of the oxidative pentose pathway (OPPP) playing critical role(s) in both plants and algae physiology [40], particularly upon nutrient starvation and abiotic stresses [38,41]. G6PDH was efficiently used for homologous overexpression and heterologous expression in microalgae. The increased NAPDH content in P. tricornutum induced by PtG6PDH overexpression stimulated an increase fatty acid production upon control and high CO2 level conditions [39]. Similarly, heterologous expression of PtG6PDH and NoG6PDH (from Nannochloropsis oceanica) in Chlorella pyrenoidosa showed an increase in lipid content of about 1.79 and 3.09-fold-change, respectively.
Among lipids, long-chain polyunsaturated fatty acid (PUFA, ω-3) represent a major fraction of microalgae biomass such as Cyclotella cryptica, Nitzschia laevis, Navicula incerta, Navicula pelliculosa, and others [42,43,44]. Particularly, eicosapentaenoic acid (EPA 20:5) and docosahexaenoic acid (DHA 22:6) showed an interesting commercial potential [42,43,44]. Homologous overexpression and heterologous expression approaches have been investigated for an enhanced production of ω-3 species (Table 1). It is worth remembering that P. tricornutum represents a model organism for diatoms, mostly studied for biomass, lipid, and EPA production [43]. Recently, P. tricornutum overexpressing malonyl CoA-acyl carrier protein transacylase (PtMCAT) and fatty acid desaturase 5b (PtFAD5b) significantly improved the content of EPA [45]. Similarly, Hamilton et al. [46], obtained an engineered P.tricornutum strain expressing Δ-5 elongase from Ostreococcus tauri, and a glucose transporter from the moss Physcomitrella patens. This strain accumulated ω-3 long-chain polyunsaturated fatty acid and showed an unconventional ability to grow in the dark [46]. Recently, aquaculture industry has started to replace fish oil and fish meal with plant seed meals and vegetable oils but the critical constraints in the use of this type of oils is the presence of anti-nutritional factors namely phytate [47]. Pudney et al. [48], proposed the heterologous expression of phytases from Aspergillus niger and Escherichia coli to overcome this constraint. P. tricornutum strains expressing the phytases from these two species showed an enhanced content of EPA and DHA [48].
Successful manipulations were also obtained using red microalgae as well as Porphyridium sp. [49,50]. These are important marine single-cell economic microalgae suitable for large-scale production of phycoerythrin, polysaccharides and PUFAs [49]. There are few cases of Porphyridium sp. engineered strains, mainly obtained by Agrobacterium-mediated and biolistic transformation methods [46]. Interestingly, Jeon et al. [50], recently reported the use of CRISP/CAS9 approach to manipulate Porphyridium sp. increasing the production of phycoerythrin. The authors manipulated the chlorophyll synthase gene (CHS1), obtaining an improved strain able to produce an increased content of phycoerythrin from 44% to 63% depending by the light sources [50].
Analogous to phycoerythrin, astaxanthin is a desirable product for human consumption. This is mainly produced by the green microalga Haematococcus pluvialis in response to adverse conditions namely excess of light, salinity, and nutrient starvation [51]. A number of mutagenesis approaches were used on H. pluvialis using physical and chemical mutagens such as UV radiation, ethyl methanesulphonate (EMS), diethyl sulphate (DES) and other. In some case desirable improved strains were obtained showing high growth rates at the vegetative stage and high astaxanthin accumulation rates at the encystment stage [51]. In recent years, genetic engineering approaches were also obtained in H. pluvialis. For example, Waissman-Levy et al. [52], manipulated the nuclear genome of H. pluvialis by insertion of the hexose uptake protein (HUP1) gene from the green microalga Parachlorella kesslerii. The engineered strain was able to grow upon heterotrophy conditions in glucose-supplemented media [52].
Apart from the utilization of microalgae for the production of biomass, biofuels (e.g., lipids), and recombinant proteins, the exploitation of these organisms as makers of proteins and complex molecules of pharmacological interest is a major theme of current research. The flexible metabolism of photosynthetic cells represents an intriguing tool for the synthesis of those compounds requiring complex biosynthesis not always possible in a single cell compartment as in bacteria.
Table 1. List of microalgae engineered strains
Table 1. List of microalgae engineered strains
Host OrganismsGeneDonor OrganismEnzymeApproachEffectsReferences
Phaeodactylum tricornutumPtGPAT2P. tricornutumGlycerol-3-phosphate acyltransferase 2OverexpressionHyperaccumulation of TAG[20]
Phaeodactylum tricornutumPtPGMP. tricornutumPhosphoglucomutaseOverexpressionIncreased synthesis of chrysolaminarin[12]
Phaeodactylum tricornutumOsElo5O. tauriΔ5-elongaseEndogenous expressionImproved accumulation of EPA and DHA—dark cultivation[46]
Phaeodactylum tricornutumPpGTP. patensGlucose transporterEndogenous expressionImproved accumulation of EPA and DHA—dark cultivation[46]
Phaeodactylum tricornutumPtTL_TAG lipaseKnockdownHyperaccumulation of TAG[27]
Thalassiosira pseudonanaTpTL_TAG lipaseKnockdownHyperaccumulation of TAG[26]
Phaeodactylum tricornutumPtDGAT2BP. tricornutum2 acyl-CoA:diacylglycerol acyltransferaseEndogenous overexpressionIncreased DHA and TAG content[53]
Phaeodactylum tricornutumPtG6PDHP. tricornutumGlucose-6-phosphate dehydrogenaseOverexpressionEnhanced lipid and w-3 accumulation [39]
Phaeodactylum tricornutumAnPhyAA. nigerPhytaseEndogenous expressionImproved accumulation of EPA and DHA[48]
Phaeodactylum tricornutumEcAppAE. coliPhytaseEndogenous expressionImproved accumulation of EPA and DHA[48]
Phaeodactylum tricornutumPtMCATP. tricornutumMalonyl CoA-acyl carrier protein transacylaseOverexpressionImproved accumulation of EPA [45]
Phaeodactylum tricornutumPtFAD5bP. tricornutumFatty acid desaturase 5bOverexpressionImproved accumulation of EPA [45]
Phaeodactylum tricornutumPtGPAT1P. tricornutumGlycerol-3-phosphate acyltransferase OverexpressionIncreased lipid content[19]
Phaeodactylum tricornutumPtLPAAT1P. tricornutumLysophosphatidic acid acyltransferaseOverexpressionIncreased lipid content[19]
Phaeodactylum tricornutumPtPTPP. tricornutumPlastidial pyruvate transporterOverexpressionIncreased production of biomass and lipids[54]
Phaeodactylum tricornutumPtTES1_Hotdog-fold thioesteraseTALEN- mutagenesisHyperaccumulation of TAG[28]
Chlorella ellipsoideaAtLEC1A. thalianaLeafy cotyledon 1 transcription factorEndogenous expressionLipid overexpression [33]
Synechocystis sp.SaACCS. albaAcetyl-CoA carboxylaseEndogenous expressionLipid overexpression [55]
Chlamydomonas reinhardtiiCrGAPDHCh. reinhardtiiGlyceraldehyde-3-phosphate dehydrogenaseOverexpressionEnhanced carbon fixation [56]
Chlorella vulgarisCvNR_Nitrate reductaseCRISP-cas9 editing Reduced growth upon specific conditions[57]
Chlorella vulgarisCvAPT_Adenine phosphoribosyltransferaseCRISP-cas9 editing Reduced growth upon specific conditions[57]
Nannochloropsis oceanicaAtDXSA. thaliana1-deoxy-D-xylulose 5-phosphate synthaseEndogenous expressionImproved CO2 absorption, biomass and lipids[34]
Nannochloropsis salinaNsMEN. salinaMalic enzymeOverexpressionIncreased production of lipids and fatty acids[35]
Nannochloropsis salinaCrLCIACh. reinhardtiiAnion transporterEndogenous expressionIncreased production of fatty acids[36]
Nannochloropsis oceanicaNoAPL-Apetala 2 like transcription factorInsertional mutagenesisIncreased production of lipids[37]
Chlamydomonas reinhardtiiCrSBP1C. reinhardtiiSedoheptulose-1,7-bisphosphataseOverexpressionPhotosynthetic and growth rates improvement[58]
Neochloris oleoabundansNoGPAT_Glycerol-3-phosphate acyltransferase OverexpressionIncreased lipid content[25]
Neochloris oleoabundansNoLPAAT_Lysophosphatidic acid acyltransferaseOverexpressionIncreased lipid content[25]
Porphyridium purpureumPpCHS1_Chlorophyll synthaseCRISP-cas9 editingIncreased phycoerythrin content[50]
Haematococcus pluvialisPkHUP1Parachlorella kessleriiHexose uptake proteinEndogenous overexpressionDark cultivation[52]
Chlorella pyrenoidosaPtG6PDHP. tricornutumGlucose-6-phosphate dehydrogenaseEndogenous overexpressionIncreased lipid content[38]
Chlorella pyrenoidosaNoG6PDHN. oceanicaGlucose-6-phosphate dehydrogenaseEndogenous overexpressionIncreased lipid content[38]

3. Microalgae Engineering for the Production of Pharmacological Proteins

Microalgae represent an important biotechnology resource for the production of recombinant proteins for pharmacological applications. Successful results were reported by the genetic manipulation of microalgae for the production of antigens for vaccines, antibodies, immunotoxins, hormones, and antimicrobial agents [59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87]. Strategies and techniques have been developed for the exploitation of microalgae as expression systems for a number of advanced genetic toolkits [62,63,64,65].
Recombinant protein production was attained by engineering both nuclear and chloroplast microalgal genomes [66]. Chloroplast manipulation produces a high level of transgene expression [66,67,68,69,70,71], but the recombinant proteins obtained could be not always subjected to critical post-translation modifications (PTMs), thus affecting their activity and functionality [63]. On the other hand, nuclear transformation produced a lower protein accumulation, but retained PTMs, such as N-glycosylation [59,67]. Furthermore, nuclear manipulation provided tools for the targeting of proteins into specific subcellular compartments or secreting them into culture media [72] while regulatory elements (promoters and terminators) and selectable markers were tested to identify improved strategies for higher levels of recombinant protein [15,73].
Recently, the utilization of different engineering strategies, many biopharmaceutical compounds have been effectively produced in microalgae, and Chlamydomonas reinhardtii has been widely used for this type of applications.
A list of engineered strains is shown in Table 2. Antigens, antibiotics, and other bioactive molecules against animal pathogens have been produced. Examples include swine fever virus [74], white spot syndrome virus [64,72,73], lethal shrimp yellow head virus [77,78], Aeromonas hydrophila bacteria [79], and vibriosis disease [80].
In addition, recombinant proteins for therapy against human diseases and pathogens have been produced in microalgae. Dong et al. [60] obtained a Ch. reinhardtii strain able to produce the antimicrobial peptide Mytichitin-A. This peptide—not toxic for human cells—caused the inhibition of bacteria growth at the concentration of 60–80 ug/mL [57]. An interesting example of antimicrobial peptide against both fish and human pathogens is piscidin-4. This was recently produced in H. pluvialis by biolistic manipulation of chloroplast of this microalga [81].
Human interferon α (huIFN α) is widely used to treat cancer and some viral infections including hepatitis C; huIFN α was engineered and expressed in Ch. reinhardtii by Agrobacterium nuclear transformation [82]. The recombinant algae-expressed IFN α2 reduces virus propagation against the vesicular stomatitis virus [82].
Human interleukin-2 (iL-2), used in different cancers therapies, was successfully expressed in Ch. reinhardtii and Dunaliella salina producing about 0.94% and 0.59% of iL-2 respectively [68]. The stability of the recombinant algae-made interleukins was analyzed after five months from the transformation using ELISA assay, showing a stable and correct conformation of recombinant protein [68].
In recent years, microalgae were tested as suitable platforms for drugs production, even to counteract virus and other pathogens. Particularly, the receptor binding domain (RBD) of SARS-CoV-2, able to bind the angiotensin-converting 2 receptor (ACE2) to entry into human host cells was produced in Chlorella vulgaris and Ch. reinhardtii [83,84]. Thus, overexpressed RBD in algae was correctly folded, functional, and able to bind the human ACE2 receptor. These progresses open new pharmaceutical possibilities in the use of microalgae in the study and in the battle against the pandemic virus. Other aquatic microorganisms were similarly used as host organism in genetic engineering approaches for pharmaceutical purposes. Interesting results were obtained using the non-photosynthetic marine microorganism Schizochytrium sp. (Phylum: Heterokonta; Order: Labyrinthulales) [85,86,87].
Zika virus (ZIKV) infection cause severe symptoms including fever, rash, conjunctivitis, muscle and joint pains. ZK microalgae-made vaccine was tested by oral administration in mice showing immunogenic potential and inducing both systemic (IgG) and mucosal (IgA) humoral responses. Promising antigens against ZIKV were obtained by Agrobacterium-mediated transformation in Schizochytrium sp. [85].
RAGE (receptor of advanced glycation end-products) is a receptor related to development and progression of Alzheimer’s disease (AD), its expression was increased in the brain in AD patients, inducing the release of proinflammatory mediators; thus, RAGE is considered an interesting target for AD therapy [86]. RAGE fused to adjuvant carrier (LTB) was produced successfully in Schizochytrium sp. Furthermore, algae-based recombinant vaccine showed high stability to heat treatment (up to 60 °C), therefore, this was proposed as a possible vaccine for the treatment of AD [86].
CelTOS (cell-traversal protein for ookinetes and sporozoites) is crucial protein, which mediated the malaria virus infection. Shamriz et al. [65] manipulated C. reinhardtii chloroplast obtaining a CelTOS antigen from Plasmodium falciparum developing a sensitive ELISA test to identify malaria infection.
One of the great challenges is the development of an algae-based vaccines against cancer. Hernandez-Ramirez et al. [87] expressed in Schizochytrium sp. multi-epitope protein (BCB) against breast cancer (BC). They selected different B cell epitopes developed from tumor associated antigens (TAAs) recognized by the immune system. Biomass from transgenic algae expressing BCB were administered to mice, recording specific antibodies production [87].
Another goal is the production of monoclonal antibodies from microalgae. Nowadays, monoclonal antibodies represent an innovative solution against cancer and other human diseases. Vanier et al. [71] expressed human anti-hepatitis B (HBsAg) antibodies in the diatom P. tricornutum, reporting the production of fully assembled IgG antibody secreted in the culture medium. Microalgal antibodies were tested on THP-1 cell line to evaluate Fcγ receptor interactions, which are specific cell-superficial receptor for IgG, binding to human Fcγ receptor, and inducing an immune response. Monoclonal IgG antibodies against Marburg Virus (MARV), responsible for hemorrhagic fever with high fatality rates in Western Africa has been produced [88]. MARV sequences for both heavy and light chains from murine hybridoma were expressed in P. tricornutum, and the recombinant protein was secreted in the culture medium at concentration of 1300 ng/mL. ELISA demonstrated an efficient binding for algal antibodies [88].
In recent years, researchers explored the possible applications of extracellular vesicles (EVs) as drug carriers [89,90,91]. EVs are small cell-derived membranous particles delimited by a lipid-bilayer, containing proteins, lipids, and nucleic acids. EVs are released by cells into the environment under physiological and pathological conditions for cell communication [89,90]. EVs showed many interesting characteristics for potential use as therapeutic molecules and drug delivery systems. Indeed, EVs showed low immunological response, low toxicity, and these vesicles are able to cross various barriers, and showed the ability to transport and protect their cargo [89,92]. EVs have been evaluated for their potential to produce nanomedicine-based therapeutics; thus, the research has been focused on the possibility to use engineered EVs as vectors in specific therapies [91]. EVs from 18 different strains of microalgae have been isolated and characterized [92]. The EVs were approximately 90–160 nm in size, and microalgae-based EVs protein showed positive reactivity for the target protein of the EVs. Moreover, EVs did not show toxicity on either normal and cancer mammalian cell lines. These results suggest that EVs produced by microalgae could represent promising tools for therapeutic purposes.
It should be noted that all these studies require critical control points, in order to carefully regulate growth conditions and the activation of specific metabolic pathways. Therefore, the possibility to utilize extremophilic organisms as advanced bioreactors able to synthetize compounds upon particular conditions is a promising strategy for these aims.
Table 2. List of pharmaceutical products obtained by engineered microalgae
Table 2. List of pharmaceutical products obtained by engineered microalgae
Organism hostProductApplicationTransformation MethodLocalizationOutcomeExpression YieldsReferences
Ch. reinhardtiiE2 protein Swine fever virus vaccineBiolisticChloroplastStrong immunogenic response in mice1.5–2% TSP[74]
Ch. reinhardtiiVP28White spot syndrome virus vaccineGlass beadChloroplastShrimp survival up to 87%ND[75]
D. salinaVP28White spot syndrome virus vaccineGlass beadChloroplast59% protection rate78 mg/100 culture[76]
Ch. reinhardtiiVP28 Chloroplast Up to 10% TSP[69]
Ch. reinhardtiiAntiviral dsRNAYellow head virus RNAi-based vaccineGlass beadsChloroplastReduced mortalityUp to 16 ng dsRNA/L culture[77]
Ch. reinhardtiidsRNA-YHVYellow head virus antiviralGlass beadsNucleus22% Shrimp survival 45 ng/100-mL culture[78]
Chlorella sp.AMPs Scy-hepcA. hydrophila bacteria oral antibioticsElectroporationNucleusIn vitro inhibitory effects on A. hydrophila; in vivo S. macrocephalus Up to 0.078%TSP[79]
H. pluvialisPiscidin-4 peptideAntibacterial activityBiolisticChloroplastStable expressionND[81]
Nannochloropsis spOmpK fragment geneVibrio species oral vaccine NucleusFifth generation stable immunogenic peptide productionND[80]
Ch. reinhardtiiMytichitin-A peptideAntibacterial activityElectroporationNucleusHigh inhibition of bacteria growth (MIC assays); No toxicity on HEK293 cells.0.28% TSP[60]
Ch. reinhardtiiSARS-CoV-2-RBDAntigen proteins against SARS-CoV-2Geminiviral vector TransientELISA assay showed specific binding with the anti-RBD antibody1.14 µg/g[83]
Ch. vulgarisSARS-CoV-2-RBDAntigen proteins against SARS-CoV-2Geminiviral vector TransientELISA assay showed specific binding with the anti-RBD antibody1.161 µg/g[83]
Ch. reinhardtiiSARS-CoV-2-RBDAntigen proteins against SARS-CoV-2ElectroporationTransientBind human ACE2 receptor0.1% TSP[84]
Ch. reinhardtiiHuman Interferon-αChronic viral diseases and cancers treatAgrobacteriumNucleusIn vivo e in vitro antitumoral activity, anticancer proprieties, antiviral activityND[82]
Ch. reinhardtii,Human interleukin-2Interleukin productionAgrobacteriumNucleusELISA assay showed biological activity, high stabilityUp to 0.94% TSP[72]
D. salina; C. vulgarisHuman interleukin-2Interleukin productionAgrobacteriumNucleusELISA assay showed biological activity, high stabilityUp to 0.59% TSP[72]
Schizochytrium sp.ZK antigenZika virus oral vaccineAlgevir technologyTransientIgG and IgA production Up to 365.3 μg g−1 FW[85]
Schizochytrium sp.LTB:RAGE antigenAlzheimer’s disease vaccineAlgevir technologyTransientELISA assay showed high stability up to of 60 °CUp to 380 μg g−1 FW [86]
Ch. reinhardtii,PfCelTOS antigenMalaria antigen for diagnosis testsBiolisticChloroplastStable expressionND[64]
Schizochytrium sp.Multiepitope protein (BCB)Breast cancer vaccineAlgevir technologyTransientTumor cell line 4T1 reactivity; IgG production in mice immunized with BCBUp to 637 μg/g[87]
P. tricornutumHepatitis B AntibodyAntibodies against Hepatitis BBiolisticNucleusBinding FcγRI2 mg/L[71]
P. tricornutumMonoclonal antibodiesAntibodies against Marburg virusBiolisticNucleusElisa assay showed binding efficiency1300 ng/ml[88]
T. pseudonanaAntibody for EA1Biosensor for anthracis detectionBiolisticNucleusDetection of detected EA1 epitope in lysed sporesND[61]

4. Extremophilic Microalgae as Bioreactors

A renewed attention has been focused on organisms from extreme environments [93,94]. This is particularly true for microalgae, which show a wide range survival capabilities under extreme and stress conditions, namely hypersalinity, high and low temperatures or toxic heavy metals levels [94,95,96,97]. The use of extremophilic microalgae showed notable benefits for various applications [93,98]. Commercial cultivations of microalgae are usually obtained by growth in photobioreactors and open ponds [54,98]. On the other hand, industrial biotechnology requires elevated temperatures, difficult sterilization procedures, and expensive downstream processing which benefit from the use of thermophilic microalgae [99,100]. Extreme operating conditions tolerate by specific microalgae strains were proposed to improve these industrial processes, thus overcoming contaminations, loss of biomass, meteorological constraints, and others [94,101,102]. Example of biotechnological applications are the cultivation of Chlorella to mitigate the effects of industrial pollution, by absorbing CO2; the use of Arthrospira platensis for bioremediation of contaminated effluents; the utilization of a number of microalgal species for biofuel productions [100].
Thermophilic microalgae cultivated in large-scale open ponds are able to produce bulk amount of lipids, which can be utilised for biodiesel production technology [103]. Particularly, in recent years increasing attention were posed on the regulation of fatty acid dehydrogenase (FAD) in freeze resistant microalgae [59,104,105]. This studies contributed to both elucidation of the regulation of algal membrane assembly, and fatty acid metabolism. Promising results were obtained using the Δ12FAD from Chlamydomonas sp. ICE-L [59] and the Δ5FAD from Lobosphaera incise [105]. Both enzymes showed cold-resistant mechanisms to adapt their biochemical functions. Δ12FAD from Chlamydomonas sp. ICE-L contributes to membrane fluidity for adaptation to Antarctic extreme environment; Δ5FAD contributes to arachidonic acid synthesis, thus avoiding the risk of photodamage upon chilling. A number of snow algae were tested for their ability to produce lipid species in low temperatures [106]. These analysis reported an enriched production of unsaturated fatty acyl chains, especially C18:1n-9 and C18:3n-3, thus indicating these species as good candidates to improve the yields of microalgal biomass and oil products at low temperatures [106].
Thermophillic microalgae are studied for the applications of phycobiliproteins [107], a group of water-soluble proteins linked to chromophores, involved in light-harvesting processes. Phycobiliproteins find use in a wide range of commercial purposes (e.g., colorant for food and textile industries) and the exploitation of these proteins would give additional value [97]. A number of industrial processes require temperatures exceeding the stability of proteins from mesophiles [107]. Phycocianins from the cyanobacterium A. platensis are actually produced as dye in food industries but high temperatures (62 °C) limited their use [108]. Different authors proposed the use of phycocianins from the thermoacidophilic red alga Cyanidioschyzon merolae to overcome this problem [97,109]. The temperatures and pH tolerability of phycocianins from Cyanidioschyzon merolae were recently characterized by Yoshida et al. [96], confirming their thermotolerant properties. Similar evaluations were reported for phycobiliproteins in textile industry where the use of mesophiles proteins at high temperatures resulted in a decrease of color intensity [97].
A number of extromophilic microalgae were recently characterized for the ability in the production of bio-compounds with high economic interest [98]. For example, the snow green algae namely Chlamydomonas nivalis, Raphidonema sp., and Chloromonas sp. showed ability in the production of astaxanthin, α-Tocopherol, xanthophylles, and glycerol [110,111,112]. On the other hand, the heat tolerant green alga Desmodesmus and the acidophilic red alga Galdieria sulphuraria were tested for the production of lutein and blue pigment phycocianin, respectively [113,114]. These results demonstrate the unexplored potential represented by microalgae from extreme environments, thus encouraging the characterization of new species.

5. Conclusions

Here, we reviewed the remarkable value of photosynthetic organisms and their potential emerged in recent years. Through the last decades, there has been significant research into the exploitaion of microalgae as sustainable resources for the production of high-value products and proteins. It is clear that microalgae can play a crucial role in modern biotechnological processes role at energetic, pharmaceutical, and nutraceutical levels. It is worth to point out that even if the current knowledge about natural and engineering microalgae is broad and wide, present notions are still not sufficient, and significant efforts are required to reach the goals. The cloning technologies to obtain suitable platform or bio-factories to produce proteins or metabolites of interest are now widely accessible and at least readily implementable by modern technologies as well as synthetic biology approaches. It is now time to further expand this research and the applications of microalgae in order to develop more refined tools and strategies for an efficient sustainable and ecological breakthrough.

Author Contributions

D.C., S.L., and S.E. conceived the idea and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fischer, R.; Emans, N.; Schuster, F.; Hellwig, S.; Drossard, J. Towards molecular farming in the future: Using plant-cell-suspension cultures as bioreactors. Biotechnol. Appl. Biochem. 1999, 30, 109–112. [Google Scholar]
  2. Fischer, R.; Stoger, E.; Schillberg, S.; Christou, P.; Twyman, R.M. Plant-based production of biopharmaceuticals. Curr. Opin. Plant Biol. 2004, 7, 152–158. [Google Scholar] [CrossRef]
  3. Lau, O.S.; Sun, S.S.M. Plant seeds as bioreactors for recombinant protein production. Biotechnol. Adv. 2009, 27, 1015–1022. [Google Scholar] [CrossRef]
  4. Liang, Z.C.; Liang, M.H.; Jiang, J.G. Transgenic microalgae as bioreactors. Crit. Rev. Food Sci. Nutr. 2020, 60, 3195–3213. [Google Scholar] [CrossRef]
  5. De Zoeten, G.; Penswick, J.; Horisberger, M.; Ahl, P.; Schultze, M.; Hohn, T. The expression, localization, and effect of a human interferon in plants. Virology 1989, 172, 213–222. [Google Scholar] [CrossRef]
  6. Hiatt, A.; Cafferkey, R.; Bowdish, K. Production of antibodies in transgenic plants. Nature 1989, 342, 76–78. [Google Scholar] [CrossRef]
  7. Shanmugaraj, B.; Bulaon, C.J.I.; Phoolcharoen, W. Plant molecular farming: A viable platform for recombinant biopharmaceutical production. Plants 2020, 9, 842. [Google Scholar] [CrossRef]
  8. Foley, J.A.; Defries, R.; Asner, G.P.; Barford, C.; Bonan, G.; Carpenter, S.R.; Chapin, F.S.; Coe, M.T.; Daily, G.C.; Gibbs, H.K.; et al. Global consequences of land use. Science 2005, 309, 570–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Hamed, I. The evolution and versatility of microalgal biotechnology: A review. Compr. Rev. Food Sci. Food Saf. 2016, 15, 1104–1123. [Google Scholar] [CrossRef] [PubMed]
  10. Hashemian, M.; Ahmadzadeh, H.; Hosseini, M.; Lyon, S.; Pourianfar, H. Production of microalgae-derived high-protein biomass to enhance food for animal feedstock and human consumption. Adv. Bioprocess. Altern. Fuels Biobased Chem. Bioprod. 2019, 393–405. [Google Scholar] [CrossRef]
  11. Gallo, C.; Landi, S.; d’Ippolito, G.; Nuzzo, G.; Manzo, E.; Sardo, A.; Fontana, A. Diatoms synthesize sterols by inclusion of animal and fungal genes in the plant pathway. Sci. Rep. 2020, 10. [Google Scholar] [CrossRef]
  12. Yang, R.; Wei, D.; Xie, J. Diatoms as cell factories for high-value products: Chrysolaminarin, eicosapentaenoic acid, and fucoxanthin. Crit. Rev. Biotechnol. 2020, 40, 993–1009. [Google Scholar] [CrossRef]
  13. Zhao, C.; Yaschenko, A.; Alonso, J.M.; Stepanova, A.N. Leveraging synthetic biology approaches in plant hormone research. Curr. Opin. Plant. Biol. 2021, 60. [Google Scholar] [CrossRef]
  14. George, J.; Kahlke, T.; Abbriano, R.M.; Kuzhiumparambil, U.; Ralph, P.J.; Fabris, M. Metabolic engineering strategies in diatoms reveal unique phenotypes and genetic configurations with implications for algal genetics and synthetic biology. Front. Bioeng. Biotechnol. 2020, 8. [Google Scholar] [CrossRef] [PubMed]
  15. Rosales-Mendoza, S.; García-Silva, I.; González-Ortega, O.; Sandoval-Vargas, J.M.; Malla, A.; Vimolmangkang, S. The potential of algal biotechnology to produce antiviral compounds and biopharmaceuticals. Molecules 2020, 25, 49. [Google Scholar] [CrossRef] [PubMed]
  16. Vella, F.M.; Sardo, A.; Gallo, C.; Landi, S.; Fontanta, A.; d’Ippolito, G. Annual outdoor cultivation of the diatom Thalassiosira weissflogii: Productivity, limits and perspectives. Algal Res. 2019, 42. [Google Scholar] [CrossRef]
  17. Landi, S.; Esposito, S. Bioinformatic characterization of sulfotransferase provides new insights for the exploitation of sulfated polysaccharides in Caulerpa. Int. J. Mol. Sci. 2020, 21, 681. [Google Scholar] [CrossRef] [PubMed]
  18. Fabris, M.; Abbriano, R.M.; Pernice, M.; Sutherland, D.L.; Commault, A.S.; Hall, C.C.; Labeeuw, L.; McCauley, J.I.; Kuzhiuparambil, U.; Ray, P.; et al. Emerging technologies in algal biotechnology: Toward the establishment of a sustainable, algae-based bioeconomy. Front. Plant Sci. 2020, 11, 279. [Google Scholar] [CrossRef] [Green Version]
  19. Wang, X.; Dong, H.P.; Wei, W.; Balamurugan, S.; Yang, W.D.; Liu, J.S.; Li, H.Y. Dual expression of plastidial GPAT1 and LPAT1 regulates triacylglycerol production and the fatty acid profile in Phaeodactylum tricornutum. Biotechnol. Biofuels 2018, 11. [Google Scholar] [CrossRef]
  20. Wang, X.; Liu, S.F.; Li, R.Y.; Yang, W.D.; Liu, J.S.; Lin, C.S.K.; Balamurugan, S.; Li, H.Y. TAG pathway engineering via GPAT2 concurrently potentiates abiotic stress tolerance and oleaginicity in Phaeodactylum tricornutum. Biotechnol. Biofuels 2020, 13. [Google Scholar] [CrossRef]
  21. Sun, X.M.; Ren, L.J.; Zhao, Q.Y.; Ji, X.J.; Huang, H. Enhancement of lipid accumulation in microalgae by metabolic engineering. BBA Lipids 2019, 4, 552–566. [Google Scholar] [CrossRef] [PubMed]
  22. Li-Beisson, Y.; Shorrosh, B.; Beisson, F.; Andersson, M.X.; Arondel, V.; Bates, P.D.; Baud, S.; Bird, D.; Debono, A.; Durrett, T.P.; et al. Acyl-lipid metabolism. Arab. Book 2010, 8, e0133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zulu, N.N.; Zienkiewicz, K.; Vollheyde, K.; Feussner, I. Current trends to comprehend lipid metabolism in diatoms. Prog. Lipid Res. 2018, 70, 1–16. [Google Scholar] [CrossRef]
  24. Li-Beisson, Y.; Thelen, J.J.; Fedosejevs, E.; Harwood, J.L. The lipid biochemistry of eukaryotic algae. Prog. Lipid Res. 2019, 74, 31–68. [Google Scholar] [CrossRef] [PubMed]
  25. Chungjatupornchai, W.; Fa-Aroonsawat, S. Enhanced triacylglycerol production in oleaginous microalga Neochloris oleoabundans by co-overexpression of lipogenic genes: Plastidial LPAAT1 and ER-located DGAT2. J. Biosci. Bioeng. 2021, 131, 124–130. [Google Scholar] [CrossRef] [PubMed]
  26. Trentacoste, E.M.; Shrestha, R.P.; Smith, S.R.; Glé, C.; Hartmann, A.; Hildebrand, M.; Gerwick, W.H. Metabolic engineering of lipid catabolism increases microalgal lipid accumulation without compromising growth. Proc. Natl. Acad. Sci. USA 2013, 110, 19748–19753. [Google Scholar] [CrossRef] [Green Version]
  27. Barka, F.; Angstenberger, M.; Ahrendt, T.; Lorenzen, W.; Bode, H.B.; Büchel, C. Identification of a triacylglycerol lipase in the diatom Phaeodactylum tricornutum. Biochim. Biophys. Acta 2016, 1861, 239–248. [Google Scholar] [CrossRef]
  28. Hao, X.; Luo, L.; Jouhet, J.; Rébeillé, F.; Maréchal, E.; Hu, H.; Pan, Y.; Tan, X.; Chen, Z.; You, L.; et al. Enhanced triacylglycerol production in the diatom Phaeodactylum tricornutum by inactivation of a Hotdog-fold thioesterase gene using TALEN-based targeted mutagenesis. Biotechnol. Biofuels 2018, 11, 312. [Google Scholar] [CrossRef]
  29. Stukenberg, D.; Zauner, S.; Dell’Aquila, G.; Maier, U.G. Optimizing CRISPR/Cas9 for the diatom Phaeodactylum tricornutum. Front. Plant Sci. 2018, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Hopes, A.; Nekrasov, V.; Kamoun, S.; Mock, T. Editing of the urease gene by CRISPR-Cas in the diatom Thalassiosira pseudonana. Plant Methods. 2016, 12. [Google Scholar] [CrossRef] [Green Version]
  31. Moosburner, M.A.; Gholami, P.; McCarthy, J.K.; Tan, M.; Bielinski, V.A.; Allen, A.E. Multiplexed knockouts in the model diatom Phaeodactylum by episomal delivery of a selectable Cas9. Front. Microbiol. 2020, 11. [Google Scholar] [CrossRef] [Green Version]
  32. Mu, J.; Tan, H.; Zheng, Q.; Fu, F.; Liang, Y.; Zhang, J.; Yang, X.; Wang, T.; Chong, K.; Wang, X.-J.; et al. LEAFY COTYLEDON1 is a key regulator of fatty acid biosynthesis in Arabidopsis. Plant Physiol. 2008, 148, 1042–1054. [Google Scholar] [CrossRef] [Green Version]
  33. Liu, X.; Zhang, D.; Zhang, J.; Chen, Y.; Liu, X.; Fan, C.; Wang, R.R.; Hou, Y.; Hu, Z. Overexpression of the transcription factor AtLEC1 significantly improved the lipid content of Chlorella ellipsoidea. Front. Bioeng. Biotechnol. 2021, 9. [Google Scholar] [CrossRef] [PubMed]
  34. Han, X.; Song, X.; Li, F.; Lu, Y. Improving lipid productivity by engineering a control-knob gene in the oleaginous microalga Nannochloropsis oceanica. Metab. Eng. Commun. 2020, 11, e00142. [Google Scholar] [CrossRef]
  35. Jeon, S.; Koh, H.G.; Cho, J.M.; Kang, N.K.; Chang, Y.K. Enhancement of lipid production in Nannochloropsis salina by overexpression of endogenous NADP-dependent malic enzyme. Algal Res. 2021. [Google Scholar] [CrossRef]
  36. Vikramathithan, J.; Hwangbo, K.; Lim, J.M.; Lim, K.M.; Kang, D.Y.; Park, Y.; Jeong, W.J. Overexpression of Chlamydomonas reinhardtii LCIA (CrLCIA) gene increases growth of Nannochloropsis salina CCMP1776. Algal Res. 2020. [Google Scholar] [CrossRef]
  37. Südfeld, C.; Hubáček, M.; Figueiredo, D.; Naduthodi, M.I.S.; van der Oost, J.; Wijffels, R.H.; Barbosa, M.J.; D’Adamo, S. High-throughput insertional mutagenesis reveals novel targets for enhancing lipid accumulation in Nannochloropsis oceanica. Metab. Eng. 2021, 66, 239–258. [Google Scholar] [CrossRef] [PubMed]
  38. Xue, J.; Chen, T.T.; Zheng, J.W.; Balamurugan, S.; Liu, Y.H.; Yang, W.D.; Liu, J.S.; Li, H.Y. Glucose-6-phosphate dehydrogenase from the oleaginous microalga Nannochloropsis uncovers its potential role in promoting lipogenesis. Biotechnol. J. 2020, 15, e1900135. [Google Scholar] [CrossRef]
  39. Wu, S.; Gu, W.; Huang, A.; Li, Y.; Kumar, M.; Lim, P.E.; Huan, L.; Gao, S.; Wang, G. Elevated CO2 improves both lipid accumulation and growth rate in the glucose-6-phosphate dehydrogenase engineered Phaeodactylum tricornutum. Microb. Cell Fact. 2019, 18, 161. [Google Scholar] [CrossRef]
  40. Esposito, S. Nitrogen assimilation, abiotic stress and glucose 6-phosphate dehydrogenase: The full circle of ductants. Plants 2016, 11, 24. [Google Scholar] [CrossRef]
  41. Landi, S.; De Lillo, A.; Nurcato, R.; Grillo, S.; Esposito, S. In-field study on traditional Italian tomato landraces: The constitutive activation of the ROS scavenging machinery reduces effects of drought stress. Plant Physiol. Biochem. 2017, 118, 150–160. [Google Scholar] [CrossRef] [PubMed]
  42. Borowitzka, M.A. High-value products from microalgae-their development and commercialisation. J. Appl. Phycol. 2013, 25, 743–756. [Google Scholar] [CrossRef]
  43. Rodolfi, L.; Zittelli, G.C.; Bassi, N.; Padovani, G.; Biondi, N.; Bonini, G.; Tredici, M.R. Microalgae for oil: Strain selection. Induction of lipid synthesis and outdoor mass cultivation in a low-cost photobioreactor. Biotechnol. Bioeng. 2009, 102, 100–112. [Google Scholar] [CrossRef] [PubMed]
  44. Cupo, A.; Landi, S.; Morra, S.; Nuzzo, G.; Gallo, C.; Manzo, E.; Fontana, A.; d’Ippolito, G. Autotrophic vs. Heterotrophic cultivation of the marine diatom cyclotella cryptica for EPA production. Mar. Drugs 2021, 19, 355. [Google Scholar] [CrossRef]
  45. Wang, X.; Luo, S.W.; Luo, W.; Yang, W.D.; Liu, J.S.; Li, H.Y. Adaptive evolution of microalgal strains empowered by fulvic acid for enhanced polyunsaturated fatty acid production. Bioresour. Technol. 2019, 277, 204–210. [Google Scholar] [CrossRef] [PubMed]
  46. Hamilton, M.L.; Powers, S.; Napier, J.A.; Sayanova, O. Heterotrophic production of omega-3 long-chain polyunsaturated fatty acids by trophically converted marine diatom Phaeodactylum tricornutum. Mar. Drugs 2016, 14, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Nasopoulou, C.; Zabetakis, I. Benefits of fish oil replacement by plant originated oils in compounded fish feeds. A review. LWT Food Sci. Technol. 2012, 47, 217–224. [Google Scholar] [CrossRef]
  48. Pudney, A.; Gandini, C.; Economou, C.K.; Smith, R.; Goddard, P.; Napier, J.A.; Spicer, A.; Sayanova, O. Multifunctionalizing the marine diatom Phaeodactylum tricornutum for sustainable co-production of omega-3 long chain polyunsaturated fatty acids and recombinant phytase. Sci. Rep. 2019, 9, 11444. [Google Scholar] [CrossRef]
  49. Li, S.; Ji, L.; Shi, Q.; Wu, H.; Fan, J. Advances in the production of bioactive substances from marine unicellular microalgae Porphyridium spp. Bioresour. Technol. 2019, 292, 122048. [Google Scholar] [CrossRef]
  50. Jeon, M.S.; Han, S.I.; Jeon, M.; Choi, Y.E. Enhancement of phycoerythrin productivity in Porphyridium purpureum using the clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 ribonucleoprotein system. Bioresour. Technol. 2021, 330, 124974. [Google Scholar] [CrossRef] [PubMed]
  51. Li, X.; Wang, X.; Duan, C.; Yi, S.; Gao, Z.; Xiao, C.; Agathos, S.N.; Wang, G.; Li, J. Biotechnological production of astaxanthin from the microalga Haematococcus pluvialis. Biotechnol. Adv. 2021, 43, 107602. [Google Scholar] [CrossRef]
  52. Waissman-Levy, N.; Leu, S.; Khozin-Goldberg, I.; Boussiba, S. Manipulation of trophic capacities in Haematococcus pluvialis enables low-light mediated growth on glucose and astaxanthin formation in the dark. Algal Res. 2019, 40, 101497. [Google Scholar] [CrossRef]
  53. Haslam, R.P.; Hamilton, M.L.; Economou, C.K.; Smith, R.; Hassall, K.L.; Napier, J.A.; Sayanova, O. Overexpression of an endogenous type 2 diacylglycerol acyltransferase in the marine diatom Phaeodactylum tricornutum enhances lipid production and omega-3 long-chain polyunsaturated fatty acid content. Biotechnol. Biofuels 2020, 13, 87. [Google Scholar] [CrossRef] [PubMed]
  54. Seo, S.; Kim, J.; Lee, J.W.; Nam, O.; Chang, K.S.; Jin, E. Enhanced pyruvate metabolism in plastids by overexpression of putative plastidial pyruvate transporter in Phaeodactylum tricornutum. Biotechnol. Biofuels 2020, 13, 120. [Google Scholar] [CrossRef]
  55. Fathy, W.; Essawy, E.; Tawfik, E.; Khedr, M.; Abdelhameed, M.S.; Hammouda, O.; Elsayed, K. Recombinant overexpression of the Escherichia coli acetyl-CoA carboxylase gene in Synechocystis sp. boosts lipid production. J. Basic Microbiol. 2021. [Google Scholar] [CrossRef]
  56. Zhu, Z.; Cao, H.; Li, X.; Rong, J.; Cao, X.; Tian, J. A carbon fixation enhanced Chlamydomonas reinhardtii strain for achieving the double-win between growth and biofuel production under non-stressed conditions. Front. Bioeng. Biotechnol. 2021, 8, 603513. [Google Scholar] [CrossRef]
  57. Kim, J.; Chang, K.S.; Lee, S.; Jin, E. Establishment of a genome editing tool using CRISPR-Cas9 in Chlorella vulgaris UTEX395. Int. J. Mol. Sci. 2021, 22, 480. [Google Scholar] [CrossRef] [PubMed]
  58. Hammel, A.; Sommer, F.; Zimmer, D.; Stitt, M.; Mühlhaus, T.; Schroda, M. Overexpression of sedoheptulose-1,7-bisphosphatase enhances photosynthesis in Chlamydomonas reinhardtii and has no effect on the abundance of other calvin-benson cycle enzymes. Front. Plant Sci. 2020, 11, 868. [Google Scholar] [CrossRef]
  59. He, Y.; Zheng, Z.; An, M.; Chen, H.; Qu, C.; Liu, F.; Wang, Y.; Miao, J.; Hou, X. Molecular cloning and functional analysis of a Δ12-fatty acid desaturase from the Antarctic microalga Chlamydomonas sp. ICE-L. 3 Biotech 2019, 9, 328. [Google Scholar] [CrossRef] [PubMed]
  60. Dong, B.; Cheng, R.Q.; Liu, Q.Y.; Wang, J.; Fan, Z.C. Multimer of the antimicrobial peptide mytichitin-A expressed in Chlamydomonas reinhardtii exerts a broader antibacterial spectrum and increased potency. J. Biosci. Bioeng. 2018, 125, 175–179. [Google Scholar] [CrossRef]
  61. Ford, N.R.; Xiong, Y.; Hecht, K.A.; Squier, T.C.; Rorrer, G.L.; Roesijadi, G. Optimizing the design of diatom biosilica-targeted fusion proteins in biosensor construction for Bacillus anthracis detection. Biology 2020, 9, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Sproles, A.E.; Fields, F.J.; Smalley, T.N.; Le, C.H.; Badary, A.; Mayfield, S.P. Recent advancements in the genetic engineering of microalgae. Algal Res. 2021. [Google Scholar] [CrossRef]
  63. Barolo, L.; Abbriano, R.M.; Commault, A.S.; George, J.; Kahlke, T.; Fabris, M.; Padula, M.P.; Lopez, A.; Ralph, P.J.; Pernice, M. Perspectives for glyco-engineering of recombinant biopharmaceuticals from microalgae. Cells 2020, 9, 633. [Google Scholar] [CrossRef] [Green Version]
  64. Shamriz, S.; Ofoghi, H. Expression of recombinant PfCelTOS antigen in the chloroplast of Chlamydomonas reinhardtii and its potential use in detection of malaria. Mol. Biotechnol. 2019, 61, 102–110. [Google Scholar] [CrossRef]
  65. Yan, N.; Fan, C.; Chen, Y.; Hu, Z. The potential for microalgae as bioreactors to produce pharmaceuticals. Int. J. Mol. Sci. 2016, 17, 962. [Google Scholar] [CrossRef] [Green Version]
  66. Jensen, P.E.; Scharff, L.B. Engineering of plastids to optimize the production of high-value metabolites and proteins. Curr. Opin. Biotechnol. 2019, 59, 8–15. [Google Scholar] [CrossRef]
  67. Barrera, D.J.; Rosenberg, J.N.; Chiu, J.G.; Chang, Y.N.; Debatis, M.; Ngoi, S.M.; Chang, J.T.; Shoemaker, C.B.; Oyler, G.A.; Mayfield, S.P. Algal chloroplast produced camelid VH H antitoxins are capable of neutralizing botulinum neurotoxin. Plant Biotechnol. J. 2015, 13, 117–124. [Google Scholar] [CrossRef] [Green Version]
  68. Hempel, F.; Lau, J.; Klingl, A.; Maier, U.G. Algae as protein factories: Expression of a human antibody and the respective antigen in the diatom Phaeodactylum tricornutum. PLoS ONE 2011, 6, e28424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Surzycki, R.; Greenham, K.; Kitayama, K.; Dibal, F.; Wagner, R.; Rochaix, J.D.; Ajam, T.; Surzycki, S. Factors effecting expression of vaccines in microalgae. Biologicals 2009, 37, 133–138. [Google Scholar] [CrossRef]
  70. Sun, M.; Qian, K.; Su, N.; Chang, H.; Liu, J.; Shen, G. Foot-and-mouth disease virus VP1 protein fused with cholera toxin B subunit expressed in Chlamydomonas reinhardtii chloroplast. Biotechnol. Lett. 2003, 25, 1087–1092. [Google Scholar] [CrossRef] [PubMed]
  71. Vanier, G.; Stelter, S.; Vanier, J.; Hempel, F.; Maier, U.G.; Lerouge, P.; Ma, J.; Bardor, M. Alga-made anti-hepatitis B antibody binds to human Fcγ receptors. Biotechnol. J. 2018, 13, e1700496. [Google Scholar] [CrossRef]
  72. Dehghani, J.; Adibkia, K.; Movafeghi, A.; Pourseif, M.M.; Omidi, Y. Designing a new generation of expression toolkits for engineering of green microalgae; robust production of human interleukin-2. Bioimpacts 2020, 10, 259–268. [Google Scholar] [CrossRef] [PubMed]
  73. Velmurugan, N.; Deka, D. Transformation techniques for metabolic engineering of diatoms and haptophytes: Current state and prospects. Appl. Microbiol. Biotechnol. 2018, 102, 4255–4267. [Google Scholar] [CrossRef]
  74. He, D.M.; Qian, K.X.; Shen, G.F.; Zhang, Z.F.; Li, Y.N.; Su, Z.L.; Shao, H.B. Recombination and expression of classical swine fever virus (CSFV) structural protein E2 gene in Chlamydomonas reinhardtii chroloplasts. Colloids Surf. B Biointerfaces 2007, 55, 26–30. [Google Scholar] [CrossRef]
  75. Kiataramgul, A.; Maneenin, S.; Purton, S.; Areechon, N.; Hirono, I.; Brocklehurst, T.W.; Unajak, S. An oral delivery system for controlling white spot syndrome virus infection in shrimp using transgenic microalgae. Aquaculture 2020, 521, 735022. [Google Scholar] [CrossRef]
  76. Feng, S.; Feng, W.; Zhao, L.; Gu, H.; Li, Q.; Shi, K.; Guo, S.; Zhang, N. Preparation of transgenic Dunaliella salina for immunization against white spot syndrome virus in crayfish. Arch. Virol. 2014, 159, 519–525. [Google Scholar] [CrossRef] [PubMed]
  77. Charoonnart, P.; Worakajit, N.; Zedler, J.A.Z.; Meetam, M.; Robinson, C.; Saksmerprome, V. Generation of microalga Chlamydomonas reinhardtii expressing shrimp antiviral dsRNA without supplementation of antibiotics. Sci. Rep. 2019, 9, 3164. [Google Scholar] [CrossRef] [Green Version]
  78. Somchai, P.; Jitrakorn, S.; Thitamadee, S.; Meetam, M.; Saksmerprome, V. Use of microalgae Chlamydomonas reinhardtii for production of double-stranded RNA against shrimp virus. Aquac. Rep. 2016, 3, 178–183. [Google Scholar] [CrossRef] [Green Version]
  79. He, Y.; Peng, H.; Liu, J.; Chen, F.; Zhou, Y.; Ma, X.; Chen, H.; Wang, K. Chlorella sp. transgenic with Scy-hepc enhancing the survival of Sparus macrocephalus and hybrid grouper challenged with Aeromonas hydrophila. Fish Shellfish Immunol. 2018, 73, 22–29. [Google Scholar] [CrossRef] [PubMed]
  80. Abidin, A.A.Z.; Othman, N.A.; Yusoff, F.M.; Yusoff, Z.N.B. Determination of transgene stability in Nannochloropsis sp. transformed with immunogenic peptide for oral vaccination against vibriosis. Aquac. Int. 2021, 29, 477–486. [Google Scholar] [CrossRef]
  81. Wang, K.; Cui, Y.; Wang, Y.; Gao, Z.; Liu, T.; Meng, C.; Qin, S. Chloroplast genetic engineering of a unicellular green alga Haematococcus pluvialis with expression of an antimicrobial peptide. Mar. Biotechnol. 2020, 22, 572–580. [Google Scholar] [CrossRef] [PubMed]
  82. El-Ayouty, Y.; El-Manawy, I.; Nasih, S.; Hamdy, E.; Kebeish, R. Engineering Chlamydomonas reinhardtii for expression of functionally active human interferon-α. Mol. Biotechnol. 2019, 61, 134–144. [Google Scholar] [CrossRef]
  83. Malla, A.; Rosales-Mendoza, S.; Phoolcharoen, W.; Vimolmangkang, S. Efficient transient expression of recombinant proteins using DNA viral vectors in freshwater microalgal species. Front. Plant. Sci. 2021, 12, 650820. [Google Scholar] [CrossRef]
  84. Berndt, A.; Smalley, T.; Ren, B.; Badary, A.; Sproles, A.; Fields, F.; Torres-Tiji, Y.; Heredia, V.; Mayfield, S. Recombinant production of a functional SARS- CoV-2 spike receptor binding domain in the green algae Chlamydomonas reinhardtii. bioRxiv 2021. [Google Scholar] [CrossRef]
  85. Márquez-Escobar, V.A.; Bañuelos-Hernández, B.; Rosales-Mendoza, S. Expression of a Zika virus antigen in microalgae: Towards mucosal vaccine development. J. Biotechnol. 2018, 282, 86–91. [Google Scholar] [CrossRef]
  86. Ortega-Berlanga, B.; Bañuelos-Hernández, B.; Rosales-Mendoza, S. Efficient expression of an Alzheimer’s disease vaccine candidate in the microalga Schizochytrium sp. using the Algevir system. Mol. Biotechnol. 2018, 60, 362–368. [Google Scholar] [CrossRef] [PubMed]
  87. Hernández-Ramírez, J.; Wong-Arce, A.; González-Ortega, O.; Rosales-Mendoza, S. Expression in algae of a chimeric protein carrying several epitopes from tumor associated antigens. Int. J. Biol. Macromol. 2020, 147, 46–52. [Google Scholar] [CrossRef] [PubMed]
  88. Hempel, F.; Maurer, M.; Brockmann, B.; Mayer, C.; Biedenkopf, N.; Kelterbaum, A.; Becker, S.; Maier, U.G. From hybridomas to a robust microalgal-based production platform: Molecular design of a diatom secreting monoclonal antibodies directed against the Marburg virus nucleoprotein. Microb. Cell Fact. 2017, 16, 131. [Google Scholar] [CrossRef] [PubMed]
  89. Pedrioli, G.; Piovesana, E.; Vacchi, E.; Balbi, C. Extracellular vesicles as promising carriers in drug delivery: Considerations from a cell biologist’s perspective. Biology 2021, 10, 376. [Google Scholar] [CrossRef]
  90. Doyle, L.M.; Wang, M.Z. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
  91. Villata, S.; Canta, M.; Cauda, V. EVs and bioengineering: From cellular products to engineered nanomachines. Int. J. Mol. Sci. 2020, 21, 6048. [Google Scholar] [CrossRef]
  92. Picciotto, S.; Barone, M.E.; Fierli, D.; Aranyos, A.; Adamo, G.; Božič, D.; Romancino, D.P.; Stanly, C.; Parkes, R.; Morsbach, S.; et al. Isolation of extracellular vesicles from microalgae: Towards the production of sustainable and natural nanocarriers of bioactive compounds. Biomater. Sci. 2021, 9, 2917–2930. [Google Scholar] [CrossRef]
  93. Raddadi, N.; Cherif, A.; Daffonchio, D.; Neifar, M.; Fava, F. Biotechnological applications of extremophiles, extremozymes and extremolytes. Appl. Microbiol. Biotechnol. 2015, 99, 7907–7913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Malavasi, V.; Soru, S.; Cao, G. Extremophile microalgae: The potential for biotechnological application. J. Phycol. 2020, 56, 559–573. [Google Scholar] [CrossRef]
  95. Vinogradova, O.M.; Darienko, T.M. Terrestrial algae of hypersaline environments of the Central Syvash islands (Kherson Region, Ukraine). Biologia 2008, 63, 813–823. [Google Scholar] [CrossRef] [Green Version]
  96. Yoshida, C.; Murakami, M.; Niwa, A.; Takeya, M.; Osanai, T. Efficient extraction and preservation of thermotolerant phycocyanins from red alga Cyanidioschyzon merolae. J. Biosci. Bioeng. 2021, 131, 161–167. [Google Scholar] [CrossRef]
  97. Puzorjov, A.; McCormick, A.J. Phycobiliproteins from extreme environments and their potential applications. J. Exp. Bot. 2020, 71, 3827–3842. [Google Scholar] [CrossRef]
  98. Varshney, P.; Mikulic, P.; Vonshak, A.; Beardall, J.; Wangikar, P.P. Extremophilic micro-algae and their potential contribution in biotechnology. Bioresour. Technol. 2015, 184, 363–372. [Google Scholar] [CrossRef]
  99. Chen, G.Q.; Jiang, X.R. Next generation industrial biotechnology based on extremophilic bacteria. Curr. Opin. Biotechnol. 2018, 50, 94–100. [Google Scholar] [CrossRef] [PubMed]
  100. Patel, A.; Matsakas, L.; Rova, U.; Christakopoulos, P. A perspective on biotechnological applications of thermophilic microalgae and cyanobacteria. Bioresour. Technol. 2019, 278, 424–434. [Google Scholar] [CrossRef]
  101. Souza, L.D.; Simioni, C.; Bouzon, Z.L.; Schneider, R.C.; Gressler, P.; Miotto, M.C.; Rossi, M.J.; Rörig, L.R. Morphological and ultrastructural characterization of the acidophilic and lipid-producer strain Chlamydomonas acidophila LAFIC-004 (Chlorophyta) under different culture conditions. Protoplasma 2017, 254, 1385–1398. [Google Scholar] [CrossRef]
  102. Soru, S.; Malavasi, V.; Caboni, P.; Concas, A.; Cao, G. Behavior of the extremophile green alga Coccomyxa melkonianii SCCA 048 in terms of lipids production and morphology at different pH values. Extremophiles 2019, 23, 79–89. [Google Scholar] [CrossRef]
  103. Abu-Ghosh, S.; Dubinsky, Z.; Banet, G.; Iluz, D. Optimizing photon dose and frequency to enhance lipid productivity of thermophilic algae for biofuel production. Bioresour. Technol. 2018, 260, 374–379. [Google Scholar] [CrossRef]
  104. de Jaeger, L.; Springer, J.; Wolbert, E.J.H.; Martens, D.E.; Eggink, G.; Wijffels, R.H. Gene silencing of stearoyl-ACP desaturase enhances the stearic acid content in Chlamydomonas reinhardtii. Bioresour. Technol. 2017, 245, 1616–1626. [Google Scholar] [CrossRef]
  105. Zorin, B.; Pal-Nath, D.; Lukyanov, A.; Smolskaya, S.; Kolusheva, S.; Didi-Cohen, S.; Boussiba, S.; Cohen, Z.; Khozin-Goldberg, I.; Solovchenko, A. Arachidonic acid is important for efficient use of light by the microalga Lobosphaera incisa under chilling stress. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2017, 1862, 853–868. [Google Scholar] [CrossRef]
  106. Hulatt, C.J.; Berecz, O.; Egeland, E.S.; Wijffels, R.H.; Kiron, V. Polar snow algae as a valuable source of lipids? Bioresour. Technol. 2017, 235, 338–347. [Google Scholar] [CrossRef]
  107. Pagels, F.; Guedes, A.C.; Amaro, H.M.; Kijjoa, A.; Vasconcelos, V. Phycobiliproteins from cyanobacteria: Chemistry and biotechnological applications. Biotechnol Adv. 2019, 37, 422–443. [Google Scholar] [CrossRef]
  108. Chaiklahan, R.; Chirasuwan, N.; Bunnag, B. Stability of phycocyanin extracted from Spirulina sp.: Influence of temperature, pH and preservatives. Process Biochem. 2012, 47, 659–664. [Google Scholar] [CrossRef]
  109. Rahman, D.Y.; Sarian, F.D.; van Wijk, A.; Martinez-Garcia, M.; van der Maarel, M.J.E.C. Thermostable phycocyanin from the red microalga Cyanidioschyzon merolae, a new natural blue food colorant. J. Appl. Phycol. 2017, 29, 1233–1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Remias, D.; Lütz-Meindl, U.; Lütz, C. Photosynthesis, pigments and ultrastructure of the alpine snow alga Chlamydomonas nivalis. Eur. J. Phycol. 2005, 40, 259–268. [Google Scholar] [CrossRef]
  111. Leya, T.; Rahn, A.; Lütz, C.; Remias, D. Response of arctic snow and permafrost algae to high light and nitrogen stress by changes in pigment composition and applied aspects for biotechnology. FEMS Microbiol. Ecol. 2009, 67, 432–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Seckbach, J.; Oren, A.; Stan-lotter, H. Polyextremophiles Life under Multiple Forms of Stress, Cellular Origin, Life in Extreme Habitats and Astrobiology; Springer: Dordrecht, The Netherlands, 2013. [Google Scholar]
  113. Sloth, J.K.; Wiebe, M.G.; Eriksen, N.T. Accumulation of phycocyanin in heterotrophic and mixotrophic cultures of the acidophilic red alga Galdieria sulphuraria. Enzym. Microb. Technol. 2006, 38, 168–175. [Google Scholar] [CrossRef]
  114. Xie, Y.; Ho, S.H.; Chen, C.N.N.; Chen, C.Y.; Ng, I.S.; Jing, K.J.; Chang, J.S.; Lu, Y. Phototrophic cultivation of a thermo-tolerant Desmodesmus sp. for lutein production: Effects of nitrate concentration, light intensity and fed-batch operation. Bioresour. Technol. 2013, 144, 435–444. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Recognized approaches (Transformation techniques, promoters, selective markers) used to microalgae genetic engineering.
Figure 1. Recognized approaches (Transformation techniques, promoters, selective markers) used to microalgae genetic engineering.
Plants 10 01686 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Castiglia, D.; Landi, S.; Esposito, S. Advanced Applications for Protein and Compounds from Microalgae. Plants 2021, 10, 1686. https://doi.org/10.3390/plants10081686

AMA Style

Castiglia D, Landi S, Esposito S. Advanced Applications for Protein and Compounds from Microalgae. Plants. 2021; 10(8):1686. https://doi.org/10.3390/plants10081686

Chicago/Turabian Style

Castiglia, Daniela, Simone Landi, and Sergio Esposito. 2021. "Advanced Applications for Protein and Compounds from Microalgae" Plants 10, no. 8: 1686. https://doi.org/10.3390/plants10081686

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

Article Metrics

Back to TopTop