You are currently viewing a new version of our website. To view the old version click .
Marine Drugs
  • Review
  • Open Access

24 April 2022

Emerging Trends in Genetic Engineering of Microalgae for Commercial Applications

,
and
1
Laboratory of Natural Substances, Biomolecules and Biotechnological Applications, University of Oum El Bouaghi, Oum El Bouaghi 04000, Algeria
2
College of Marine Sciences, Hainan University, Haikou 570228, China
3
College of Agricultural Sciences, Panzhihua University, Panzhihua 617000, China
*
Author to whom correspondence should be addressed.

Abstract

Recently, microalgal biotechnology has received increasing interests in producing valuable, sustainable and environmentally friendly bioproducts. The development of economically viable production processes entails resolving certain limitations of microalgal biotechnology, and fast evolving genetic engineering technologies have emerged as new tools to overcome these limitations. This review provides a synopsis of recent progress, current trends and emerging approaches of genetic engineering of microalgae for commercial applications, including production of pharmaceutical protein, lipid, carotenoids and biohydrogen, etc. Photochemistry improvement in microalgae and CO2 sequestration by microalgae via genetic engineering were also discussed since these subjects are closely entangled with commercial production of the above mentioned products. Although genetic engineering of microalgae is proved to be very effective in boosting performance of production in laboratory conditions, only limited success was achieved to be applicable to industry so far. With genetic engineering technologies advancing rapidly and intensive investigations going on, more bioproducts are expected to be produced by genetically modified microalgae and even much more to be prospected.

1. Introduction

Microalgae (including cyanobacteria) are predominantly unicellular photosynthetic organisms which constitute the base of aquatic food webs. As an ancestor of plants with billions of years of evolutionary history, they distinctively adapted to extreme habitats and developed massive phylogenetic and biochemical diversity [1,2]. They have colonized almost all biotopes and been acclimated to severe environments, living in salt marshes, deserts or environments with very low light [3]. Not only do some microalgae tolerate hostile environmental conditions but also need these conditions to thrive. As a consequence of diurnal, seasonal, vertical and geographic variations plus fluctuations in nutrient availability, temperature, light and other factors, the distribution and metabolic activities of microalgae and the biomolecules they produce may be significantly more heterogeneous than previously believed [2].
In recent decades, microalgae emerged as new, attractive, promising and scalable platforms for the production of some biomolecules [4]. Among these biomolecules, some primary and secondary metabolites, such as carotenoids and proteins, were already commercialized as customer products, which intrigues further R&D to prospect more bioproducts from microalgae [5]. The efforts of developing these bioproducts from microalgae are underway, but with a number of constraints including: low product yield, slow growth rates, high cost of production facility, frequent contamination, high cost of harvesting, high energy consumption of cell lysis and complicated process of extraction of the desired metabolites [6]. The primary focus of the process development is to increase the yield of these biomolecules and the growth rates of microalgae. The evolution of genetic engineering technologies of microalgae progressed considerably for the past decades, and the research results obtained helped to boost the economic viability of commercial microalgal productions. This purpose of this study is to summarize the recent development and the emerging trends of molecular biotechnology applied to microalgae to increase the cells’ growth performance as well as to improve the synthesis rates of primary and secondary metabolites from this valuable group of organisms.

1.1. Phylogenetical and Biochemical Diversity of Microalgae

Microalgae could be prokaryotic or eukaryotic and are phylogenetically very diverse. The eukaryotic microalgae might be traced back 1.9 × 109 years and are much younger than the cyanobacteria with 2.7 × 109 years of phylogenetic history [1]. After the progenitor of the algae arose through an endosymbiosis with cyanobacterium, two evolutionary lines appeared simultaneously, namely green and red algae. These two lines acquired a number of various distinctive characteristics. Further groups of algae did not appear until much later through the secondary endosymbiosis, while the green and red algae are transformed into plastids in a eukaryotic host. This process gave rise to the heterokont algae, the Dinoflagellates, the Cryptophytes and the Euglenida [1]. Because of several biochemical and cellular disparities, two principal groups of green microalgae are identified: the Chlorophyta and the Conjugaphyta. While the second group is nearly five times larger than the Chlorophyta, none of the Conjugaphyta is yet applied for biotechnological engineering [1].
Algal diversity is very large and represents an almost unexploited natural resource. In accordance with some early assessments, there might be tens of thousands to millions of microalgae species, only a tiny fraction of which were isolated or described [2]. A more accurate number of estimation might be 72,500 species, which is still roughly twice as that of plants [7]. In recent decades, huge microalgal collections were generated by researchers from various countries. An example is the collection of freshwater microalgae from the Coimbra College (Coimbra, Portugal), considered to be among the largest in the world, with over 4000 different strains with more than 1000 species. The collection mirrors the broad array of microalgae ready to be used in various applications [8]. Microalgae are therefore a group that is not well studied in biotechnology terms. Of the great variety of microalgal species thought to exist, just some thousands of strains are preserved in collections around the world, with just some hundreds explored for chemical composition and only tens cultivated in manufacturing (tons per year) [9].
The wide variability of microalgae provides a large range of potential applications as source of feeding, stock of biomaterials and bioreactor of biotechnologically important molecules [8,10]. This diverse phylogeny is also expressed in a large biochemical diversity of pigments, photosynthetic storage products, mucilage materials, fatty acids, oils and hydrocarbons, sterols and secondary bioactive compounds, relating secondary metabolites [2,11]. Except for having a specific compound available that makes these organisms interesting, their diversity and the possibility of harvesting and cultivating under various conditions allows for using them as natural bioreactors for producing multiple chemicals through biorefinery or integrated processes [11].

1.2. Various Applications of Microalgae

The use of microalgae by humans was practiced since many years as food, feed, medicines and fertilizers. In the 14th century, the Aztecs harvested Arthrospira, formerly spirulina, a cyanobacteria in Lake Texcoco. They used tecuitlatl (cake made with spirulina) as a main dietary component. Most likely, the utilization of this cyanobacterium as food in Chad happened at the same period, or even earlier to the Kanem Empire (9th century) [9,12]. In a world with limited resources (energy, water and arable land, etc.) and intensifying anthropogenic pressure on the environment, the improvement of biotechnological processes to supply sustainable energy and renewable biomaterials from cleaner industrial processes constitutes a key challenge.
The genetic, phylogenetic and compositional nature of microalgal diversity is considerable, which makes them appealing for bioprospecting and the eventual industrial utilization of various biomolecules [13]. Nowadays, the principal industrial products from green microalgae are carotenoids and biomass for food, health and aquaculture [14]. These products are obtained from a restricted number of species; cyanobacterium from the Arthrospira genus constitutes 50% of world production, followed by green microalgae from the genera Chlorella, Dunaliella, Haematococcus, Nannochloropsis and the diatom Odontella [13]. Microalgae are usually selected based on both their growth properties and their aptitude to produce considerable amounts of specific metabolites. The efficiency of biomass production is a pivotal element of financial success in most of today’s commercial systems [14].
The production of such microalgal compounds depends on the variability of process and environmental conditions, which all have a direct effect on the value and quality of the target products. Quality control must therefore meet industry standards relating to toxicity and safety, the concentration of antioxidants and product characteristics relevant to marketability such as the taste and smell [14]. The market of microalgae applications remains in development, and the exploitation will spread into new areas. Given the huge microalgal diversity and modern technological progress of biotechnology, these microorganisms constitute so far major potential resources for novel products and applications [1]. Furthermore, as being broadly unexplored, the microalgae offer a major opportunity for discovery. The rediscovery rate (finding metabolites already described) should be superior to that of other groups of better-studied organisms.

1.3. Recent Development of Microalgal Biotechnology

During the current decades, the microalgal biotechnology acquired extensive and significant importance. Applications vary from biomass production for feeding to useful products for environmental applications [1]. Microalgal biotechnology is currently going through an unparalleled interest and investment worldwide [8]. For the last decade, researchers and industry developed various microalgal cultivation technologies that are in use today to produce biomass. Along with conventional fermentation reactors, two techniques are frequently applied in microalgae cultivation, specifically, outdoor and indoor production within photobioreactors. It is therefore important to develop and improve the diverse microalgae culture technologies to minimize production costs [11]. Growth rates significantly fluctuate between species and greatly depend on cultivation methods, especially the design of photobioreactors and the bioprocess conditions. In outdoor cultivation, extremophile species are preferred as they minimize the risk of contamination by competing organisms [14].
Recently, there has been a fast and significant progress in molecular engineering. As a result, the genetic technology applied to microalgae extended from the conventional process to systematic and synthetic regulation of metabolic pathways (Figure 1). Systems biology (including genomics, transcriptomics, proteomics and metabolomics) use two or more omics methods in order to define and to study a complex biological system. Synthetic biology aims to use standard biopartition as a basis for a new and fast biological implementation (e.g., transcriptome analysis combined to genome sequence might serve to predict promoter potency and operability). Metabolic engineering seeks to construct a strong host for the high production level of the chemical substance through genome editing and design of genetic circuits to redirect molecular flow of some metabolites [15].
Figure 1. Genetic technology applied to microalgae to optimize production of target metabolites.

3. Opportunities, Challenges and Prospects

According to recent literature reviews, the whole genomic sequencing of more than 40 microalgal species have been completed, and much more are underway [117]. Recent approaches of next-generation sequencing technologies including sequencing-by-synthesis, single-molecule real-time sequencing and pyrosequencing found wide applications in the functional genomic research of microalgae, such as profiling of mRNAs and small RNAs, recognizing transcription factor regions, genome annotation, detection of aberrant transcription, mutation mapping and polymorphic identifying of noncoding RNA [128,129]. All these techniques were implemented to facilitate sequencing of several microalgal species and offered novel and rapid ways for genome-wide characterization. In parallel, several bioinformatics methodologies were developed to assemble, map and evaluate huge quantities of relatively or extremely short nucleotide sequences generated with next-generation sequencing data [130]. Emerging comparative genomics allow the investigation of the variability and operability of algal genes by intra- and inter-species comparisons [131]. Three recent reviews summarize the multi-omics analysis, recent genome-scale metabolic simulation, genetic engineering and evolutionary engineering of microalgae previously reported and might be able to provide detailed information and knowledge in this fields [15,132,133].
Three organelles available to microalgae, the nucleus, mitochondria and chloroplast, could be subjected to genetic engineering for commercial production of microalgal biomass or metabolites. Plenty of investigations were reported to be able to improve targeted metabolite production by engineering single genes that are crucial for the synthesis of those metabolites. The alternative approach is transcriptional engineering (TE), which regulates a wide range of genes involving multiple metabolic pathways simultaneously [134]. TFs control specific target gene expression by binding specific DNA sequences with cis elements of the gene target and by acting with the RNA polymerase to allow or disallow the transcription. Through knowledge of TFs and possible genetic targets, the transcriptional monitoring mechanism can be engineered to regulate the expressed genes, thus modifying relevant metabolic pathways [57].
Advanced genome-editing techniques for microalgae such as zinc finger nuclease (ZFN), meganuclease (MN), TALEN and CRISPR/Cas9 techniques recently emerged as powerful tools for genetic engineering of various species. These molecular scissors are able to perform a targeted gene modification through one of two major mechanisms, non-homologous end-joining, which results in a base pair alteration mutation over the break site, or homologous recombination, which drives gene insertion or gene replacement to the targeted locus [135,136]. The new approaches may be used not only to optimize specific characteristics of certain microalgal species, but also to develop novel traits into existing microalgal systems to fulfill industrial necessities [137].
All these techniques and approaches of genetic engineering of microalgae show their effectiveness to boost the production of high-value compounds. It is possible and necessary to extrapolate the knowledge base on cyanobacterial genetic engineering to the eukaryotic microalgal genetic engineering [133]. Though still challenging, increasingly more newly developed genetic tools and methods have been applied to enhance the transformation and expression systems such as removing the cell wall to improve the transformation efficiency [133]. Adjustment of the cultivation condition may result in great influences on the content of some biocompounds in microalgae. With a suitable and adequate association of genetic manipulation and culture process, optimal productivity could be achieved [133].
There are various key obstacles to overcome in microalgal genetic editing. The design and regulation of multiple target sites are primarily challenging and are then followed by the establishment of intact metabolic pathways and post-translocation stability. Sufficient genomic and transcriptomic data and information are essential in achieving the above goals. The low transformation efficiency requires pretreatments to eliminate cell walls or the use of a cell wall-deficient mutant deficient mutant [133]. In addition, label-free genetic editing might be instrumental for industrial application due to safety and regulatory reasons. To counter these limitations, the fundamental study of molecular elements, such as identification and cloning of promoters, enhancers and terminator, should be carried out more intensively. The innovation and toolkits for genetic engineering of microalgae are also needed to be specifically improved.
The main challenges appear to be the high cost of operation, infrastructure, maintenance, mass production, bioproduct accumulation and extraction [138]. Using industrial or agricultural waste contained with less microbial load should be adapted to a medium for sustainability and thus saving costs for the industrial scale. Indeed, fundamental knowledge and research are also necessary, making more research on various cultivation conditions a good option within the next few years. Many plant chemicals that are of pharmaceutical interest are waiting to be produced by the benefits of genetic engineering of microbial synthesis on an industrial scale. In terms of sustainability, combined with economic, environmental and short life cycle benefits, hetero- and autotrophic microalgae may reach this goal [139]. The lower biomass productivity, harvesting technology and poorly developed downstream biorefinery are the major reasons of high production cost of the bioproducts. With the improvement in each process step in microalgal cultivation, substantial progress towards a cost-efficient microalgal-based biorefinery technology needs to be developed [140]. The challenges to meet the economic demand are multifaceted, including both product quality and cost effectiveness. Improving yield and product quality in some microalgal hosts remains to be addressed [139]. Although a small number of microalgal hosts are approaching commercialization, as the demand for therapeutics and other industries is continually growing, there are still some limitations from genetic engineering of microalgal hosts, such as difficult engineering due to the lack of a high-efficiency genetic toolbox, less-available molecular specific toolkits, short-term stability of the genetic system and less efficient manipulation outside laboratory [139].
Safety is a primary consideration for microalgal products for human food and animal feed applications, and environmental safety associated with the cultivation of wild-type microalgae and genetically modified (GM) microalgal strains is also of great concern [140]. It is important that the food security agencies establish the permissible guidelines of GM-microalgal biomass for human and animal consumption, and furthering the potential of genetically modified microalgae needs to be assessed to ensure successful commercialization in order to support future energy and food security without risk to human and environmental health. The key safety legislation must be established to evaluate the environmental risk assessment (ERA) system that may be able to respond to rapidly evolving research and development [140].

4. Conclusions

In order to fulfill requirements of the world’s growing population, inexpensive as well as environmentally friendly production processes, including exploitation of microalgae, should be more aggressively developed. Although development of genetic engineering technologies makes it possible for researchers to engineer and improve production efficiency of microalgae, only a few models species were chosen to be used so far. With advances made in sequencing technologies as well as the availability of genomic data for a variety of microalgae strains, these quickly emerging technologies allow researchers to investigate and explore the wide range of this valuable group of organisms for commercial applications. In addition, synthesis genes are becoming available and applicable to modify algal strains, resulting in both time-saving and cost-effective genetic engineering procedures. Finally, in parallel and with all these technological advances, thorough studies must be carried out in order to evaluate and understand the effects of these genetically modified strains on the environment. The stability of these strains on a large scale is primordial before their use in industrial applications.

Author Contributions

Writing—original draft preparation, S.B.G.; writing—original draft preparation, Z.L.; writing—review and editing, funding acquisition, J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Joint Program of Sichuan University and Panzhihua City for Industrial Innovation, China, grant number 2019CDPZH-19 and by the Science and Technology Planning Program in Sichuan, China, grant number 2019YFN0121.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was also supported by The Algerian Ministry of Higher Education and Scientific Research.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Pulz, O.; Gross, W. Valuable products from biotechnology of microalgae. Appl. Microbiol. Biotechnol. 2004, 65, 635–648. [Google Scholar] [CrossRef]
  2. Metting, F. Biodiversity and application of microalgae. J. Ind. Microbiol. 1996, 17, 477–489. [Google Scholar] [CrossRef]
  3. Cadoret, J.-P.; Garnier, M.; Saint-Jean, B. Microalgae, functional genomics and biotechnology. In Advances in Botanical Research; Elsevier: Amsterdam, The Netherlands, 2012; Volume 64, pp. 285–341. [Google Scholar]
  4. Mobin, S.M.A.; Chowdhury, H.; Alam, F. Commercially important bioproducts from microalgae and their current applications–A review. Energy Procedia 2019, 160, 752–760. [Google Scholar] [CrossRef]
  5. Camacho, F.; Macedo, A.; Malcata, F. Potential industrial applications and commercialization of microalgae in the functional food and feed industries: A Short Review. Mar. Drugs 2019, 17, 312. [Google Scholar] [CrossRef]
  6. 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. 2020, 43, 107602. [Google Scholar] [CrossRef]
  7. Guiry, M.D. How many species of microalgae are there? J. Phycol. 2012, 48, 1057–1063. [Google Scholar] [CrossRef]
  8. Mata, T.M.; Martins, A.A.; Caetano, N.S. Microalgae for biodiesel production and other applications: A review. Renew. Sustain. Energy Rev. 2010, 14, 217–232. [Google Scholar] [CrossRef]
  9. Olaizola, M. Commercial development of microalgal biotechnology: From the test tube to the marketplace. Biomol. Eng. 2003, 20, 459–466. [Google Scholar] [CrossRef]
  10. Harun, R.; Singh, M.; Forde, G.M.; Danquah, M.K. Bioprocess engineering of microalgae to produce a variety of consumer products. Renew. Sustain. Energy Rev. 2010, 14, 1037–1047. [Google Scholar] [CrossRef]
  11. Skjånes, K.; Rebours, C.; Lindblad, P. Potential for green microalgae to produce hydrogen, pharmaceuticals and other high value products in a combined process. Crit. Rev. Biotechnol. 2013, 33, 172–215. [Google Scholar] [CrossRef]
  12. Barsanti, L.; Gualtieri, P. Algae: Anatomy, Biochemistry, and Biotechnology; CRC Press: Boca Raton, FL, USA, 2005. [Google Scholar]
  13. Person, J.; Lando, D.; Mathieu, D. Livre Turquoise: Algues, Filières du Futur; Édition Adebiotech: Romainville, France, 2011. [Google Scholar]
  14. Borowitzka, M.A. High-value products from microalgae—their development and commercialisation. J. Appl. Phycol. 2013, 25, 743–756. [Google Scholar] [CrossRef]
  15. Choi, K.R.; Jang, W.D.; Yang, D.; Cho, J.S.; Park, D.; Lee, S.Y. Systems metabolic engineering strategies: Integrating systems and synthetic biology with metabolic engineering. Trends Biotechnol. 2019, 37, 817–837. [Google Scholar] [CrossRef] [PubMed]
  16. 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, 53, 102158. [Google Scholar] [CrossRef]
  17. Fajardo, C.; De Donato, M.; Carrasco, R.; Martínez-Rodríguez, G.; Mancera, J.M.; Fernández-Acero, F.J. Advances and challenges in genetic engineering of microalgae. Rev. Aquac. 2020, 12, 365–381. [Google Scholar] [CrossRef]
  18. Rasala, B.A.; Mayfield, S.P. Photosynthetic biomanufacturing in green algae; production of recombinant proteins for industrial, nutritional, and medical uses. Photosynth. Res. 2015, 123, 227–239. [Google Scholar] [CrossRef]
  19. Vanier, G.; Hempel, F.; Chan, P.; Rodamer, M.; Vaudry, D.; Maier, U.G.; Lerouge, P.; Bardor, M. Biochemical characterization of human anti-hepatitis B monoclonal antibody produced in the microalgae Phaeodactylum tricornutum. PLoS ONE 2015, 10, e0139282. [Google Scholar] [CrossRef]
  20. Ramos-Martinez, E.M.; Fimognari, L.; Sakuragi, Y. High-yield secretion of recombinant proteins from the microalga Chlamydomonas reinhardtii. Plant Biotechnol. J. 2017, 15, 1214–1224. [Google Scholar] [CrossRef]
  21. Davis, A.; Crum, L.T.; Corbeil, L.B.; Hildebrand, M. Expression of Histophilus somni IbpA DR2 protective antigen in the diatom Thalassiosira pseudonana. Appl. Microbiol. Biotechnol. 2017, 101, 5313–5324. [Google Scholar] [CrossRef]
  22. Wannathong, T.; Waterhouse, J.C.; Young, R.E.; Economou, C.K.; Purton, S. New tools for chloroplast genetic engineering allow the synthesis of human growth hormone in the green alga Chlamydomonas reinhardtii. Appl. Microbiol. Biotechnol. 2016, 100, 5467–5477. [Google Scholar] [CrossRef]
  23. Stoffels, L.; Taunt, H.N.; Charalambous, B.; Purton, S. Synthesis of bacteriophage lytic proteins against Streptococcus pneumoniae in the chloroplast of Chlamydomonas reinhardtii. Plant Biotechnol. J. 2017, 15, 1130–1140. [Google Scholar] [CrossRef]
  24. Corchero, J.L.; Gasser, B.; Resina, D.; Smith, W.; Parrilli, E.; Vázquez, F.; Abasolo, I.; Giuliani, M.; Jäntti, J.; Ferrer, P. Unconventional microbial systems for the cost-efficient production of high-quality protein therapeutics. Biotechnol. Adv. 2013, 31, 140–153. [Google Scholar] [CrossRef] [PubMed]
  25. 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] [PubMed]
  26. Hempel, F.; Maier, U.G. Microalgae as Solar-Powered Protein Factories. In Advanced Technologies for Protein Complex Production and Characterization; Vega, M.C., Ed.; Springer International Publishing: Cham, Switzerland, 2016; pp. 241–262. [Google Scholar]
  27. Hirschl, S.; Ralser, C.; Asam, C.; Gangitano, A.; Huber, S.; Ebner, C.; Bohle, B.; Wolf, M.; Briza, P.; Ferreira, F. Expression and characterization of functional recombinant Bet v 1.0101 in the chloroplast of Chlamydomonas reinhardtii. Int. Arch. Allergy Immunol. 2017, 173, 44–50. [Google Scholar] [CrossRef] [PubMed]
  28. Barahimipour, R.; Neupert, J.; Bock, R. Efficient expression of nuclear transgenes in the green alga Chlamydomonas: Synthesis of an HIV antigen and development of a new selectable marker. Plant Mol. Biol. 2016, 90, 403–418. [Google Scholar] [CrossRef] [PubMed]
  29. Scranton, M.A.; Ostrand, J.T.; Georgianna, D.R.; Lofgren, S.M.; Li, D.; Ellis, R.C.; Carruthers, D.N.; Dräger, A.; Masica, D.L.; Mayfield, S.P. Synthetic promoters capable of driving robust nuclear gene expression in the green alga Chlamydomonas reinhardtii. Algal Res. 2016, 15, 135–142. [Google Scholar] [CrossRef]
  30. Zedler, J.A.; Mullineaux, C.W.; Robinson, C. Efficient targeting of recombinant proteins to the thylakoid lumen in Chlamydomonas reinhardtii using a bacterial Tat signal peptide. Algal Res. 2016, 19, 57–62. [Google Scholar] [CrossRef]
  31. Zedler, J.A.; Gangl, D.; Guerra, T.; Santos, E.; Verdelho, V.V.; Robinson, C. Pilot-scale cultivation of wall-deficient transgenic Chlamydomonas reinhardtii strains expressing recombinant proteins in the chloroplast. Appl. Microbiol. Biotechnol. 2016, 100, 7061–7070. [Google Scholar] [CrossRef]
  32. Mishra, A.; Medhi, K.; Malaviya, P.; Thakur, I.S. Omics approaches for microalgal applications: Prospects and challenges. Bioresour. Technol. 2019, 291, 121890. [Google Scholar] [CrossRef]
  33. 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, 513. [Google Scholar] [CrossRef]
  34. 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]
  35. 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]
  36. Wu, C.; Zheng, C.; Wang, J.; Jiang, P. Recombinant Expression of Thrombolytic Agent Reteplase in Marine Microalga Tetraselmis subcordiformis (Chlorodendrales, Chlorophyta). Mar. Drugs 2021, 19, 315. [Google Scholar] [CrossRef]
  37. Maeda, Y.; Tsuru, Y.; Matsumoto, N.; Nonoyama, T.; Yoshino, T.; Matsumoto, M.; Tanaka, T. Prostaglandin production by the microalga with heterologous expression of cyclooxygenase. Biotechnol. Bioeng. 2021, 118, 2734–2743. [Google Scholar] [CrossRef] [PubMed]
  38. 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]
  39. 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] [PubMed]
  40. 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]
  41. 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]
  42. Park, S.; Nguyen, T.H.T.; Jin, E. Improving lipid production by strain development in microalgae: Strategies, challenges and perspectives. Bioresour. Technol. 2019, 292, 121953. [Google Scholar] [CrossRef]
  43. Yan, J.; Cheng, R.; Lin, X.; You, S.; Li, K.; Rong, H.; Ma, Y. Overexpression of acetyl-CoA synthetase increased the biomass and fatty acid proportion in microalga Schizochytrium. Appl. Microbiol. Biotechnol. 2013, 97, 1933–1939. [Google Scholar] [CrossRef]
  44. Ma, Y.-H.; Wang, X.; Niu, Y.-F.; Yang, Z.-K.; Zhang, M.-H.; Wang, Z.-M.; Yang, W.-D.; Liu, J.-S.; Li, H.-Y. Antisense knockdown of pyruvate dehydrogenase kinase promotes the neutral lipid accumulation in the diatom Phaeodactylum tricornutum. Microb. Cell Factories 2014, 13, 100. [Google Scholar] [CrossRef]
  45. Xue, J.; Niu, Y.-F.; Huang, T.; Yang, W.-D.; Liu, J.-S.; Li, H.-Y. Genetic improvement of the microalga Phaeodactylum tricornutum for boosting neutral lipid accumulation. Metab. Eng. 2015, 27, 1–9. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, J.; Kwak, H.S.; Sim, S.J.; Jin, E. Overexpression of malic enzyme isoform 2 in Chlamydomonas reinhardtii PTS42 increases lipid production. Bioresour. Technol. Rep. 2019, 7, 100239. [Google Scholar] [CrossRef]
  47. Radakovits, R.; Jinkerson, R.E.; Darzins, A.; Posewitz, M.C. Genetic engineering of algae for enhanced biofuel production. Eukaryot. Cell 2010, 9, 486–501. [Google Scholar] [CrossRef] [PubMed]
  48. Niu, Y.-F.; Zhang, M.-H.; Li, D.-W.; Yang, W.-D.; Liu, J.-S.; Bai, W.-B.; Li, H.-Y. Improvement of neutral lipid and polyunsaturated fatty acid biosynthesis by overexpressing a type 2 diacylglycerol acyltransferase in marine diatom Phaeodactylum tricornutum. Mar. Drugs 2013, 11, 4558–4569. [Google Scholar] [CrossRef]
  49. Hamilton, M.L.; Haslam, R.P.; Napier, J.A.; Sayanova, O. Metabolic engineering of Phaeodactylum tricornutum for the enhanced accumulation of omega-3 long chain polyunsaturated fatty acids. Metab. Eng. 2014, 22, 3–9. [Google Scholar] [CrossRef]
  50. Ahmad, I.; Sharma, A.K.; Daniell, H.; Kumar, S. Altered lipid composition and enhanced lipid production in green microalga by introduction of brassica diacylglycerol acyltransferase 2. Plant Biotechnol. J. 2015, 13, 540–550. [Google Scholar] [CrossRef]
  51. Klaitong, P.; Fa-Aroonsawat, S.; Chungjatupornchai, W. Accelerated triacylglycerol production and altered fatty acid composition in oleaginous microalga Neochloris oleoabundans by overexpression of diacylglycerol acyltransferase 2. Microb. Cell Factories 2017, 16, 61. [Google Scholar] [CrossRef]
  52. Rismani-Yazdi, H.; Haznedaroglu, B.Z.; Hsin, C.; Peccia, J. Transcriptomic analysis of the oleaginous microalga Neochloris oleoabundans reveals metabolic insights into triacylglyceride accumulation. Biotechnol. Biofuels 2012, 5, 74. [Google Scholar] [CrossRef]
  53. Tan, K.W.M.; Lee, Y.K. The dilemma for lipid productivity in green microalgae: Importance of substrate provision in improving oil yield without sacrificing growth. Biotechnol. Biofuels 2016, 9, 255. [Google Scholar] [CrossRef]
  54. Guo, X.; Fan, C.; Chen, Y.; Wang, J.; Yin, W.; Wang, R.R.; Hu, Z. Identification and characterization of an efficient acyl-CoA: Diacylglycerol acyltransferase 1 (DGAT1) gene from the microalga Chlorella ellipsoidea. BMC Plant Biol. 2017, 17, 48. [Google Scholar] [CrossRef]
  55. Rismani-Yazdi, H.; Haznedaroglu, B.Z.; Bibby, K.; Peccia, J. Transcriptome sequencing and annotation of the microalgae Dunaliella tertiolecta: Pathway description and gene discovery for production of next-generation biofuels. BMC Genom. 2011, 12, 148. [Google Scholar] [CrossRef] [PubMed]
  56. Courchesne, N.M.D.; Parisien, A.; Wang, B.; Lan, C.Q. Enhancement of lipid production using biochemical, genetic and transcription factor engineering approaches. J. Biotechnol. 2009, 141, 31–41. [Google Scholar] [CrossRef] [PubMed]
  57. Bajhaiya, A.K.; Moreira, J.Z.; Pittman, J.K. Transcriptional engineering of microalgae: Prospects for high-value chemicals. Trends Biotechnol. 2017, 35, 95–99. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, J.; Hao, Q.; Bai, L.; Xu, J.; Yin, W.; Song, L.; Xu, L.; Guo, X.; Fan, C.; Chen, Y. Overexpression of the soybean transcription factor GmDof4 significantly enhances the lipid content of Chlorella ellipsoidea. Biotechnol. Biofuels 2014, 7, 128. [Google Scholar] [CrossRef] [PubMed]
  59. Ngan, C.Y.; Wong, C.-H.; Choi, C.; Yoshinaga, Y.; Louie, K.; Jia, J.; Chen, C.; Bowen, B.; Cheng, H.; Leonelli, L. Lineage-specific chromatin signatures reveal a regulator of lipid metabolism in microalgae. Nat. Plants 2015, 1, 15107. [Google Scholar] [CrossRef] [PubMed]
  60. Eungrasamee, K.; Miao, R.; Incharoensakdi, A.; Lindblad, P.; Jantaro, S. Improved lipid production via fatty acid biosynthesis and free fatty acid recycling in engineered Synechocystis sp. PCC 6803. Biotechnol. Biofuels 2019, 12, 8. [Google Scholar] [CrossRef]
  61. Athanasakoglou, A.; Kampranis, S.C. Diatom isoprenoids: Advances and biotechnological potential. Biotechnol. Adv. 2019, 37, 107417. [Google Scholar] [CrossRef]
  62. Shin, Y.S.; Jeong, J.; Nguyen, T.H.T.; Kim, J.Y.H.; Jin, E.; Sim, S.J. Targeted knockout of phospholipase A2 to increase lipid productivity in Chlamydomonas reinhardtii for biodiesel production. Bioresour. Technol. 2019, 271, 368–374. [Google Scholar] [CrossRef]
  63. Kao, P.-H.; Ng, I.-S. CRISPRi mediated phosphoenolpyruvate carboxylase regulation to enhance the production of lipid in Chlamydomonas reinhardtii. Bioresour. Technol. 2017, 245, 1527–1537. [Google Scholar] [CrossRef]
  64. Cui, H.; Zhao, C.; Xu, W.; Zhang, H.; Hang, W.; Zhu, X.; Ji, C.; Xue, J.; Zhang, C.; Li, R. Characterization of type-2 diacylglycerol acyltransferases in Haematococcus lacustris reveals their functions and engineering potential in triacylglycerol biosynthesis. BMC Plant Biol. 2021, 21, 20. [Google Scholar] [CrossRef]
  65. Song, I.; Kim, S.; Kim, J.; Oh, H.; Jang, J.; Jeong, S.J.; Baek, K.; Shin, W.-S.; Sim, S.J.; Jin, E. Macular pigment-enriched oil production from genome-edited microalgae. Microb. Cell Factories 2022, 21, 27. [Google Scholar] [CrossRef] [PubMed]
  66. 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] [PubMed]
  67. Zou, L.-G.; Chen, J.-W.; Zheng, D.-L.; Balamurugan, S.; Li, D.-W.; Yang, W.-D.; Liu, J.-S.; Li, H.-Y. High-efficiency promoter-driven coordinated regulation of multiple metabolic nodes elevates lipid accumulation in the model microalga Phaeodactylum tricornutum. Microb. Cell Factories 2018, 17, 54. [Google Scholar] [CrossRef] [PubMed]
  68. Kwon, S.; Kang, N.K.; Koh, H.G.; Shin, S.-E.; Lee, B.; Jeong, B.-r.; Chang, Y.K. Enhancement of biomass and lipid productivity by overexpression of a bZIP transcription factor in Nannochloropsis salina. Biotechnol. Bioeng. 2018, 115, 331–340. [Google Scholar] [CrossRef]
  69. Xi, Y.; Yin, L.; Chi, Z.Y.; Luo, G. Characterization and RNA-seq transcriptomic analysis of a Scenedesmus obliqnus mutant with enhanced photosynthesis efficiency and lipid productivity. Sci. Rep. 2021, 11, 11795. [Google Scholar] [CrossRef]
  70. 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]
  71. Osorio, H.; Jara, C.; Fuenzalida, K.; Rey-Jurado, E.; Vásquez, M. High-efficiency nuclear transformation of the microalgae Nannochloropsis oceanica using Tn5 Transposome for the generation of altered lipid accumulation phenotypes. Biotechnol. Biofuels 2019, 12, 134. [Google Scholar] [CrossRef]
  72. 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]
  73. 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, 61, 330–338. [Google Scholar] [CrossRef]
  74. Grama, B.S.; Chader, S.; Khelifi, D.; Agathos, S.N.; Jeffryes, C. Induction of canthaxanthin production in a Dactylococcus microalga isolated from the Algerian Sahara. Bioresour. Technol. 2014, 151, 297–305. [Google Scholar] [CrossRef]
  75. Grama, B.S.; Delhaye, A.; Agathos, S.N.; Jeffryes, C. β-Carotene and Other Carotenoids and Pigments from Microalgae. In Industrial Biotechnology of Vitamins, Biopigments, and Antioxidants; Wiley: Hoboken, NJ, USA, 2016. [Google Scholar]
  76. Ma, R.; Zhao, X.; Xie, Y.; Ho, S.-H.; Chen, J. Enhancing lutein productivity of Chlamydomonas sp. via high-intensity light exposure with corresponding carotenogenic genes expression profiles. Bioresour. Technol. 2019, 275, 416–420. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, J.; Sun, Z.; Gerken, H.; Huang, J.; Jiang, Y.; Chen, F. Genetic engineering of the green alga Chlorella zofingiensis: A modified norflurazon-resistant phytoene desaturase gene as a dominant selectable marker. Appl. Microbiol. Biotechnol. 2014, 98, 5069–5079. [Google Scholar] [CrossRef] [PubMed]
  78. Anila, N.; Simon, D.P.; Chandrashekar, A.; Ravishankar, G.; Sarada, R. Metabolic engineering of Dunaliella salina for production of ketocarotenoids. Photosynth. Res. 2016, 127, 321–333. [Google Scholar] [CrossRef] [PubMed]
  79. Kathiresan, S.; Chandrashekar, A.; Ravishankar, G.; Sarada, R. Regulation of astaxanthin and its intermediates through cloning and genetic transformation of β-carotene ketolase in Haematococcus pluvialis. J. Biotechnol. 2015, 196, 33–41. [Google Scholar] [CrossRef]
  80. Huang, W.; Ye, J.; Zhang, J.; Lin, Y.; He, M.; Huang, J. Transcriptome analysis of Chlorella zofingiensis to identify genes and their expressions involved in astaxanthin and triacylglycerol biosynthesis. Algal Res. 2016, 17, 236–243. [Google Scholar] [CrossRef]
  81. Baek, K.; Yu, J.; Jeong, J.; Sim, S.J.; Bae, S.; Jin, E. Photoautotrophic production of macular pigment in a Chlamydomonas reinhardtii strain generated by using DNA-free CRISPR-Cas9 RNP-mediated mutagenesis. Biotechnol. Bioeng. 2018, 115, 719–728. [Google Scholar] [CrossRef]
  82. Kim, M.; Ahn, J.; Jeon, H.; Jin, E. Development of a Dunaliella tertiolecta strain with increased zeaxanthin content using random mutagenesis. Mar. Drugs 2017, 15, 189. [Google Scholar] [CrossRef]
  83. Galarza, J.I.; Gimpel, J.A.; Rojas, V.; Arredondo-Vega, B.O.; Henríquez, V. Over-accumulation of astaxanthin in Haematococcus pluvialis through chloroplast genetic engineering. Algal Res. 2018, 31, 291–297. [Google Scholar] [CrossRef]
  84. Cui, H.; Xu, W.; Zhu, X.; Zhao, C.; Cui, Y.; Ji, C.; Zhang, C.; Xue, J.; Qin, S.; Jia, X.; et al. Characterization of a Haematococcus pluvialis Diacylglycerol Acyltransferase 1 and Its Potential in Unsaturated Fatty Acid-Rich Triacylglycerol Production. Front. Plant Sci. 2021, 12, 771300. [Google Scholar] [CrossRef]
  85. Chen, Y.; Bi, C.; Zhang, J.; Hou, H.; Gong, Z. Astaxanthin biosynthesis in transgenic Dunaliella salina (Chlorophyceae) enhanced tolerance to high irradiation stress. S. Afr. J. Bot. 2020, 133, 132–138. [Google Scholar] [CrossRef]
  86. Perozeni, F.; Cazzaniga, S.; Baier, T.; Zanoni, F.; Zoccatelli, G.; Lauersen, K.J.; Wobbe, L.; Ballottari, M. Turning a green alga red: Engineering astaxanthin biosynthesis by intragenic pseudogene revival in Chlamydomonas reinhardtii. Plant Biotechnol. J. 2020, 18, 2053–2067. [Google Scholar] [CrossRef] [PubMed]
  87. Eilers, U.; Bikoulis, A.; Breitenbach, J.; Büchel, C.; Sandmann, G. Limitations in the biosynthesis of fucoxanthin as targets for genetic engineering in Phaeodactylum tricornutum. J. Appl. Phycol. 2016, 28, 123–129. [Google Scholar] [CrossRef]
  88. Kadono, T.; Kira, N.; Suzuki, K.; Iwata, O.; Ohama, T.; Okada, S.; Nishimura, T.; Akakabe, M.; Tsuda, M.; Adachi, M. Effect of an Introduced Phytoene Synthase Gene Expression on Carotenoid Biosynthesis in the Marine Diatom Phaeodactylum tricornutum. Mar. Drugs 2015, 13, 5334–5357. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, J.Y.; Jeon, H.; Sim, S.J.; Lee, Y.; Jin, E. Homologous sense and antisense expression of a gene in Dunaliella tertiolecta. Planta 2015, 242, 1051–1058. [Google Scholar] [CrossRef] [PubMed]
  90. Rathod, J.P.; Vira, C.; Lali, A.M.; Prakash, G. Metabolic Engineering of Chlamydomonas reinhardtii for Enhanced β-Carotene and Lutein Production. Appl. Biochem. Biotechnol. 2020, 190, 1457–1469. [Google Scholar] [CrossRef]
  91. Morikawa, T.; Uraguchi, Y.; Sanda, S.; Nakagawa, S.; Sawayama, S. Overexpression of DnaJ-Like Chaperone Enhances Carotenoid Synthesis in Chlamydomonas reinhardtii. Appl. Biochem. Biotechnol. 2018, 184, 80–91. [Google Scholar] [CrossRef] [PubMed]
  92. Esquível, M.G.; Amaro, H.M.; Pinto, T.S.; Fevereiro, P.S.; Malcata, F.X. Efficient H2 production via Chlamydomonas reinhardtii. Trends Biotechnol. 2011, 29, 595–600. [Google Scholar] [CrossRef] [PubMed]
  93. Chien, L.-F.; Kuo, T.-T.; Liu, B.-H.; Lin, H.-D.; Feng, T.-Y.; Huang, C.-C. Solar-to-bioH2 production enhanced by homologous overexpression of hydrogenase in green alga Chlorella sp. DT. Int. J. Hydrogen Energy 2012, 37, 17738–17748. [Google Scholar] [CrossRef]
  94. Quintana, N.; Van der Kooy, F.; Van de Rhee, M.D.; Voshol, G.P.; Verpoorte, R. Renewable energy from Cyanobacteria: Energy production optimization by metabolic pathway engineering. Appl. Microbiol. Biotechnol. 2011, 91, 471–490. [Google Scholar] [CrossRef]
  95. Melis, A. Photosynthetic H2 metabolism in Chlamydomonas reinhardtii (unicellular green algae). Planta 2007, 226, 1075–1086. [Google Scholar] [CrossRef]
  96. Beer, L.L.; Boyd, E.S.; Peters, J.W.; Posewitz, M.C. Engineering algae for biohydrogen and biofuel production. Curr. Opin. Biotechnol. 2009, 20, 264–271. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, H.-C.; Newton, A.J.; Melis, A. Role of SulP, a nuclear-encoded chloroplast sulfate permease, in sulfate transport and H2 evolution in Chlamydomonas reinhardtii. Photosynth. Res. 2005, 84, 289–296. [Google Scholar] [CrossRef] [PubMed]
  98. Sun, Y.; Chen, M.; Yang, H.; Zhang, J.; Kuang, T.; Huang, F. Enhanced H2 photoproduction by down-regulation of ferredoxin-NADP+ reductase (FNR) in the green alga Chlamydomonas reinhardtii. Int. J. Hydrogen Energy 2013, 38, 16029–16037. [Google Scholar] [CrossRef]
  99. McNeely, K.; Xu, Y.; Bennette, N.; Bryant, D.A.; Dismukes, G.C. Redirecting reductant flux into hydrogen production via metabolic engineering of fermentative carbon metabolism in a cyanobacterium. Appl. Environ. Microbiol. 2010, 76, 5032–5038. [Google Scholar] [CrossRef]
  100. Kosourov, S.N.; Ghirardi, M.L.; Seibert, M. A truncated antenna mutant of Chlamydomonas reinhardtii can produce more hydrogen than the parental strain. Int. J. Hydrogen Energy 2011, 36, 2044–2048. [Google Scholar] [CrossRef]
  101. Lin, H.-D.; Liu, B.-H.; Kuo, T.-T.; Tsai, H.-C.; Feng, T.-Y.; Huang, C.-C.; Chien, L.-F. Knockdown of PsbO leads to induction of HydA and production of photobiological H2 in the green alga Chlorella sp. DT. Bioresour. Technol. 2013, 143, 154–162. [Google Scholar] [CrossRef]
  102. Wu, S.; Xu, L.; Huang, R.; Wang, Q. Improved biohydrogen production with an expression of codon-optimized hemH and lba genes in the chloroplast of Chlamydomonas reinhardtii. Bioresour. Technol. 2011, 102, 2610–2616. [Google Scholar] [CrossRef]
  103. Toepel, J.; Illmer-Kephalides, M.; Jaenicke, S.; Straube, J.; May, P.; Goesmann, A.; Kruse, O. New insights into C hlamydomonas reinhardtii hydrogen production processes by combined microarray/RNA-seq transcriptomics. Plant Biotechnol. J. 2013, 11, 717–733. [Google Scholar] [CrossRef]
  104. Yang, S.; Guarnieri, M.T.; Smolinski, S.; Ghirardi, M.; Pienkos, P.T. De novo transcriptomic analysis of hydrogen production in the green alga Chlamydomonas moewusii through RNA-Seq. Biotechnol. Biofuels 2013, 6, 118. [Google Scholar] [CrossRef]
  105. Terashima, M.; Specht, M.; Naumann, B.; Hippler, M. Characterizing the anaerobic response of Chlamydomonas reinhardtii by quantitative proteomics. Mol. Cell. Proteom. 2010, 9, 1514–1532. [Google Scholar] [CrossRef]
  106. Chader, S.; Mahmah, B.; Chetehouna, K.; Amrouche, F.; Abdeladim, K. Biohydrogen production using green microalgae as an approach to operate a small proton exchange membrane fuel cell. Int. J. Hydrogen Energy 2011, 36, 4089–4093. [Google Scholar] [CrossRef]
  107. Eroglu, E.; Melis, A. Microalgal hydrogen production research. Int. J. Hydrogen Energy 2016, 41, 12772–12798. [Google Scholar] [CrossRef]
  108. Khanna, N.; Esmieu, C.; Mészáros, L.S.; Lindblad, P.; Berggren, G. In vivo activation of an [FeFe] hydrogenase using synthetic cofactors. Energy Environ. Sci. 2017, 10, 1563–1567. [Google Scholar] [CrossRef]
  109. Ban, S.; Lin, W.; Luo, Z.; Luo, J. Improving hydrogen production of Chlamydomonas reinhardtii by reducing chlorophyll content via atmospheric and room temperature plasma. Bioresour. Technol. 2019, 275, 425–429. [Google Scholar] [CrossRef] [PubMed]
  110. Durall, C.; Lindblad, P. Mechanisms of carbon fixation and engineering for increased carbon fixation in cyanobacteria. Algal Res. 2015, 11, 263–270. [Google Scholar] [CrossRef]
  111. González-Fernández, C.; Ballesteros, M. Linking microalgae and cyanobacteria culture conditions and key-enzymes for carbohydrate accumulation. Biotechnol. Adv. 2012, 30, 1655–1661. [Google Scholar] [CrossRef] [PubMed]
  112. Wei, L.; Shen, C.; El Hajjami, M.; You, W.; Wang, Q.; Zhang, P.; Ji, Y.; Hu, H.; Hu, Q.; Poetsch, A. Knockdown of carbonate anhydrase elevates Nannochloropsis productivity at high CO2 level. Metab. Eng. 2019, 54, 96–108. [Google Scholar] [CrossRef]
  113. Atsumi, S.; Higashide, W.; Liao, J.C. Direct photosynthetic recycling of carbon dioxide to isobutyraldehyde. Nat. Biotechnol. 2009, 27, 1177–1180. [Google Scholar] [CrossRef]
  114. Shih, P.M.; Zarzycki, J.; Niyogi, K.K.; Kerfeld, C.A. Introduction of a synthetic CO2-fixing photorespiratory bypass into a cyanobacterium. J. Biol. Chem. 2014, 289, 9493–9500. [Google Scholar] [CrossRef]
  115. Hu, J.; Li, T.; Xu, W.; Zhan, J.; Chen, H.; He, C.; Wang, Q. Small antisense RNA RblR positively regulates RuBisCo in Synechocystis sp. PCC 6803. Front. Microbiol. 2017, 8, 231. [Google Scholar] [CrossRef][Green Version]
  116. Gimpel, J.A.; Henríquez, V.; Mayfield, S.P. In metabolic engineering of eukaryotic microalgae: Potential and challenges come with great diversity. Front. Microbiol. 2015, 6, 1376. [Google Scholar] [CrossRef] [PubMed]
  117. Fu, W.; Nelson, D.R.; Mystikou, A.; Daakour, S.; Salehi-Ashtiani, K. Advances in microalgal research and engineering development. Curr. Opin. Biotechnol. 2019, 59, 157–164. [Google Scholar] [CrossRef] [PubMed]
  118. Melis, A. Solar energy conversion efficiencies in photosynthesis: Minimizing the chlorophyll antennae to maximize efficiency. Plant Sci. 2009, 177, 272–280. [Google Scholar] [CrossRef]
  119. Polle, J.E.; Kanakagiri, S.-D.; Melis, A. tla1, a DNA insertional transformant of the green alga Chlamydomonas reinhardtii with a truncated light-harvesting chlorophyll antenna size. Planta 2003, 217, 49–59. [Google Scholar] [CrossRef]
  120. Mussgnug, J.H.; Thomas-Hall, S.; Rupprecht, J.; Foo, A.; Klassen, V.; McDowall, A.; Schenk, P.M.; Kruse, O.; Hankamer, B. Engineering photosynthetic light capture: Impacts on improved solar energy to biomass conversion. Plant Biotechnol. J. 2007, 5, 802–814. [Google Scholar] [CrossRef]
  121. Beckmann, J.; Lehr, F.; Finazzi, G.; Hankamer, B.; Posten, C.; Wobbe, L.; Kruse, O. Improvement of light to biomass conversion by de-regulation of light-harvesting protein translation in Chlamydomonas reinhardtii. J. Biotechnol. 2009, 142, 70–77. [Google Scholar] [CrossRef]
  122. Kirst, H.; Formighieri, C.; Melis, A. Maximizing photosynthetic efficiency and culture productivity in cyanobacteria upon minimizing the phycobilisome light-harvesting antenna size. Biochim. Biophys. Acta (BBA)-Bioenerg. 2014, 1837, 1653–1664. [Google Scholar] [CrossRef]
  123. Jeong, J.; Baek, K.; Kirst, H.; Melis, A.; Jin, E. Loss of CpSRP54 function leads to a truncated light-harvesting antenna size in Chlamydomonas reinhardtii. Biochim. Biophys. Acta (BBA)-Bioenerg. 2017, 1858, 45–55. [Google Scholar] [CrossRef]
  124. Mussgnug, J.H.; Wobbe, L.; Elles, I.; Claus, C.; Hamilton, M.; Fink, A.; Kahmann, U.; Kapazoglou, A.; Mullineaux, C.W.; Hippler, M.; et al. NAB1 Is an RNA Binding Protein Involved in the Light-Regulated Differential Expression of the Light-Harvesting Antenna of Chlamydomonas reinhardtii. Plant Cell 2005, 17, 3409–3421. [Google Scholar] [CrossRef]
  125. Berger, H.; Blifernez-Klassen, O.; Ballottari, M.; Bassi, R.; Wobbe, L.; Kruse, O. Integration of carbon assimilation modes with photosynthetic light capture in the green alga Chlamydomonas reinhardtii. Mol. Plant 2014, 7, 1545–1559. [Google Scholar] [CrossRef]
  126. Hasunuma, T.; Matsuda, M.; Senga, Y.; Aikawa, S.; Toyoshima, M.; Shimakawa, G.; Miyake, C.; Kondo, A. Overexpression of flv3 improves photosynthesis in the cyanobacterium Synechocystis sp. PCC6803 by enhancement of alternative electron flow. Biotechnol. Biofuels 2014, 7, 493. [Google Scholar] [CrossRef] [PubMed]
  127. Rea, G.; Lambreva, M.; Polticelli, F.; Bertalan, I.; Antonacci, A.; Pastorelli, S.; Damasso, M.; Johanningmeier, U.; Giardi, M.T. Directed evolution and in silico analysis of reaction centre proteins reveal molecular signatures of photosynthesis adaptation to radiation pressure. PLoS ONE 2011, 6, e16216. [Google Scholar] [CrossRef] [PubMed]
  128. Morozova, O.; Marra, M.A. Applications of next-generation sequencing technologies in functional genomics. Genomics 2008, 92, 255–264. [Google Scholar] [CrossRef] [PubMed]
  129. Mardis, E.R. The impact of next-generation sequencing technology on genetics. Trends Genet. 2008, 24, 133–141. [Google Scholar] [CrossRef] [PubMed]
  130. Horner, D.S.; Pavesi, G.; Castrignano, T.; De Meo, P.D.O.; Liuni, S.; Sammeth, M.; Picardi, E.; Pesole, G. Bioinformatics approaches for genomics and post genomics applications of next-generation sequencing. Brief. Bioinform. 2010, 11, 181–197. [Google Scholar] [CrossRef] [PubMed]
  131. Guarnieri, M.T.; Pienkos, P.T. Algal omics: Unlocking bioproduct diversity in algae cell factories. Photosynth. Res. 2015, 123, 255–263. [Google Scholar] [CrossRef] [PubMed]
  132. Vavricka, C.J.; Hasunuma, T.; Kondo, A. Dynamic metabolomics for engineering biology: Accelerating learning cycles for bioproduction. Trends Biotechnol. 2020, 38, 68–82. [Google Scholar] [CrossRef]
  133. Lin, W.-R.; Tan, S.-I.; Hsiang, C.-C.; Sung, P.-K.; Ng, I.-S. Challenges and opportunity of recent genome editing and multi-omics in cyanobacteria and microalgae for biorefinery. Bioresour. Technol. 2019, 291, 121932. [Google Scholar] [CrossRef]
  134. Hegde, K.; Chandra, N.; Sarma, S.J.; Brar, S.K.; Veeranki, V.D. Genetic engineering strategies for enhanced biodiesel production. Mol. Biotechnol. 2015, 57, 606–624. [Google Scholar] [CrossRef]
  135. Daboussi, F.; Leduc, S.; Marechal, A.; Dubois, G.; Guyot, V.; Perez-Michaut, C.; Amato, A.; Falciatore, A.; Juillerat, A.; Beurdeley, M. Genome engineering empowers the diatom Phaeodactylum tricornutum for biotechnology. Nat. Commun. 2014, 5, 3831. [Google Scholar] [CrossRef]
  136. Jinkerson, R.E.; Jonikas, M.C. Molecular techniques to interrogate and edit the Chlamydomonas nuclear genome. Plant J. 2015, 82, 393–412. [Google Scholar] [CrossRef] [PubMed]
  137. Valverde, F.; Romero-Campero, F.J.; León, R.; Guerrero, M.G.; Serrano, A. New challenges in microalgae biotechnology. Eur. J. Protistol. 2016, 55, 95–101. [Google Scholar] [CrossRef] [PubMed]
  138. Hachicha, R.; Elleuch, F.; Ben Hlima, H.; Dubessay, P.; de Baynast, H.; Delattre, C.; Pierre, G.; Hachicha, R.; Abdelkafi, S.; Michaud, P.; et al. Biomolecules from Microalgae and Cyanobacteria: Applications and Market Survey. Appl. Sci. 2022, 12, 1924. [Google Scholar] [CrossRef]
  139. Jareonsin, S.; Pumas, C. Advantages of Heterotrophic Microalgae as a Host for Phytochemicals Production. Front. Bioeng. Biotechnol. 2021, 9, 58. [Google Scholar] [CrossRef]
  140. Mehariya, S.; Goswami, R.K.; Karthikeysan, O.P.; Verma, P. Microalgae for high-value products: A way towards green nutraceutical and pharmaceutical compounds. Chemosphere 2021, 280, 130553. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.