Elsevier

Algal Research

Volume 53, March 2021, 102158
Algal Research

Review article
Recent advancements in the genetic engineering of microalgae

https://doi.org/10.1016/j.algal.2020.102158Get rights and content

Highlights

  • The genetic toolkit for engineering microalgae is currently expanding.

  • New molecular tools have been utilized to produce algal-derived therapeutics.

  • Microalgae production strains have been improved using evolutionary engineering.

Abstract

The development of more sustainable food, feed, and bio-products is critical to mitigating the environmental stresses facing our world today. Algae, which includes seaweeds, eukaryotic microalgae, and cyanobacteria, are a promising platform to achieving this, as they have low energy and space requirements, are safe for human and animal consumption, and can be manipulated to produce a diversity of valuable bioproducts. This review focuses on microalgae, both eukaryotic and cyanobacteria. In the past, addressing the major challenges of bringing microalgal production systems to an economically viable scale only had a relatively small genetic toolset to work with, in comparison to other microbial systems such as bacteria and yeast. Expanding the molecular tools available for genetic engineering of microalgae will lead to higher product yields, and accelerate the development of new microalgal bioproducts for commercial applications, thereby supporting the shift towards more environmentally friendly products. In this review, we highlight significant advances from recent years on the design of microalgal expression vectors, discovery of genetic regulatory elements (promoters and transcription factors), optimization of transformation methods, and development of new strain improvement techniques, all aimed at advancing microalgae to become a more efficient biomanufacturing platform. We then discuss how these tools have been applied to improving recombinant protein production, and to enhance metabolic pathway engineering.

Introduction

Microalgae, both eukaryotic and cyanobacteria taxa, are being developed as a sustainable platform for the biomanufacturing of a variety of desirable products including foods, feeds, fuels, materials, and medicines. Due to their rapid growth, efficient use of resources, and natural genetic diversity, a wide variety of algal-derived compounds can be targeted for cost-competitive production (Fig. 1). Unlike other microorganisms, including yeast and bacteria, multiple species of microalgae are safe for human consumption and have obtained GRAS (Generally Recognized as Safe) status from the US FDA, potentially reducing purification costs of algal-derived therapeutics and expanding the potential for valuable food and feed ingredients [1]. Historically, a focus of this field has been on the commercial production of biofuels from microalgae, however due to the low cost of fossil fuels, economically viable biofuels from algae have yet to be achieved at any significant scale and focus has largely shifted towards coproduct production to offset the costs [[2], [3], [4]]. In order to achieve economic viability of commodity goods produced in algae, the entire algae biomass must be valorized through the production of valuable coproducts. Many commercially desirable compounds exist naturally in microalgae, including a variety of lipids, vitamins, pigments, and the documented highly nutritional biomass itself [5], with the potential to express high value recombinant proteins using synthetic biology. Currently, the largest challenge to developing more algae bioproducts at a commercial level is bringing production from lab scale to an industrial scale. Optimization of strains, and expansion of genetic toolsets for manipulating the strains into producing high yields of target products will be instrumental in accomplishing this.

Regardless of the intended product, molecular and genetic tools offer powerful approaches to enhance the production of targeted molecules. Development of a sophisticated suite of genetic tools in microalgae is still incomplete, compared to other industrial microorganism platforms. Nevertheless, the toolsets currently available have greatly facilitated the speed and accuracy with which one can engineer algae to optimize productivity. Metabolic engineering involves targeting specific parts of metabolic pathways within cells to change the flux of the metabolites towards a desired product [6]. Recent metabolic engineering efforts in photosynthetic microorganisms have been focused on overexpressing or knocking out key genes that encode enzymes in pathways of interest. Studies involving metabolic engineering of photosynthetic microorganisms have focused on a variety of products, with many in the fatty acid or isoprenoid biosynthetic pathways. Engineering recombinant proteins in microalgae, on the other hand, involves inserting an exogenous gene encoding for the desired protein into the algal genome in a strategic way, enabling the protein to be expressed at high levels and targeted to a specific subcellular region of the algal cells. One challenge in utilizing molecular tools in algae has been the genetic difference between species, where the interspecies genetic variability limits the use of genetic tools from one species to another. However, as the cost of genome sequencing continues to decline, more microalgal strains can be easily characterized, and the tools then adapted to their genetic code. As a result, research in this field is broadening and more widely applicable genetic toolkits are being developed to enable various species of microalgae as industrial production systems. Here we review the most current research advances and discuss how these new tools are enabling us to bring algae forward as a true commercial platform.

Section snippets

Development of new tools for genetic engineering of algae

The development of microalgae as an effective platform for engineered bioproducts will require a more complete molecular toolbox. Having an array of verified robust tools will enable manipulation of the algal genome to produce optimal quantities of the target products. Other microorganisms, such as bacteria and yeast, have been successfully developed to produce a highly diverse set of industrial products, using the expansive engineering tool sets available [[7], [8], [9]]. Microalgae are

Recent applications in metabolic engineering of natural metabolites

Metabolic engineering of photosynthetic microorganisms can be used to increase their productivity and efficiency, to create a more sustainable and cost-effective production platform. Recent metabolic engineering efforts in microalgae have been focused on the fatty acid and isoprenoid biosynthesis pathways, because of their value in the food, feed, fuel, and pharmaceutical industries.

Recent applications of recombinant protein expression to produce biopharmaceuticals

Genetic tools in algae have allowed for new applications in the production and delivery of recombinant proteins as biopharmaceuticals (Table 4). Biopharmaceuticals are medicinal products synthesized in organisms to prevent or treat disease, and include recombinantly produced vaccines, antibodies, enzymes, hormones, antibiotics, cytokines, growth factors, thrombolytic agents, and immune signaling proteins [164,165]. Recombinant protein biopharmaceutical production is a greatly emerging market

Recent applications of random mutagenesis and genome shuffling to improve algal strains

The implementation of evolutionary engineering has long been used as a genetic tool to drive the development of production strains of algae. This iterative process begins by introducing genetic diversity into a population using non-recombinant strategies followed by selective pressures and intensive screening to identify new strains with the desired phenotype. Genetic diversification is generally introduced either through random mutagenesis, using physical or chemical mutagens to cause

Conclusions

Microalgae have great potential as a sustainable and safe platform for the biomanufacturing of commercially desirable molecules. The progress being made in this field will inevitably yield strains suitable for production of useful industrial products with applications in fuels, materials, foods, feeds and therapeutics. While genetic tool development in algae lags behind those in bacteria and yeast, recent discoveries are effectively expanding the molecular toolkit and simplifying the

CRediT authorship contribution statement

Ashley Sproles: Conceptualization, Formal analysis, Writing – Original draft preparation, Writing – Review & Editing; Frank Fields: Conceptualization, Formal analysis, Writing – Original draft preparation, Writing – Review & Editing, Visualization; Tressa Smalley: Formal analysis, Writing – original draft preparation; Chau Le: Formal analysis, Writing – Original draft preparation; Stephen Mayfield: Writing – Review & Editing, Supervision.

Acknowledgements and funding sources

The graphical figure was created with BioRender.com. Funding was provided by the U.S. Department of Energy, as part of the PEAK (DE-EE0008246) and BEEPS (DE-EE0008491) projects.

Declaration of competing interest

The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Stephen Mayfield holds an equity position in Algenesis Materials, a company that could potentially benefit from this publication.

References (230)

  • S. Kobayashi et al.

    Application of an engineered chromatic acclimation sensor for red-light-regulated gene expression in cyanobacteria

    Algal Res.

    (2019)
  • M.A. Scranton et al.

    Synthetic promoters capable of driving robust nuclear gene expression in the green alga Chlamydomonas reinhardtii

    Algal Res.

    (2016)
  • T. Gaj et al.

    ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering

    Trends Biotechnol.

    (2013)
  • W.R. Lin et al.

    Challenges and opportunity of recent genome editing and multi-omics in cyanobacteria and microalgae for biorefinery

    Bioresour. Technol.

    (2019)
  • W.Z. Jiang et al.

    A gene-within-a-gene Cas9/sgRNA hybrid construct enables gene editing and gene replacement strategies in Chlamydomonas reinhardtii

    Algal Res.

    (2017)
  • H. Li et al.

    CRISPR-Cas9 for the genome engineering of cyanobacteria and succinate production

    Metab. Eng.

    (2016)
  • G.C. Gordon et al.

    CRISPR interference as a titratable, trans-acting regulatory tool for metabolic engineering in the cyanobacterium Synechococcus sp. strain PCC 7002

    Metab. Eng.

    (2016)
  • D. Kaczmarzyk et al.

    Diversion of the long-chain acyl-ACP pool in Synechocystis to fatty alcohols through CRISPRi repression of the essential phosphate acyltransferase PlsX

    Metab. Eng.

    (2018)
  • A.K. Bajhaiya et al.

    Transcriptional engineering of microalgae: prospects for high-value chemicals

    Trends Biotechnol.

    (2017)
  • Y. Ma et al.

    Evaluation of the potential of 9 Nannochloropsis strains for biodiesel production

    Bioresour. Technol.

    (2014)
  • Y. Torres-Tiji et al.

    Microalgae as a future food source

    Biotechnol. Adv.

    (2020)
  • E. Stephens et al.

    An economic and technical evaluation of microalgal biofuels

    Nat. Biotechnol.

    (2010)
  • R.H. Wijffels et al.

    An outlook on microalgal biofuels

    Science (80-)

    (2010)
  • M.P. Naghshbandi et al.

    Metabolic engineering of microalgae for biofuel production

  • G.J. Gopal et al.

    Strategies for the production of recombinant protein in Escherichia coli

    Protein J.

    (2013)
  • S.E. O’Connor

    Preface

    (2016)
  • D. Porro et al.

    Production of recombinant proteins and metabolites in yeasts

    Appl. Microbiol. Biotechnol.

    (2011)
  • E. Celińska et al.

    Golden Gate Assembly system dedicated to complex pathway manipulation in Yarrowia lipolytica

    Microb. Biotechnol.

    (2017)
  • C. Engler et al.

    A Golden Gate modular cloning toolbox for plants

    ACS Synth. Biol.

    (2014)
  • S.V. Iverson et al.

    CIDAR MoClo: improved MoClo assembly standard and new E. coli part library enable rapid combinatorial design for synthetic and traditional biology

    ACS Synth. Biol.

    (2016)
  • M.E. Lee et al.

    A highly characterized yeast toolkit for modular, multipart assembly

    ACS Synth. Biol.

    (2015)
  • A. Martella et al.

    EMMA: an extensible mammalian modular assembly toolkit for the rapid design and production of diverse expression vectors

    ACS Synth. Biol.

    (2017)
  • S.J. Moore et al.

    EcoFlex: a multifunctional MoClo kit for E. coli synthetic biology

    ACS Synth. Biol.

    (2016)
  • P. Crozet et al.

    Birth of a photosynthetic chassis: A MoClo toolkit enabling synthetic biology in the microalga Chlamydomonas reinhardtii

    ACS Synth. Biol.

    (2018)
  • E. Poliner et al.

    Identification of circadian rhythms in Nannochloropsis species using bioluminescence reporter lines

    Plant J.

    (2019)
  • E. Poliner et al.

    Advanced genetic tools enable synthetic biology in the oleaginous microalgae Nannochloropsis sp

    Plant Cell Rep.

    (2018)
  • I. Weiner et al.

    Enhancing heterologous expression in Chlamydomonas reinhardtii by transcript sequence optimization

    Plant J.

    (2018)
  • A. Eichler-Stahlberg et al.

    Strategies to facilitate transgene expression in Chlamydomonas reinhardtii

    Planta

    (2009)
  • M. Schroda et al.

    The HSP70A promoter as a tool for the improved expression of transgenes in Chlamydomonas

    Plant J.

    (2000)
  • J. Wu et al.

    Efficient expression of green fluorescent protein (GFP) mediated by a chimeric promoter in Chlamydomonas reinhardtii

    Chin. J. Oceanol. Limnol.

    (2008)
  • K.E. Apt et al.

    Stable nuclear transformation of the diatom Phaeodactylum tricornutum

    Mol. Gen. Genet.

    (1996)
  • D. Wu-Scharf et al.

    Transgene and transposon silencing in Chlamydomonas reinhardtii by a DEAH-Box RNA helicase

    Science (80-)

    (2000)
  • J.A. Casas-Mollano et al.

    SET3p monomethylates histone H3 on lysine 9 and is required for the silencing of tandemly repeated transgenes in Chlamydomonas

    Nucleic Acids Res.

    (2007)
  • J.A. Casas-Mollano et al.

    The MUT9p kinase phosphorylates histone H3 threonine 3 and is necessary for heritable epigenetic silencing in Chlamydomonas

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • D. Strenkert et al.

    Heat shock factor 1 counteracts epigenetic silencing of nuclear transgenes in Chlamydomonas reinhardtii

    Nucleic Acids Res.

    (2013)
  • J. Hou et al.

    Isolation and functional validation of salinity and osmotic stress inducible promoter from the maize type-II H+-pyrophosphatase gene by deletion analysis in transgenic tobacco plants

    PLoS One

    (2016)
  • E. Englund et al.

    Evaluation of promoters and ribosome binding sites for biotechnological applications in the unicellular cyanobacterium Synechocystis sp. PCC 6803

    Sci. Rep.

    (2016)
  • A.L. Markley et al.

    Synthetic biology toolbox for controlling gene expression in the cyanobacterium Synechococcus sp. strain PCC 7002

    ACS Synth. Biol.

    (2015)
  • K. Thiel et al.

    Translation efficiency of heterologous proteins is significantly affected by the genetic context of RBS sequences in engineered cyanobacterium Synechocystis sp. PCC 6803

    Microb. Cell Factories

    (2018)
  • B. Wang et al.

    A genetic toolbox for modulating the expression of heterologous genes in the cyanobacterium Synechocystis sp. PCC 6803

    ACS Synth. Biol.

    (2018)
  • Cited by (0)

    View full text