Skip to main content

Microalgae biofuels: illuminating the path to a sustainable future amidst challenges and opportunities

Abstract

The development of microalgal biofuels is of significant importance in advancing the energy transition, alleviating food pressure, preserving the natural environment, and addressing climate change. Numerous countries and regions across the globe have conducted extensive research and strategic planning on microalgal bioenergy, investing significant funds and manpower into this field. However, the microalgae biofuel industry has faced a downturn due to the constraints of high costs. In the past decade, with the development of new strains, technologies, and equipment, the feasibility of large-scale production of microalgae biofuel should be re-evaluated. Here, we have gathered research results from the past decade regarding microalgae biofuel production, providing insights into the opportunities and challenges faced by this industry from the perspectives of microalgae selection, modification, and cultivation. In this review, we suggest that highly adaptable microalgae are the preferred choice for large-scale biofuel production, especially strains that can utilize high concentrations of inorganic carbon sources and possess stress resistance. The use of omics technologies and genetic editing has greatly enhanced lipid accumulation in microalgae. However, the associated risks have constrained the feasibility of large-scale outdoor cultivation. Therefore, the relatively controllable cultivation method of photobioreactors (PBRs) has made it the mainstream approach for microalgae biofuel production. Moreover, adjusting the performance and parameters of PBRs can also enhance lipid accumulation in microalgae. In the future, given the relentless escalation in demand for sustainable energy sources, microalgae biofuels should be deemed a pivotal constituent of national energy planning, particularly in the case of China. The advancement of synthetic biology helps reduce the risks associated with genetically modified (GM) microalgae and enhances the economic viability of their biofuel production.

Graphical Abstract

Highlights

  • Highly adaptable microalgae strains are the preferred choice for large-scale biofuel production.

  • Omics technologies and genetic editing have significantly increased lipid accumulation in microalgae, but rigorous risk assessment is required.

  • Adjusting the parameters of PBRs can lead to efficient lipid accumulation.

  • A promising future for the microalgae biofuel industry is highly anticipated in China.

Background

For several decades, increasing global population, rapid industrialization, and urbanization have led to a significant rise in energy demand. In 2021, primary energy use surged by 5.8%, predominantly from fossil fuel [1]. The availability of secure, affordable, and low-carbon energy is of increasing importance, given the declining reserves of fossil fuels, the global warming crisis, and the geopolitical tensions that exist around the world [2]. Currently, various forms of clean energy, such as solar, ocean tides, hydroelectric, geothermal, wind, and biofuels, are being researched and implemented by numerous countries to satisfy the increasing energy demands [3, 4]. Among them, biodiesel stands out as the only biofuel capable of replacing fossil fuels, while other clean energies are constrained by their ecological footprint, economic performance, dependence on the environment, and geographical location, making them more suitable for power generation and heating purposes only [5]. Hence, biofuel production is receiving global attention and holds promising prospects for development and future demand. In terms of global biofuel production in 2021, besides the traditional biofuel producers such as the United States, Brazil, and the European Union, India, Switzerland, and Argentina also made noteworthy contributions to the growth in biofuel production [1]. Simultaneously, there is a continuous emergence of new national-level policies encouraging the gradual substitution of fossil fuels with biofuels [6, 7]. As one of the developing countries, China faces the problem of fossil fuels shortage and carbon emission. In recent years, China is also striving to find a sustainable development path of energy, and the biofuel industry is considered to be an important means to ensure energy security. The present situation and policy support of biofuel industry in China are introduced in this paper.

Biofuels, derived from a diverse range of sources, such as higher plants, algae, microorganisms, crop straw, and livestock manure, offer a wide array of product types, including biodiesel, biohydrogen, bioethanol, biogas, and solid fuels. They are typically divided into four generations based on the feedstock sources (Fig. 1). The first-generation of biofuels often utilizes edible materials, such as corn, soybeans, wheat, sunflower oil, and palm oil as feedstocks, which can result in competition between food and fuel sources [8]. Second-generation biofuels, primarily derived from non-edible sources, such as switchgrass, poplar, straw, and food waste, are more sustainable than the first generation. However, both face limitations due to grain, land, and water resources [9].

Fig. 1
figure 1

Overview of feedstocks and productions of different biofuel generations. 1st generation biofuels utilize corn, soybeans, wheat, sunflower oil, and palm oil as feedstocks; 2nd generation biofuels utilize switchgrass, poplar, straw, and food waste as feedstocks; 3rd and 4th generation biofuels utilize microalgae as feedstocks, with microalgae being editable in 4th generation

Microalgae, which are important feedstocks for third- and fourth-generation biofuels, can grow in seawater, wastewater, food waste, and even saline–alkaline soil, and can obtain higher biomass than land plants using limited nutrients [10,11,12,13,14,15]. Microalgae exhibit a fast growth rate, a higher photosynthetic efficiency, and a better land utilization rate, which can yield a range of biofuel products [3, 16]. In the past two decades, scientists have made significant strides in researching microalgae biodiesel. Several species, including Chlorella vulgaris, Nannochloropsis oceanica, Dunaliella salina, Botryococcus, Desmodesmus, Neochloris, Scenedesmus, and Tetraselmis, have been identified as suitable for biodiesel production, with some of them producing biodiesel in laboratory scale with properties meeting ASTM standards [17,18,19,20,21,22,23,24]. Synechococcus strains also exhibit compliance with the EN 14214 standards in terms of iodine value and cetane number while also demonstrating lower cold filter plug point values [25]. Moreover, the fuel properties of diesel and microalgae biodiesel mixture have been extensively studied [26,27,28]. While various studies have documented methods of producing lipid from microalgae by manipulating culture conditions, fundamental limitations cannot be overcome if unsuitable strains are chosen for biofuel production. It is essential to conduct thorough investigations into species-specific characteristics regarding lipid production from microalgae. Hence, the selection of robust strains that could harvest a high amount of desired products is a crucial step in the large-scale production of biofuels [29], and this article will provide a comprehensive review on this topic.

Due to differences in cell size, lipid content and fatty acids (FAs) composition among microalgae species, not all of them are useful for biofuel production. Recently, genetically engineered microalgae have attracted the interest of researchers, and numerous species have been modified, including Chlamydomonas, C. vulgaris, Phaeodactylum tricornutum, Thalassiosira pseudonana, Galdieria sulphuraria, B. braunii, D. salina, etc. [30]. This approach employs genetic engineering to enhance and adjust the standardized traits of microalgae, resulting in the production of high-yield lipids. It proves to be more effective, precise, and rapid compared to random mutagenesis, offering extensive potential applications [31]. The resources, methods, and limitations of microalgae genetic engineering are all discussed in this article.

While the market potential for microalgae biofuels has been widely recognized, several economic, biotechnological, and environmental challenges must be addressed before microalgal products can enter the global market [32]. These include cost-effective dehydration and harvesting methods, process improvements in product extraction, as well as the thorough exploration of genomic resources and genetic tools [33, 34]. Thus, the objective of this paper is to outline the development direction of microalgae biofuels, taking into account the current achievements and challenges.

Selection and cultivation of microalgae strains

Types of microalgae from biofuel sources

The success of large-scale microalgae culture relies heavily on the selection of appropriate species. It should be noted that the lipid productivity of microalgae, which is a critical criterion for strain selection, has been a focus in previous studies. The type of lipids synthesized by microalgae can affect the properties of biodiesel [35]. Triacylglycerol (TAG) is the preferred lipid component for use as biodiesel feedstocks, and is usually expressed as the total lipid content as a percentage of dry weight [36]. While a recent study challenges the notion that natural cyanobacteria lack neutral lipid TAG, it remains a fact that TAG content in cyanobacteria is generally low [37, 38]. Unlike the abundant diacylglycerol (DG) found in cyanobacteria, TAG constitutes the primary lipid component in green algae [39, 40]. Moreover, some strains of cyanobacteria have been reported to possess higher lipid productivity, although it generally remains lower than that of green algae [41,42,43]. Therefore, the focus of this study primarily lies in green algae.

Green algae (Chlorophyceae) have the largest group of oleaginous microalgae, with high lipid abundance, and are good potential biodiesel feedstocks [37, 44, 45]. Typically, the average total lipid content of oleaginous green algae is 25.5%, while nutrient deficiency or stress conditions can increase the total lipid content substantially (up to 45.7%) [37]. The lipid content in many oleaginous green algae significantly surpasses that of typical lipid-producing crops, such as maize, soybean, canola, castor, sunflower, and palm oil. This high lipid content contributes to significant biodiesel production. However, for most microalgae, high biomass does not necessarily equate to high lipid productivity, and high lipid content may actually lead to lower biomass productivity. Consequently, it is not accurate to concentrate solely on lipid content and not biomass, since lipid productivity is determined by the combination of both factors. In the past decade, researchers have been striving to screen new microalgal strains to achieve higher lipid accumulation. Table 1 documents the biomass, biomass productivity, lipid content and lipid productivity of various green algae under different culture conditions. For Neochloris oleoabundans and Botryococcus braunii, the maximum lipid content can reach more than 74%, while their respective lipid productivity has not been the most impressive (Table 1). On the other hand, Chromochloris zofingiensis displays the highest lipid productivity, as well as a high biomass productivity and lipid content, and seems to be a particularly suitable option for biodiesel production (Table 1). Lipid productivity reflects the actual situation of microalgal growth and lipid accumulation per unit time, avoiding the misleading conclusions that may arise from relying solely on lipid content or biomass productivity. The study by Yu et al. [46] also found that, under similar lipid content in Chlorella, accumulating more biomass per unit time can effectively reduce production costs. Therefore, comparing lipid productivity is a crucial step in strain selection for microalgal biofuel production.

Table 1 Biomass and lipid in various green algae species

In addition to lipid productivity, the FAs composition is also essential for the success of biodiesel production as they can affect the characteristics of biodiesel. Microalgae FAs consist of both saturated and unsaturated FAs, featuring hydrocarbon chains ranging from 4 to 36 carbons [82]. The main saturated fatty acids (SFA), monounsaturated fatty acids (MUFA), and polyunsaturated fatty acids (PUFA) in microalgae are palmitic acid (C16:0), oleic acid (C18:1), linoleic acid (C18:2) and linolenic acid (C18:3), respectively [83]. Table S1 is not a strict comparison of the performance among 16 oleaginous microalgal species with FAs compositional profiles reported in the literature, and provides the percentage of each FAs to the total. Comparison with the FAs profiles of different microalgal species reveals several interesting features. First, SFA and MUFA are dominant in most of the algae examined. The sum of SFA and MUFA of A. fusiformis, B. braunii, Chlamydomonas sp., C. vulgaris, Nannochloris sp., N. oleoabundans, S. incrassatulus, T. obliquus, and P. subcapitata were more than two-thirds of the total FAs (Additional file 1: Table S1). Among these, the content of MUFA is considered an optimal choice for microalgal biodiesel production [84]. Second, all of these oleaginous microalgae have considerable amounts of C16 and C18 species. The C16–C18 of 16 microalgae accounted for 89.24% of the total FAs on average (Additional file 1: Table S1). Moreover, these oleaginous microalgae commonly exhibit elevated levels of C16:0 and C18:1, which are the most prevalent components in biodiesel [85]. Therefore, the selection of suitable strains for biodiesel production should take into account the FAs composition in microalgae, contributing to enhanced biodiesel performance.

Factors affecting lipid accumulation

Although oleaginous microalgae are abundant in nature, basal lipid levels can vary substantially by modifying lipid metabolism in different ways. Common approaches to increased lipid productivity include nutrient deficiencies, extreme stress, and environmental perturbations. This paper summarized the effects of inorganic carbon sources (CO2 and NaHCO3), nitrogen and phosphorus limitation, saline stress, light, and temperature on the lipid accumulation of microalgae (Fig. 2).

Fig. 2
figure 2

Methodology of enhancing lipid accumulation in microalgae cells. Nutrient deficiency, saline stress, light, temperature, CO2, and NaHCO3 can induce lipid accumulation in microalgae

CO2 and NaHCO3 concentration

Phototrophic microalgae can produce biomass by absorbing CO2. Published data showed that 1–5% CO2 concentration is appropriate for numerous microalgae growth; however, higher CO2 concentration will inhibit the growth of microalgae [86, 87]. In this concentration range, increased CO2 concentration helps Tribonema minus [88], Chlamydomonas [89], and Nannocholoropsis oculate [90] to accumulate more lipids and achieve excellent lipid production. At the same time, excessively low or high CO2 concentrations lead to a reduction in lipid content. This implies that the optimal levels of CO2 affect lipid production and their subsequent accumulation within the cellular structure. In large-scale production, industrial flue gas is an economical source of CO2 for microalgae biofuel production, with CO2 concentrations typically ranging from 10% to 20% [91]. Currently, certain microalgae species have been discovered thriving in elevated CO2 environments, with a few demonstrating enhanced lipid accumulation capabilities even under a 15% CO2 culture condition [91,92,93,94,95]. Currently, Chile and Colombia have already applied microalgae photobioreactors for carbon capture [96], providing feasibility for the large-scale application of high-CO2-adaptive microalgae in utilizing flue gas for biofuel production. Although, it is important to note that these microalgae are merely a limited selection of representatives.

NaHCO3 is a valuable carbon source for some microalgae and is much easier to handle than gaseous CO2 in storage, transportation, and processing. In recent studies, exogenous NaHCO3 has been found to significantly increase the total lipid content of D. salina, D. tertiolecta, Scenedesmus, and Nannochloris, and the addition of NaHCO3 under nutrient deficiency conditions has further significantly increased the lipid content of D. salina [13, 97,98,99]. Besides, some microalgae even prefer NaHCO3 to CO2 for lipid biosynthesis [100]. This may be attributed to the enhanced activity of key enzymes in lipid metabolism due to the presence of NaHCO3, stimulating microalgae to assimilate extracellular inorganic carbon. In our previous study, we observed that the addition of exogenous high concentrations of NaHCO3 led to a significant accumulation of lipid bodies in microalgae, and the gene UTP-glucose-1-phosphate uridylyltransferase 3, associated with lipid synthesis, was upregulated [101]. Similarly, high concentrations of NaHCO3 increased the activity of Ribulose-1, 5-bisphosphate carboxylase/oxygenase (Rubisco), promoting the generation of 3-phosphoglycerate, which serves as a substrate for FAs production [102]. Another interpretation suggests that NaHCO3 enhances the photosynthetic capacity of microalgae, causing more carbon to be directed towards lipid accumulation. Indeed, in Nannochloris sp., we observed a significant enhancement in photosynthetic capacity after NaHCO3 treatment [13]. Furthermore, Roya et al. has also suggested that NaHCO3 increases lipid content while concomitantly enhancing the rate of CO2 fixation in Chlorella sp. [100]. In the selection and breeding of microalgae, the carbon fixation efficiency of high concentration inorganic carbon sources can be used as one of the screening criteria in the future.

Nutritional limitation

Numerous researchers have used nutritional limitation as an applied and promising strategy to alter and control the cell cycle and biochemical pathways associated with lipid production and accumulation in microalgae [103]. Nitrogen and phosphorus are regarded as fundamental factors that exert a direct influence on lipid accumulation in microalgae [104]. In particular, nitrogen deficiency is often cited as a booster of lipid augmentation in most microalgae species [105]. Nitrogen deficiency serves as a significant stimulus for lipid accumulation in various microalgae species, such as Acutodesmus dimorphus [106], Nephrochlamys yushanlensis [107, 108], C. vulgaris [109], Scenedesmus quadricauda [110], and N. oceanica [111]. These reports imply that the degradation of nitrogen compounds is the possible reason for lipid accumulation under nitrogen deficiency conditions.

During the initial stage of nitrogen deficiency, the photosynthetic capacity of microalgae decreases, and the excess carbon is more easily converted into storage compounds, such as lipids and starch. According to this view, the chlorophyll and proteins are degraded as a nitrogen source to support photosynthesis with nitrogen limitation, and the excess carbon is used to the synthesize FAs, which is also the reason for the changes in FAs composition of microalgae under nitrogen deficiency [94]. It is known that the FAs compositions of N. oceanica [112], Coelastrella multistriata [113], Scenedesmus sp. and C. sorokiniana [114], Nannochloropsis gaditana and Coccomyxa elongata [115, 116] indeed altered under nitrogen deficiency. In general, these changes involved notable increases in SFA and MUFA, accompanied by a decrease in PUFA. A decrease in PUFA and a simultaneous increase in the SFA resulted in a reduced degree of unsaturation of the total FAs pool. This may be related to oxidative damage caused by nitrogen deficiency, to which PUFA is particularly sensitive [117]. It is also possible that the production of enzymes involved in desaturation and elongation reactions is reduced at nitrogen-limited condition, leading to a decrease in PUFA production [118].

However, the growth of microalgae cells under nitrogen deficiency is inhibited usually, which is manifested as a decrease in chlorophyll content and yellowing cultures [119]. The intracellular nitrogen quota plays a crucial role in regulating the allocation of carbon between the functional pool and the storage pool, thus nitrogen deficiency consistently impacts the carbon flow in microalgae, as indicated in references [120, 121]. In the initial stages of the nitrogen-depleted condition, both carbohydrates and lipids are produced at higher levels. Nevertheless, as nitrogen levels decrease, the fixed carbon undergoes a shift from the synthesis of functional biomass to the generation of storage molecules, resulting in the redirection of previously stored carbon towards neutral lipids [121]. In that case, neutral lipid levels are significantly higher than carbohydrates [122]. Despite the unfavorable conditions that nutrient deficiency creates within microalgae cells, the prospect of increased lipid production and accumulation as a desirable outcome has motivated researchers and the industry to continue utilizing this technique [123].

Numerous investigations have explored phosphorus deficiency techniques in different strains to comprehend and optimize different output parameters. It has been found that phosphorus deficiency can increase the accumulation of lipids and FAs [113, 119, 124], and the changes of FAs composition were similar to that under nitrogen deficiency. However, the increase of lipids and FAs in phosphorus deficiency is much smaller than that in nitrogen deficiency [108, 125]. These results indicate that nitrogen deficiency is more effective than phosphate deficiency in inducing lipid accumulation in some microalgae species.

Saline stress

Saline stress has proven to be a highly effective method for enhancing lipid content, making it crucial to investigate lipid accumulation in microalgae as model organisms under saline stress in the context of biofuel development strategies. The lipid content of various microalgae species such as C. reinhardtii, A. dimorphus, C. vulgaris, and Acutodesmus obliquus has been found to increase when exposed to concentrations exceeding 0.1 M NaCl [66, 126,127,128]. It has been observed that microalgae respond to saline stress by reallocating resources to carbohydrates, which are later utilized in the biosynthesis of neutral lipids [129]. It should be emphasized that in the lipid accumulation process of microalgae, KCl, MgCl2, and CaCl2 also play crucial roles, and in certain species, they may exert a more pronounced influence than NaCl [130]. Moreover, saline stress not only increases the lipid content but also causes changes in the FAs profile [131, 132]. Although some microalgae showed an increase in MUFA, most microalgae still showed an increase in SFA after high concentration of NaCl treatments [128, 129, 133]. A previous study has shown that the FAs composition is closely related to the fluidity of cell membranes [134]. A decrease in unsaturated FAs is thought to help reduce membrane fluidity, which is critical for cells to withstand saline stress [135]. In conclusion, saline stress to a certain extent can promote microalgae to accumulate lipids. However, this promotion depends on the salt tolerance of microalgae.

Light and temperature

Light intensity is an essential parameter that plays a crucial role in the lipid content and FAs composition of microalgae. A lack of sufficient light can lead to reduced production, while excessive light can result in photoinhibition. Microalgae exposed to high light intensity (HL) lead to excellent lipid and TAG accumulation [136,137,138,139,140,141]. It has also been observed that HL conditions can induce an increase in lipid content in Synechocystis sp. PCC6803 [142]. Although excessive light causes oxidative damage to cells, cells respond to the situation by converting the light energy into lipids for storage within the cell. Hence, strains with high light-tolerance are ideal for large-scale production systems in outdoor conditions. Study of two high-light-tolerant Chlamydomonas mutants supported this claim, showing that their lipid productivity increased twice as much as that of the wild type under high-light conditions [143].

From the early fluorescent lamps to the light emitting diodes (LEDs) used today, artificial light sources for microalgae culture vary in the characteristics of the emission spectrum. The light spectrum is not only essential for microalgae growth but also critically affects lipid accumulation [144]. Several studies have specifically investigated the impact of LED light on the enhancement of lipids in microalgae, highlighting the significant role of blue light in this process [137, 145, 146]. Nevertheless, it is worth noting that certain microalgae have demonstrated sensitivity to green light, resulting in either a substantial inhibition or promotion of lipid accumulation [147, 148]. Moreover, the FAs composition of microalgae is also affected by the different light spectra [149,150,151]. These results highlight the potential of coupling light spectrum to lipid synthesis in bioprocess development.

Temperature is another primary environmental driver affecting growth and lipid content in microalgae cells; however, different species have different ranges of adaptation to temperature. The mechanisms that govern and regulate the adaptation of physiological responses to temperature stress are largely unknown [152]. For example, in Tetraselmis subcordiformis, N. oculata and Parachlorella kessleri, the response temperature of lipid accumulation of each microalga is different [153, 154]. Besides, a decrease in the culture temperature leads to the shift of desaturases activity and an increase in the unsaturation level in FAs, which ensure the cell membrane fluidity under temperature stress conditions [155,156,157,158]. These findings suggest that the lipid content and FAs composition of microalgae can be influenced to a certain extent by temperature control. However, it is important to note that the method of inducing lipids through temperature control is species-specific and challenging to implement on a large scale in production.

In summary, achieving high productivity for biofuels hinges on the rational control of factors, such as light, temperature, stress, and nutrition. When cultivating microalgae outdoors, prioritizing those with high light tolerance is crucial, and considering their salinity and alkali tolerance is also essential. This ensures that microalgae can thrive in extreme environments without interference from foreign microorganisms, a fundamental requirement for large-scale biofuel production using microalgae. However, for indoor cultivation, microalgae with a high tolerance for CO2 are preferred, as they can strike a balance between carbon sequestration and lipid production. As mentioned earlier, such microalgae can utilize industrial flue gas for biofuel production, further reducing costs and enhancing sustainability.

The cultivation systems of microalgae to induce lipid accumulation

Cultivation of microalgae appears to be relatively straightforward and can be established with limited or even no supervision, using simple nutrients and even waste-water while absorbing CO2 from the atmosphere [159]. Large-scale microalgae cultivation can occur in either open or closed systems (Fig. 3). Open systems encompass a range of setups, including small artificial ponds and large bodies of open water, each employing various agitation systems [160]. Closed system mainly refers to various types of PBRs, including plastic bags, horizontal or vertical tubes, bubble columns, airlift reactors, flat panel tubular reactors, plate reactors, and even membrane-based systems [83]. Initially, open systems were more acceptable than PBRs from a commercial point of view, as they were relatively economical to build, could accommodate large areas and were easy to operate [5]. However, such systems are unable to control the stable environmental conditions (temperature and light), and are inherently susceptible to contamination, leading to inefficient utilization of light by the cells, contamination by predators, loss due to evaporation, and escape of CO2 [161]. Due to the shortcomings of open systems, much attention has been paid to closed PBRs. On one hand, PBRs prevent evaporation and reduce both contamination risks and CO2 losses [162]. On the other hand, PBRs have strong photosynthetic efficiency leading to a high biomass and lipid productivity [163].

Fig. 3
figure 3

Microalgae cultivation systems. a Open cultivation systems, b flat plate photobioreactor, c column photobioreactor, d tubular photobioreactor

Performance of PBRs

PBRs allow for a wider selection of microalgae species, while open systems typically lead to the cultivation of only a few robust photoautotrophic species [160]. Microalgae such as C. reinhardtii, N. oceanica, Chlorella pyrenoidosa, Golenkinia sp., and others have been successfully cultivated in PBRs. Some of them exhibit superior growth performance in PBRs compared to open systems, showcasing the ability to achieve higher biomass and lipid accumulation levels [164,165,166,167]. Since the design of different PBRs directly affects biomass productivity, novel PBRs to improve the process design and operation are rising. Studies focused on how to optimize biomass production, improve contaminants removal efficiency, reduce cost and space dominance by renovating the configurations of PBRs [163, 168]. Among them, studies focused on enhancing lipid production and quality through membrane PBRs predominantly aim to utilize wastewater for inducing lipid production [169,170,171,172]. This approach is considered more environmentally friendly in terms of water resource management compared to other types of PBRs.

Unlike closed systems, PBRs can improve the efficiency of light energy utilization by microalgal cells through their unique light path design. The overall optical conditions in PBRs can be evaluated in terms of light distribution and light penetration. As an example, the utilization of light and mass transfer efficiency of microalgae in flat-plate PBRs can be significantly enhanced by optimizing the ratio of the gap in the baffle opening to the width of the bioreactor [173]. By doing so, microalgae can efficiently convert optical energy into chemical energy, leading to the efficient accumulation of biomass and lipids. In fact, studies have reported that microalgae cultivated in these improved PBRs have achieved nearly threefold increases in biomass and lipid accumulation [174]. Besides, hydrodynamics is another crucial factor that affects the performance of PBRs. Proper fluid mixing helps prevent sedimentation, influences nutrient delivery, and governs the movement of microalgal cells in the reactor, which in turn impacts their light exposure history [175]. In one study, four different PBRs were investigated at varying air flow rates to evaluate their impact on biomass and lipid production from N. oleoabundans, and it was found that the bubble column reactor with a ring sparger displayed superior performance [176]. By adding mixers and improving sparger performances, novel PBRs with efficient mixing, high rates of gas–liquid mass transfer, and well-defined fluid flow patterns can be obtained for microalgal lipid accumulation and biofuel production purposes [177,178,179,180]. The results suggest that the production of microalgal biofuels in PBRs is influenced by the reactor configuration and operating conditions. Ideally, PBRs should not cause cell disruption and damage, but cell damage occurs in PBRs (with turbulent flow) when the smallest eddies are the size of cells or much larger than the cells. Thus, optimizing the performance of PBRs is an essential technical approach to improve microalgae biomass and lipid production.

Limitations and future perspectives of PBRs

While many alternative configurations have been shown to be effective for microalgae growth, the major drawback is the high-power consumption and other operating costs of PBRs. In an effort to compare each type of PBRs, Vo et al. [181] summarized their disadvantages as follows. For example, flat plate PBRs require more space and land, have photoinhibition effects and dark zone formation, leading to high construction and energy costs. For column PBRs, which prefer internal illumination and are more costly to mix, the surface-to-volume ratio is generally lower. The utilization of tubular PBRs poses the challenge of preventing system overheating and fouling, as well as high operational costs. Similarly, soft-frame PBRs are prone to issues such as fragile materials, expensive components, culture leakage, and poor mixing resulting from the formation of dead zones. Despite being regarded as the most promising cultivation system in recent years, hybrid PBRs systems also face problems, such as membrane fouling and negative energy balance.

If the performance of the outdoor lipid production process is described in terms of productivity and yield, and the calculation is based on ground area, then theoretically the parameters obtained from the pilot plant can be applied to the full-scale plant. However, microalgae batch lipid production pilot tests are usually conducted in single plate reactors, which means that the application of these parameters to full-scale plants is difficult [182]. Naturally, without consistent data on both biomass productivity and lipid productivity, it becomes challenging to determine which types of PBRs are better suited for microalgae biofuel production. The optimal designs of PBRs are contingent upon the microalgae species being used and the intended final metabolite. There are multiple parameters within PBRs that still require optimization to improve lipid production. It is clear that sustainable development and scaling of PBRs will be the central focus of future research.

Genetic engineering of microalgae to increase lipid production

Microalgal energy density is relatively low. This is due to the fact that microalgal cells are mainly composed of water, proteins, lipids, sugars, etc., with water accounting for a large proportion, and proteins and lipids being relatively scarce in comparison. This leads to a need for greater input to produce the same amount of energy, which in turn reduces the unit value of the product, indirectly increasing the production cost. It is worth mentioning that genetic engineering can cut the cost of microalgal biofuels by 15–20% [183]. Most studies using genetic engineering to induce lipid accumulation have concentrated on either mutagenizing a single target gene or engineering multiple genes. In general, genes are either knocked out or knocked in to achieve desired outcomes, and they are often of great importance in lipid metabolism. Modifying metabolic pathways involves the up-regulation or down-regulation of transcriptional and translational genes [184]. An example of genetic engineering in microalgae is the modification of N. oleoabundans by introducing an endogenous key enzyme called diacylglycerol acyltransferase 2 (NeoDGAT2). It was observed that overexpression of NeoDGAT2 led to an increase in TAG content and productivity in N. oleoabundans [73]. Target-specific knockout of the phospholipase A2 gene in C. reinhardtii improved overall lipid production by up to 64.25% [185]. Overexpressing Malonyl CoA-acylcarrier protein transacylase gene and glycerol-3-phosphate acyltransferases gene promoted neutral lipid accumulation in microalgae, too [186, 187]. Unlike green algae, cyanobacteria are prone to stress incurred due to the synthesis of biofuel compounds [188]. Therefore, the focus of the genetic engineering of cyanobacteria is the transfer of biofuel compounds. In Synechocystis sp. PCC6803, overexpression of the thioesterase gene has been shown to stimulate the significant secretion of free FAs into the extracellular environment [189, 190]. Similar findings have also been observed in Synechococcus elongatus PCC7942 and Synechococcus sp. PCC7002 [191, 192]. Researchers have been working tirelessly to develop new genetic engineering strategies to further enhance lipid accumulation in microalgae. Table 2 shows some key features of the genetic engineering of microalgae for enhanced biofuel production. It can be observed that genetic editing is driving microalgae towards a positive direction in biofuel production. However, this requires a thorough understanding of microalgae metabolic pathways. The development of omics provides abundant background information for targeted production of microalgae products, while the evolution of genetic engineering tools also offers technical support in reducing the cost of microalgae biofuel production.

Table 2 Biotechnological modifications of microalgae for enhanced biofuel production

Types of genetic engineering resources in microalgae

Algomics resources

Omics techniques for algae, known as ‘algomics’, including transcriptomics, genomics, proteomics, metabolomics, and metagenomics, prove to be highly valuable in assisting the design of experiments and predicting potential interactions and outcomes in algae research [218]. The integration of omics methodologies expands the scope of research for addressing stress-related challenges in lipid production. The combination of these omics approaches and bioinformatics tools and databases will be crucial in realizing economically viable microalgal-based biofuels. Furthermore, the integration of omics resources enables microalgae to produce a wider range of metabolites and bioactive compounds, accelerating the discovery and utilization of new microalgal species resources.

Genome sequencing and analysis play a vital role in comprehending and understanding microalgal systems [103]. The complete genome sequence of the cyanobacterium Synechocystis sp. PCC6803 was published as early as 1996, providing precise genetic information for gene modification [219]. In contrast, research on the genomes of eukaryotic microalgae started later. In the beginning, the whole genome sequence of microalgal genome projects have been generated only for a limited species, namely, C. reinhardtii, T. pseudonana, and P. tricornutum, but the number of publicly available microalgal sequenced genomes is escalating currently [220, 221]. Investigators have used insights from the genome of C. reinhardtii to perform functional annotation of lipid biosynthetic genes in microalgae [222]. By genome editing, the gene UDP-glucose pyrophosphorylase was disrupted and a 45-fold of TAG accumulation was found in P. tricornutum [223]. With the development of next-generation sequencing technology, genome mining of microalgal strains has become much more economical and reliable. Recently, an initiated genome sequencing project called 10KP aims to sequence the genomes of at least 1000 green algae (microalgae and macroalgae) as well as 3000 photosynthetic and non-photosynthetic protists [224]. As the number of published microalgae genome sequences increases, the evolution and adaptation of microalgae can be unraveled, and metabolic engineering strategies can be employed to reduce wet lab work. Besides, there are three web-based resources available for algal genomics, including pico-PLAZA (http://bioinformatics.psb.ugent.be/pico-plaza/), AlgaePath (http://algaepath.itps.ncku.edu.tw), and ALCOdb (http://alcodb.jp). These genomics resources provide genomic information, intuitive tools for functional genomics, gene co-expression data, and details of gene expression prediction based on metabolic pathways in some microalgae [225,226,227]. To this end, genome editing tools have been added to develop or increase the productive efficiency of biofuels and other valuable products in sustainable and renewable ways. Insertion, deletion, mutation, gene knockdown, and gene knockout are common strategies used in genome editing [228].

Transcriptomics is a valuable tool that can reveal details about gene expression by analyzing RNA transcripts, providing insights into the functions of various microalgae strains. Transcriptomic data are particularly beneficial as it offers a comprehensive reference data set for the discovery of new genes and the construction of metabolic models through computational analysis. The Marine Microbial Eukaryote Transcriptome Sequencing Project has conducted transcriptome sequencing on almost 700 marine microbial species, including 140 microalgae species [229]. Subsequent to sequencing, decontamination, pooling, and dereplication methods have been employed to refine these sequences, rendering them more amenable to analysis, with lower levels of contamination, redundancy, and improved balance in the resulting phylogenies [230]. In recent years, analysis of gene expression involved in lipid production at the transcriptome level in microalgae has offered valuable insights into metabolic pathways, as well as contextual information regarding modifications at the genomic level in algae [231,232,233,234].

In contrast to transcriptomics, proteomics is concerned with regulatory pathways at the post-transcriptional level. Proteomics provides quantitative data on protein expression under varying experimental conditions, helping to elucidate how protein expression can be upregulated or downregulated in response to environmental conditions that can affect both the production of a specific product and the survival of the cell [235]. Proteomic studies have identified a variety of proteins involved in lipid enhancement. In the case of N. oculate, it has been observed that organic carbon and nitrogen derived from the breakdown of proteins and pigments are primarily directed towards the FAs synthesis [236]. Nonoyama et al. [237] reported the detection of membrane trafficking-related proteins from the proteome involved in TAG hydrolysis in oleaginous diatoms. A proteomic analysis of C. zofingiensis has shown that structural proteins are present in high abundance and that enzymes involved in lipid metabolism work in concert with other lipid metabolism-related enzymes localized in ER [54]. The proteins involved in lipid biosynthesis and in FAs oxidation have been up-regulated under hypersaline conditions in Microchloropsis gaditana [238]. Furthermore, 29 putative lipid droplet-localizing proteins of Aurantiochytrium limacinum have been identified despite proteomics and lipidomics analyses, and approximately half of the proteins were accounted for by thraustochytrid-specific lipid droplet protein 1, which was thought as a structural protein in the lipid droplet [239]. There were certainly studies that combine the results of transcriptomic and proteomic analyses to analyze the molecular mechanisms of lipid accumulation in microalgae, and the availability of quantitative data under defined experimental conditions provided strategic insights for strain improvement in microalgae [240, 241].

Metabolomics focuses on the analysis of low molecular weight compounds involved in the maintenance of cellular biological processes, to target processes or pathways and explain their mechanisms. Similar to transcriptomic and proteomic studies in microalgae, it is also desirable to investigate lipid metabolism under different environmental conditions though metabolomic to achieve efficient lipid production. Up-regulated metabolic fluxes associated with increased lipid biosynthesis were found in a comparative time course metabolomics analysis of a C. sorokiniana mutant [242]. Metabolomics was used to identify the biochemical changes induced by antibiotics in S. obliquus, and it was found that norfloxacin triggers the shift of additional common precursors from the starch pathway to the FAs synthesis pathway, resulting in a shift in carbon allocation from carbohydrates to lipids [243]. Moreover, the metabolomic analysis has indicated that the intermediates in glycolysis and the tricarboxylic acid cycle contribute to the accumulation of astaxanthin and lipids in Haematococcus pluvialis treated with melatonin [244]. Metabolomics has been proven to be instrumental in microalgal research, but there is no dedicated database for microalgal metabolomics [33].

The advancing research in algomics is opening up new possibilities for microalgae biofuel production. Particularly, the integration of multi-omics approaches allows for a comprehensive understanding of the metabolic networks in microalgae. This approach reveals intricate regulatory relationships among key genes, proteins, and metabolic products, enabling precise control of production pathways and the attainment of desired end-products, such as lipids.

Mutant resources

Forward genetics can generate extensive pools of mutant phenotypes without prior knowledge about the genetics and metabolism of the target organism through random mutagenesis and adaptive laboratory evolution [245]. The use of random mutagenesis to obtain microalgal strains with the desired phenotype is widespread. Through methods, such as 60Co γ-ray, UV exposure, and chemical mutagenesis, mutants of N. gaditana [246, 247], N. oculate [248, 249] and Scenedesmus sp. [250, 251] have been successfully generated. These mutants have exhibited robust abilities for lipid accumulation. However, random mutagenesis is non-specific, unstable and even lethal or at least detrimental to the mutagenized organisms [245]. Therefore, appropriate selection methods, such as fluorescence-activated cell sorting, are needed to prevent phenotypic reversion. Similar to random mutagenesis, adaptive laboratory evolution is another cost-effective approach used for improving microalgal strains. In this model, microalgae cells are subjected to continuous selective pressure for a long period to obtain desired traits. As mentioned earlier in this study, microalgae can induce high lipid production under nutrient deficiencies, saline stress, high light and temperatures, and examples of how microalgae adapt to these stresses will not be mentioned here.

Insertional mutagenesis necessitates the presence of a DNA transformation protocol specific to each organism, as well as the identification of the mutation site associated with a particular mutant phenotype by the presence of selective markers and/or genetic or phenotypic tags [252]. Metabolism can be diverted to the lipid synthesis pathway by insertion and integration into the microalgal genome of a foreign DNA sequence present with a selective marker [252]. Mutant libraries are good tools for studying the function of uncharacterized genes and are also very important for reverse genetic research. Insertional mutagenesis by random non-homologous end joining is the preferred method of choice for generating mutant libraries [253]. The studies found an increase in FAME and lipids in the insertional mutants of both N. salina and N. oceanica [254, 255]. However, due to the lack of efficient transformation and genetic manipulation protocols, the application range of microalgae mutant libraries are far less wide than that of Arabidopsis thaliana and yeast [33]. At present, there are 2 random insertion mutant libraries of Chlamydomonas. Chlamydomonas Resource Center (www.chlamycollection.org/products/clip-strains) applied over 62,000 insertional mutants of C. reinhardtii that covers 83% of nuclear protein-encoding genes. Institute of Hydrobiology (Chinese Academy of Sciences) built an indexed mutant library, including ~ 150,000 C. reinhardtii insertional mutants [256]. As an important microalgae resource, the establishment of mutant libraries still needs to solve the problems of efficient identification of insertion sites.

Methods of genetic engineering in microalgae

Transformation techniques

Genetic engineering encompasses several steps, including transformation, which involves the introduction of foreign DNA into the target cell. This process entails the penetration of DNA into the cell, the integration of the foreign DNA into the genome of the target cell, and finally, the stable and desired expression of the integrated DNA [257]. The efficacy of transformation is highly contingent upon the microalgal species, and it is necessary to carefully choose both the genetic modification and transformation methods based on the specific species and type of modification [103]. Several transformation methods have been utilized to introduce DNA into microalgal cells, including the use of artificial transposons, electroporation, particle bombardment, sonication, viruses, bacterial infection, silicon carbide whiskers, glass bead-based agitation, and agrobacterium-mediated transformations [258]. Among them, the glass bead-based transformation was performed early in C. reinhardtii [259], and the discovery of cell wall-deficient mutants made transformation easier to achieve. Electroporation and particle bombardment are considered to have the highest transformation efficiency, and neither method requires prior removal of microalgal cell walls [260]. Currently, some examples have been observed to increase the content of neutral lipids and total lipids in microalgae by electroporation and particle bombardment [207, 261, 262]. Furthermore, agrobacterium-mediated transformation results in a higher rate of false-positive transformants, thus it does not have a significant advantage over electroporation methods in microalgae. Other methods of DNA delivery, such as agitation with silicon carbide whiskers, artificial transposons or sonication, have been reported, but they have not been widely adopted for lipid accumulation.

Genome editing tools

Novel genome-editing tools, such as RNA interference (RNAi), zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR/Cas9), have been employed to improve the quality and quantity of desired products in microalgae [184]. RNAi is a powerful tool used for gene silencing and modulating gene functions [184]. Levitan et al. [263] used RNAi to knock down the expression of the nitrate reductase gene in P. tricornutum to obtain a larger lipid content. In the diatom T. pseudonana, strains with a targeted knockdown of a multifunctional lipase/phospholipase/acyltransferase have been shown to exhibit growth patterns similar to the wild type and demonstrate increased lipid content under both continuous light and alternating light/dark conditions [264]. ZFNs is an initial tool for genome editing in microalgae and has been used successfully in C. reinhardtii in recent years, but is rarely used to study lipid accumulation [265,266,267]. Although TALENs is feasible for enhancing lipid production in microalgae [268], similar to RNAi and ZFNs, it is still limited by their tedious design steps and high off-target events, so CRISPR–Cas9 came into being [269, 270].

In the CRISPR–Cas9 system, the Cas9 nuclease creates double-stranded breaks in DNA sequences that are determined by the guide RNA, making microalgal genome editing more efficient, accurate and simple [271, 272]. A significant amount of effort has been invested in mitigating off-target mutations and cytotoxicity related to the use of Cas9 ribonucleoproteins. Researchers have established CRISPR–Cas9-based targeted gene knockout methods in microalgae, such as C. reinhardtii and N. oceanica [267, 273]. In addition, this method has been used to explore ways to enhance lipid accumulation in microalgae and has been demonstrated in N. gaditana [274], Tetraselmis sp. [275], P. tricornutum [276], and other microalgae. The rapid pace of technological progress provides hope that CRISPR/Cas will soon be established as a routine and trustworthy technique for genome editing in any type of microalga.

Limitation and risk of genetic engineering microalgae

As previously noted, there have been numerous experiments and studies conducted on the genetic engineering methods applied to various microalgal species. The application of genetic engineering in microalgae holds immense potential for boosting lipid production. An issue with working with GM microalgae is the potential environmental and ethical impacts of their release. Unlike other GM organisms that are cultivated in controlled environments for high-value products, GM microalgae in outdoor cultivation systems can escape into the environment through means, such as wind, water, birds, animals, natural disasters, or accidents [277]. This could have unintended consequences that are difficult to predict and control. Therefore, several factors should be taken into consideration when evaluating the potential negative ecological effects of GM microalgae and their impact on the environment and human health. These factors include potential changes to food web structure, displacement of native species such as phytoplankton, local extinctions, harmful algal blooms, and serious societal, cultural, and economic impacts in the case of toxic strains [278]. To mitigate the potential risks associated with the use of GM microalgae, both regulations for their use and strict biosecurity measures are necessary to ensure the safe importation of both foreign GM and wild-type species into the local environment. Besides, there is a need for the development of ‘suicide genes’ to control the spread of hazardous algae strains in case of an accidental release into the environment.

Another significant challenge is the compatibility of transgenic microalgae with large-scale cultivation, as the conditions required for their growth and cultivation may be difficult to replicate at a larger scale. Upon exposure to the conditions of large-scale cultivation, the strains encounter a wider range of conditions that differ from the controlled laboratory environment, which in turn affects their transgenic properties. Tabatabaei et al. [279] highlighted that lower growth rates and poor gene quality are significant challenges in genetic engineering that could hinder the commercialization of algae on a global scale. In addition, some of the genetic engineering techniques used for microalgae, such as homologous recombination, can result in long and complex constructions, which may impact the stability and functionality of the modified microalgal strains. As a result, their productivity in outdoor cultivation is often not as high as it is under optimized laboratory conditions. To date, no reports of outdoor cultivation of GM microalgae have been made due to the various risks and uncertainties associated with open cultivation [280].

Microalgal biofuel production opportunities in China

China is the largest energy-consuming country in the world. In 2021, China consumed 157.65 exajoules (EJ) of primary energy, an increase of 10 EJ from 2020 [1]. The rapid economic growth has led to an increasing demand for energy in China, thus the need to enhance the development and utilization of renewable energy and strive towards energy transformation. According to the report, the consumption of renewable energy in China increased by 32.9% in 2021 compared to 2020, indicating efforts made by China in energy transformation [1]. It is expected that the share of renewable energy in China will reach 13% by 2030 [281]. Biofuel is one of the important types of renewable energy, which has been paid attention to in recent years. The main types of biofuels in China include biodiesel, bioethanol, biogas, and biohydrogen, among others. In 2021, the consumption of biofuels was 48 thousand barrels of oil equivalent per day, with a biofuels production of 64 thousand barrels of oil equivalent per day [1]. Despite the increase in the production and consumption of biofuels in China, the country still has a long way to go in comparison with the United States, Brazil, and the European Union, which have set more ambitious targets and made greater progress in incorporating biofuels into their energy mix. In addition, the Chinese government is implementing measures to advance the cultivation and utilization of biofuels and has instituted a number of policies. In 2014, the National Energy Administration of China issued the ‘Biodiesel Industry Development Policy’, which proposed exploring and optimizing microalgae cultivation and lipid extraction processes, to achieve a technological breakthrough in microalgae biodiesel production. In 2016, the ‘CHINA: 13th Five-Year Plan for Biomass Energy Development’ proposed promoting the large-scale development of biogas and steadily developing biomass power generation. In 2017, the Chinese government issued a policy to expand the production of bioethanol and promote the use of ethanol gasoline for vehicles. The ‘2021 Work Plan for Biomass Power Generation Project Construction’ stated that the total central subsidy for biomass power generation in 2021 is CNY 2.5 billion. China aspires to harness the potential of biofuels and other sustainable energy sources to fulfill its expanding energy needs, strengthen its energy security, decrease its reliance on non-renewable fuels, and decrease its carbon emissions. Presently, China’s utilization of biofuels on a commercial scale remains largely at the second-generation level, with third- and fourth-generation biofuels mainly in the research stage [262, 282, 283]. Table 3 lists several large-scale bioethanol production companies in China and their production capacity. These companies mainly use materials, such as grains to produce bioethanol. However, like the problems faced by global biofuels, China also needs to adjust its reliance on grains for biofuels to alleviate the pressure on the food supply. Therefore, large-scale production of third- and fourth-generation biofuels using microalgae as feedstock is the key challenge that China needs to overcome in the future.

Table 3 List of bioethanol producing industries in China

It is undeniable that Chinese scientists have made great progress in the field of microalgal biofuel research, including the development of microalgal strains with high lipid content, exploration of cost-effective large-scale production methods, and improvement of microalgal lipid extraction processes, among others [19, 68, 88, 140, 173, 206, 208, 276]. In the future, with the continuous improvement of technology and the reduction of costs, the microalgal biofuel industry in China is expected to experience rapid development. At the same time, government policy support and the entry of industries are also important factors. Therefore, the prospects of microalgal biofuels in China are very optimistic.

Future outlook

Substantial progress has been made in the selection and cultivation optimization of lipid-producing microalgal strains in the past decade. However, significant improvements must be made to the economic viability of current microalgal biofuel production to compete favorably with fossil fuels before ultimately replacing them [284]. Enhancing microalgae dehydration and harvesting techniques can help achieve maximum biomass yield at minimum cost. The continual enrichment of genome sequences and mutant libraries, along with the improvement of gene editing tools and synthetic biology, will provide strong support for the development of microalgal potential as a renewable energy source. The development of novel techniques for lipid extraction and purification is a key area that requires significant progress in downstream processing. For example, the feasibility discussion on the application of AI models in the fucoxanthin extraction and quantification process provides valuable insights [285]. In addition, exploring the simultaneous production of biofuels and other high-value byproducts, as well as the use of microalgal biofuels in aerospace and other fields, are also promising directions for the future. Overall, the future of microalgal biofuels hinges on the developing cost-effective methods for the most efficient technology, which will accelerate the commercialization of this type of biofuel.

Undoubtedly, microalgae constitute a vast family with a diverse classification. This discussion has only scratched the surface, focusing primarily on the research and applications of eukaryotic microalgae in biofuel production. In addition, this article primarily emphasizes strategies for enhancing lipid accumulation in microalgae and does not delve into the processes of harvesting, extraction, purification, and other production processes. As a result, there are certain limitations to consider.

Conclusions

Microalgae biofuel presents an effective solution to the fossil fuel crisis, but it faces challenges in cost-effective large-scale production. The first step towards large-scale production is selecting robust and adaptable microalgae strains. While controlling cultivation conditions and adjusting PBRs parameters can influence the yield and performance of microalgae biofuel products, gene editing and multi-omics technologies offer more precise production routes. However, it is crucial to consider the environmental and human health risks associated with large-scale outdoor cultivation of GM microalgae. China still has significant work ahead to achieve large-scale production of microalgae biofuels, but the convergence of technology and market demand promises a bright outlook. In the future, the application of synthetic biology holds the potential to reduce the risks associated with GM microalgae and enhance the economic viability of their biofuel production.

Availability of data and materials

The data sets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

DG:

Diacylglycerol

TAG:

Triacylglyceride

FAs:

Fatty acids

SFA:

Saturated fatty acids

MUFA:

Monounsaturated fatty acids

PUFA:

Polyunsaturated fatty acids

C16:0:

Palmitic acid

C18:1:

Oleic acid

C18:2:

Linoleic acid

C18:3:

Linolenic acid

Rubisco:

Ribulose-1, 5-bisphosphate carboxylase/oxygenase

HL:

High light intensity

LEDs:

Light emitting diodes

PBRs:

Photobioreactors

NeoDGAT2:

Diacylglycerol acyltransferase 2

RNAi:

RNA interference

ZFNs:

Zinc-finger nucleases

TALENs:

Transcription activator-like effector nucleases

CRISPR/Cas9:

Clustered regularly interspaced short palindromic repeats

GM:

Genetically modified

EJ:

Exajoules

References

  1. BP Statistical Review of World Energy. 2022. https://www.bp.com/content/dam/bp/business-sites/en/global/corporate/pdfs/energy-economics/statistical-review/bp-stats-review-2022-full-report.pdf Accessed 18 Oct 2022.

  2. Lv Z, Chu AMY, McAleer M, Wong W-K. Modelling economic growth, carbon emissions, and fossil fuel consumption in china: cointegration and multivariate causality. Int J Environ Res Public Health. 2019;16(21):4176.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Maliha A, Abu-Hijleh B. A review on the current status and post-pandemic prospects of third-generation biofuels. Energy Syst. 2022;14:1–32.

    Google Scholar 

  4. Saini R, Osorio-Gonzalez CS, Brar SK, Kwong R. A critical insight into the development, regulation and future prospects of biofuels in Canada. Bioengineered. 2021;12(2):9847–59.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Dutta K, Daverey A, Lin J-G. Evolution retrospective for alternative fuels: first to fourth generation. Renew Energy. 2014;69:114–22.

    Article  CAS  Google Scholar 

  6. Aziz E, Anna S, Pascal L. Biofuels and the sustainability challenge: a global assessment of sustainability issues, trends and policies for biofuels and related feedstocks. Rome, Italy: Food And Agriculture Organization of The United Nations. 2013.

  7. Soccol CR, Vandenberghe LPS, Costa B, Woiciechowski AL, Carvalho JCD, Medeiros ABP, Francisco AM, Bonomi LJ. Brazilian biofuel program an overview. J Sci Indust Res. 2005;64(11):897–904.

    Google Scholar 

  8. Saladini F, Patrizi N, Pulselli FM, Marchettini N, Bastianoni S. Guidelines for emergy evaluation of first, second and third generation biofuels. Renew Sustain Energy Rev. 2016;66:221–7.

    Article  Google Scholar 

  9. Bharti MK, Chalia S, Thakur P, Sridhara SN, Thakur A, Sharma PB. Nanoferrites heterogeneous catalysts for biodiesel production from soybean and canola oil: a review. Environ Chem Lett. 2021;19(5):3727–46.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Maeda Y, Yoshino T, Matsunaga T, Matsumoto M, Tanaka T. Marine microalgae for production of biofuels and chemicals. Curr Opin Biotechnol. 2017;50:111–20.

    Article  PubMed  Google Scholar 

  11. Lynch F, Santana-Sánchez A, Jämsä M, Sivonen K, Aro E-M, Allahverdiyeva Y. Screening native isolates of cyanobacteria and a green alga for integrated wastewater treatment, biomass accumulation and neutral lipid production. Algal Res. 2015;11:411–20.

    Article  Google Scholar 

  12. Plöhn M, Spain O, Sirin S, Silva M, Escudero-Oñate C, Ferrando-Climent L, Allahverdiyeva Y, Funk C. Wastewater treatment by microalgae. Physiol Plant. 2021;173:568–78.

    Article  PubMed  Google Scholar 

  13. Wang M, Liu H, Qiao K, Ye X, Takano T, Liu S, Bu Y. Exogenous NaHCO3 enhances growth and lipid accumulation of the highly NaHCO3-tolerant Nannochloris sp. JB17. J Appl Phycol. 2021;33:241–53.

    Article  CAS  Google Scholar 

  14. Tu Z, Liu L, Lin W, Xie Z, Luo J. Potential of using sodium bicarbonate as external carbon source to cultivate microalga in non-sterile condition. Bioresour Technol. 2018;266:109–15.

    Article  PubMed  CAS  Google Scholar 

  15. Chong JWR, Yew GY, Khoo KS, Ho S-H, Show PL. Recent advances on food waste pretreatment technology via microalgae for source of polyhydroxyalkanoates. J Environ Manage. 2021;293: 112782.

    Article  PubMed  CAS  Google Scholar 

  16. Kovacevic V, Wesseler J. Cost-effectiveness analysis of algae energy production in the EU. Energy Policy. 2010;38(10):5749–57.

    Article  Google Scholar 

  17. Al-lwayzy SH, Yusaf T, Al-Juboori RA. Biofuels from the fresh water microalgae Chlorella vulgaris (FWM-CV) for diesel engines. Energies. 2014;7(3):1829–51.

    Article  Google Scholar 

  18. Cho K, Hur S-P, Lee C-H, Ko K, Lee Y-J, Kim K-N, Kim M-S, Chung Y-H, Kim D, Oda T. Bioflocculation of the oceanic microalga Dunaliella salina by the bloom-forming dinoflagellate Heterocapsa circularisquama, and its effect on biodiesel properties of the biomass. Bioresour Technol. 2016;202:257–61.

    Article  PubMed  CAS  Google Scholar 

  19. Gui J, Chen S, Luo G, Wu Z, Fan Y, Yao L, Xu H. Nutrient deficiency and an algicidal bacterium improved the lipid profiles of a novel promising oleaginous dinoflagellate, Prorocentrum donghaiense, for biodiesel production. Appl Environ Microbiol. 2021;87(19):e01159-e11121.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Nascimento IA, Marques SSI, Cabanelas ITD, Pereira SA, Druzian JI, de Souza CO, Vich DV, de Carvalho GC, Nascimento MA. Screening microalgae strains for biodiesel production: lipid productivity and estimation of fuel quality based on fatty acids profiles as selective criteria. BioEnergy Res. 2013;6(1):1–13.

    Article  CAS  Google Scholar 

  21. Wu H, Miao X. Biodiesel quality and biochemical changes of microalgae Chlorella pyrenoidosa and Scenedesmus obliquus in response to nitrate levels. Bioresour Technol. 2014;170:421–7.

    Article  PubMed  CAS  Google Scholar 

  22. Xu D, Gao Z, Li F, Fan X, Zhang X, Ye N, Mou S, Liang C, Li D. Detection and quantitation of lipid in the microalga Tetraselmis subcordiformis (Wille) Butcher with BODIPY 505/515 staining. Bioresour Technol. 2013;127:386–90.

    Article  PubMed  CAS  Google Scholar 

  23. Hegel P, Martín L, Popovich C, Damiani C, Pancaldi S, Pereda S, Leonardi P. Biodiesel production from Neochloris oleoabundans by supercritical technology. Chem Eng Process. 2017;121:232–9.

    Article  CAS  Google Scholar 

  24. Ma Y, Wang Z, Yu C, Yin Y, Zhou G. Evaluation of the potential of 9 Nannochloropsis strains for biodiesel production. Bioresour Technol. 2014;167:503–9.

    Article  PubMed  CAS  Google Scholar 

  25. Santana-Sánchez A, Lynch F, Sirin S, Allahverdiyeva Y. Nordic cyanobacterial and algal lipids: Triacylglycerol accumulation, chemotaxonomy and bioindustrial potential. Physiol Plant. 2021;173(2):591–602.

    Article  PubMed  Google Scholar 

  26. Subramani L, Venu H. Evaluation of methyl ester derived from novel Chlorella emersonii as an alternative feedstock for DI diesel engine & its combustion, performance and tailpipe emissions. Heat Mass Transfer. 2019;55(5):1513–34.

    Article  CAS  Google Scholar 

  27. Popovich CA, Pistonesi M, Hegel P, Constenla D, Bielsa GB, Martín LA, Damiani MC, Leonardi PI. Unconventional alternative biofuels: quality assessment of biodiesel and its blends from marine diatom Navicula cincta. Algal Res. 2019;39: 101438.

    Article  Google Scholar 

  28. Tüccar G, Aydın K. Evaluation of methyl ester of microalgae oil as fuel in a diesel engine. Fuel. 2013;112:203–7.

    Article  Google Scholar 

  29. Chong JWR, Khoo KS, Chew KW, Ting H-Y, Show PL. Trends in digital image processing of isolated microalgae by incorporating classification algorithm. Biotechnol Adv. 2023;63: 108095.

    Article  PubMed  CAS  Google Scholar 

  30. Saravanan A, Deivayanai VC, Senthil Kumar P, Rangasamy G, Varjani S. CO2 bio-mitigation using genetically modified algae and biofuel production towards a carbon net-zero society. Bioresour Technol. 2022;363: 127982.

    Article  PubMed  CAS  Google Scholar 

  31. Snow AA, Smith VH. Genetically engineered algae for biofuels: a key role for ecologists. Bioscience. 2012;62(8):765–8.

    Article  Google Scholar 

  32. Roy Chong JW, Tan X, Khoo KS, Ng HS, Jonglertjunya W, Yew GY, Show PL. Microalgae-based bioplastics: future solution towards mitigation of plastic wastes. Environ Res. 2022;206: 112620.

    Article  PubMed  CAS  Google Scholar 

  33. Kumar G, Shekh A, Jakhu S, Sharma Y, Sharma TR. Bioengineering of microalgae: recent advances, perspectives, and regulatory challenges for industrial application. Front Bioeng Biotechnol. 2020;8:914.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Chong JWR, Khoo KS, Yew GY, Leong WH, Lim JW, Lam MK, Ho Y-C, Ng HS, Munawaroh HSH, Show PL. Advances in production of bioplastics by microalgae using food waste hydrolysate and wastewater: a review. Bioresour Technol. 2021;342: 125947.

    Article  PubMed  CAS  Google Scholar 

  35. Shekh AY, Shrivastava P, Gupta A, Krishnamurthi K, Devi SS, Mudliar SN. Biomass and lipid enhancement in Chlorella sp. with emphasis on biodiesel quality assessment through detailed FAME signature. Bioresour Technol. 2016;201:276–86.

    Article  PubMed  CAS  Google Scholar 

  36. Hoekman SK, Broch A, Robbins C, Ceniceros E, Natarajan M. Review of biodiesel composition, properties, and specifications. Renew Sustain Energy Rev. 2012;16(1):143–69.

    Article  CAS  Google Scholar 

  37. Hu Q, Sommerfeld M, Jarvis E, Ghirardi M, Posewitz M, Seibert M, Darzins A. Microalgal triacylglycerols as feedstocks for biofuel production: perspectives and advances. Plant J. 2008;54(4):621–39.

    Article  PubMed  CAS  Google Scholar 

  38. Aizouq M, Peisker H, Gutbrod K, Melzer M, Hölzl G, Dörmann P. Triacylglycerol and phytyl ester synthesis in Synechocystis sp. PCC6803. Proceed Nat Acad Sci. 2020;117(11):6216–22.

    Article  CAS  Google Scholar 

  39. Sheng J, Vannela R, Rittmann BE. Evaluation of methods to extract and quantify lipids from Synechocystis PCC 6803. Bioresour Technol. 2011;102(2):1697–703.

    Article  PubMed  CAS  Google Scholar 

  40. Hewelt-Belka W, Kot-Wasik Á, Tamagnini P, Oliveira P. Untargeted lipidomics analysis of the cyanobacterium Synechocystis sp. PCC 6803: lipid composition variation in response to alternative cultivation setups and to gene deletion. Int J Mol Sci. 2020;21(23):8883.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Nagappan S, Bhosale R, Duc Nguyen D, Pugazhendhi A, Tsai P-C, Chang SW, Ponnusamy VK, Kumar G. Nitrogen-fixing cyanobacteria as a potential resource for efficient biodiesel production. Fuel. 2020;279: 118440.

    Article  CAS  Google Scholar 

  42. Fuad Hossain M, Ratnayake RR, Mahbub S, Kumara KLW, Magana-Arachchi DN. Identification and culturing of cyanobacteria isolated from freshwater bodies of Sri Lanka for biodiesel production. Saudi J Biol Sci. 2020;27(6):1514–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Uma VS, Gnanasekaran D, Lakshmanan U, Dharmar P. Survey and isolation of marine cyanobacteria from eastern coast of India as a biodiesel feedstock. Biocatal Agric Biotechnol. 2020;24: 101541.

    Article  Google Scholar 

  44. Damiani MC, Popovich CA, Constenla D, Leonardi PI. Lipid analysis in Haematococcus pluvialis to assess its potential use as a biodiesel feedstock. Bioresour Technol. 2010;101(11):3801–7.

    Article  PubMed  CAS  Google Scholar 

  45. Popovich CA, Damiani C, Constenla D, Martínez AM, Freije H, Giovanardi M, Pancaldi S, Leonardi PI. Neochloris oleoabundans grown in enriched natural seawater for biodiesel feedstock: evaluation of its growth and biochemical composition. Bioresour Technol. 2012;114:287–93.

    Article  PubMed  CAS  Google Scholar 

  46. Yu N, Dieu LTJ, Harvey S, Lee D-Y. Optimization of process configuration and strain selection for microalgae-based biodiesel production. Bioresour Technol. 2015;193:25–34.

    Article  PubMed  CAS  Google Scholar 

  47. Fawzy MA, El-Otify AM, Adam MS, Moustafa SSA. The impact of abiotic factors on the growth and lipid accumulation of some green microalgae for sustainable biodiesel production. Environ Sci Pollut Res. 2021;28(31):42547–61.

    Article  CAS  Google Scholar 

  48. George B, Pancha I, Desai C, Chokshi K, Paliwal C, Ghosh T, Mishra S. Effects of different media composition, light intensity and photoperiod on morphology and physiology of freshwater microalgae Ankistrodesmus falcatus – a potential strain for bio-fuel production. Bioresour Technol. 2014;171:367–74.

    Article  PubMed  CAS  Google Scholar 

  49. He Q, Yang H, Hu C. Optimizing light regimes on growth and lipid accumulation in Ankistrodesmus fusiformis H1 for biodiesel production. Bioresour Technol. 2015;198:876–83.

    Article  PubMed  CAS  Google Scholar 

  50. Blifernez-Klassen O, Chaudhari S, Klassen V, Wördenweber R, Steffens T, Cholewa D, Niehaus K, Kalinowski J, Kruse O. Metabolic survey of Botryococcus braunii: impact of the physiological state on product formation. PLoS ONE. 2018;13(6): e0198976.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Fang L, Sun D, Xu Z, He J, Qi S, Chen X, Chew W, Liu J. Transcriptomic analysis of a moderately growing subisolate Botryococcus braunii 779 (Chlorophyta) in response to nitrogen deprivation. Biotechnol Biofuels. 2015;8:130.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Behl K, SeshaCharan P, Joshi M, Sharma M, Mathur A, Kareya MS, Jutur PP, Bhatnagar A, Nigam S. Multifaceted applications of isolated microalgae Chlamydomonas sp. TRC-1 in wastewater remediation, lipid production and bioelectricity generation. Bioresour Technol. 2020;304:122993.

    Article  PubMed  CAS  Google Scholar 

  53. Kato Y, Ho S-H, Vavricka CJ, Chang J-S, Hasunuma T, Kondo A. Evolutionary engineering of salt-resistant Chlamydomonas sp. strains reveals salinity stress-activated starch-to-lipid biosynthesis switching. Bioresour Technol. 2017;245:1484–90.

    Article  PubMed  CAS  Google Scholar 

  54. Wang X, Wei H, Mao X, Liu J. Proteomics analysis of lipid droplets from the oleaginous alga Chromochloris zofingiensis reveals novel proteins for lipid metabolism. Genom Proteom Bioinformat. 2019;17(3):260–72.

    Article  Google Scholar 

  55. Chen J-H, Wei D, Lim P-E. Enhanced coproduction of astaxanthin and lipids by the green microalga Chromochloris zofingiensis: selected phytohormones as positive stimulators. Bioresour Technol. 2020;295:122242.

    Article  PubMed  CAS  Google Scholar 

  56. Mao X, Wu T, Sun D, Zhang Z, Chen F. Differential responses of the green microalga Chlorella zofingiensis to the starvation of various nutrients for oil and astaxanthin production. Bioresour Technol. 2018;249:791–8.

    Article  PubMed  CAS  Google Scholar 

  57. Yu H, Kim J, Lee C. Nutrient removal and microalgal biomass production from different anaerobic digestion effluents with Chlorella species. Sci Rep. 2019;9(1):6123.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Ren X, Zhao X, Turcotte F, Deschênes J-S, Tremblay R, Jolicoeur M. Current lipid extraction methods are significantly enhanced adding a water treatment step in Chlorella protothecoides. Microb Cell Fact. 2017;16(1):26.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Li T, Gargouri M, Feng J, Park J-J, Gao D, Miao C, Dong T, Gang DR, Chen S. Regulation of starch and lipid accumulation in a microalga Chlorella sorokiniana. Bioresour Technol. 2015;180:250–7.

    Article  PubMed  CAS  Google Scholar 

  60. Kumar V, Nanda M, Kumar S, Chauhan PK. The effects of ultraviolet radiation on growth, biomass, lipid accumulation and biodiesel properties of microalgae. Energy Sources Part A Recov Utilizat Environ Effects. 2018;40(7):787–93.

    CAS  Google Scholar 

  61. Arora N, Philippidis GP. Insights into the physiology of Chlorella vulgaris cultivated in sweet sorghum bagasse hydrolysate for sustainable algal biomass and lipid production. Sci Rep. 2021;11(1):6779.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Deng X-Y, Gao K, Addy M, Li D, Zhang R-C, Lu Q, Ma Y-W, Cheng Y-L, Chen P, Liu Y-H, et al. Cultivation of Chlorella vulgaris on anaerobically digested swine manure with daily recycling of the post-harvest culture broth. Bioresour Technol. 2018;247:716–23.

    Article  PubMed  CAS  Google Scholar 

  63. Shiels K, Tsoupras A, Lordan R, Nasopoulou C, Zabetakis I, Murray P, Saha SK. Bioactive lipids of marine microalga Chlorococcum sp. SABC 012504 with anti-inflammatory and anti-thrombotic activities. Mar Drugs. 2021;19(1):28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Sabeela Beevi U, Sukumaran RK. Cultivation of the fresh water microalga Chlorococcum sp. RAP13 in sea water for producing oil suitable for biodiesel. J Appl Phycol. 2015;27(1):141–7.

    Article  Google Scholar 

  65. Ndikubwimana T, Zeng X, Murwanashyaka T, Manirafasha E, He N, Shao W, Lu Y. Harvesting of freshwater microalgae with microbial bioflocculant: a pilot-scale study. Biotechnol Biofuels. 2016;9(1):47.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Ahmed RA, He M, Aftab RA, Zheng S, Nagi M, Bakri R, Wang C. Bioenergy application of Dunaliella salina SA 134 grown at various salinity levels for lipid production. Sci Rep. 2017;7(1):8118.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Talebi AF, Tohidfar M, Mousavi Derazmahalleh SM, Sulaiman A, Baharuddin AS, Tabatabaei M. Biochemical modulation of lipid pathway in microalgae dunaliella sp for biodiesel production. BioMed Res Int. 2015;2015:597198.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Liang M-H, Xue L-L, Jiang J-G. Two-stage cultivation of Dunaliella tertiolecta with glycerol and triethylamine for lipid accumulation: a viable way to alleviate the inhibitory effect of triethylamine on biomass. Appl Environ Microbiol. 2019;85(4):e02614-e2618.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Benhima R, El Arroussi H, Kadmiri IM, El Mernissi N, Wahby I, Bennis I, Smouni A, Bendaou N. Nitrate reductase inhibition induces lipid enhancement of Dunaliella Tertiolecta for biodiesel production. Sci World J. 2018;2018:6834725.

    Article  Google Scholar 

  70. Zhao K, Li Y, Yan H, Hu Q, Han D. Regulation of light spectra on cell division of the unicellular green alga Haematococcus pluvialis: insights from physiological and lipidomic analysis. Cells. 2022;11(12):1956.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Pang N, Fu X, Fernandez JSM, Chen S. Multilevel heuristic LED regime for stimulating lipid and bioproducts biosynthesis in Haematococcus pluvialis under mixotrophic conditions. Bioresour Technol. 2019;288: 121525.

    Article  PubMed  CAS  Google Scholar 

  72. Du Y, Schuur B, Brilman DWF. Maximizing lipid yield in Neochloris oleoabundans Algae extraction by stressing and using multiple extraction stages with N-Ethylbutylamine as switchable solvent. Ind Eng Chem Res. 2017;56(28):8073–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. 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 Fact. 2017;16(1):61.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Jaeger LD, Carreres BM, Springer J, Schaap PJ, Eggink G, Santos V, Wijffels RH, Martens DE. Neochloris oleoabundans is worth its salt: Transcriptomic analysis under salt and nitrogen stress. PLoS ONE. 2018;13(4):e0194834.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Muñoz CF, Weusthuis RA, D’Adamo S, Wijffels RH. Effect of single and combined expression of lysophosphatidic acid acyltransferase, Glycerol-3-Phosphate acyltransferase, and diacylglycerol acyltransferase on lipid accumulation and composition in Neochloris oleoabundans. Front Plant Sci. 2019. https://doi.org/10.3389/fpls.2019.01573.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Matich EK, Butryn DM, Ghafari M, del Solar V, Camgoz E, Pfeifer BA, Aga DS, Haznedaroglu BZ, Atilla-Gokcumen GE. Mass spectrometry-based metabolomics of value-added biochemicals from Ettlia oleoabundans. Algal Res. 2016;19:146–54.

    Article  Google Scholar 

  77. Saliu F, Magoni C, Torelli A, Cozza R, Lasagni M, Labra M. Omega-3 rich oils from microalgae: a chitosan mediated in situ transesterification method. Food Chem. 2021;337: 127745.

    Article  PubMed  CAS  Google Scholar 

  78. Koyande AK, Show P-L, Guo R, Tang B, Ogino C, Chang J-S. Enhanced microalgal lipid production with media engineering of potassium nitrate as a nitrogen source. Bioengineered. 2019;10(1):574–92.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Vendruscolo RG, Fagundes MB, Maroneze MM, do Nascimento TC, de Menezes CR, Barin JS, Zepka LQ, Jacob-Lopes E, Wagner R. Scenedesmus obliquus metabolomics: effect of photoperiods and cell growth phases. Bioproc Biosyst Eng. 2019;42(5):727–39.

    Article  CAS  Google Scholar 

  80. Ma C, Zhang Y-B, Ho S-H, Xing D-F, Ren N-Q, Liu B-F. Cell growth and lipid accumulation of a microalgal mutant Scenedesmus sp Z-4 by combining light/dark cycle with temperature variation. Biotechnol Biofuels. 2017;10(1):260.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Shuba Eyasu S, Kifle D. Microalgae to biofuels: ‘Promising’ alternative and renewable energy, review. Renew Sustain Energy Rev. 2018;81:743–55.

    Article  Google Scholar 

  82. D’Alessandro EB, Antoniosi Filho NR. Concepts and studies on lipid and pigments of microalgae: a review. Renew Sustain Energy Rev. 2016;58:832–41.

    Article  Google Scholar 

  83. Ferreira GF, Ríos Pinto LF, Maciel Filho R, Fregolente LV. A review on lipid production from microalgae: Association between cultivation using waste streams and fatty acid profiles. Renew Sustain Energy Rev. 2019;109:448–66.

    Article  CAS  Google Scholar 

  84. Krzemińska I, Szymańska M, Ciempiel W, Piasecka A. Auxin supplementation under nitrogen limitation enhanced oleic acid and MUFA content in Eustigmatos calaminaris biomass with potential for biodiesel production. Sci Rep. 2023;13(1):594.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Lee S-J, Go S, Jeong G-T, Kim S-K. Oil production from five marine microalgae for the production of biodiesel. Biotechnol Bioprocess Eng. 2011;16(3):561–6.

    Article  CAS  Google Scholar 

  86. Chiu S-Y, Kao C-Y, Tsai M-T, Ong S-C, Chen C-H, Lin C-S. Lipid accumulation and CO2 utilization of Nannochloropsis oculata in response to CO2 aeration. Bioresour Technol. 2009;100(2):833–8.

    Article  PubMed  CAS  Google Scholar 

  87. de Morais MG, Costa JAV. Isolation and selection of microalgae from coal fired thermoelectric power plant for biofixation of carbon dioxide. Energy Convers Manage. 2007;48(7):2169–73.

    Article  Google Scholar 

  88. Hui W, Wenjun Z, Wentao C, Lili G, Tianzhong L. Strategy study on enhancing lipid productivity of filamentous oleaginous microalgae Tribonema. Bioresour Technol. 2016;218:161–6.

    Article  PubMed  CAS  Google Scholar 

  89. Nakanishi A, Aikawa S, Ho S-H, Chen C-Y, Chang J-S, Hasunuma T, Kondo A. Development of lipid productivities under different CO2 conditions of marine microalgae Chlamydomonas sp. JSC4. Bioresour Technol. 2014;152:247–52.

    Article  PubMed  CAS  Google Scholar 

  90. Krishnan V, Uemura Y, Thanh NT, Khalid NA, Osman N, Mansor N. Three types of Marine microalgae and Nannocholoropsis oculata cultivation for potential source of biomass production. J Phys: Conf Ser. 2015;622: 012034.

    Google Scholar 

  91. Yanlin Z, Cheng Y, Junhan L, Guangrong H. Lipid production by a CO2-tolerant green microalga, Chlorella sp. MRA-1. J Microbiol Biotechnol. 2014;24(5):683–9.

    Article  Google Scholar 

  92. Ota M, Kato Y, Watanabe H, Watanabe M, Sato Y, Smith RL, Inomata H. Fatty acid production from a highly CO2 tolerant alga, Chlorocuccum littorale, in the presence of inorganic carbon and nitrate. Bioresour Technol. 2009;100(21):5237–42.

    Article  PubMed  CAS  Google Scholar 

  93. Bagchi S, Mallick N. Carbon dioxide biofixation and lipid accumulation potential of an indigenous microalga Scenedesmus obliquus (Turpin) Kützing GA 45 for biodiesel production. RSC Adv. 2016;6:29889–98.

    Article  CAS  Google Scholar 

  94. Shin YS, Choi HI, Choi JW, Lee JS, Sung YJ, Sim SJ. Multilateral approach on enhancing economic viability of lipid production from microalgae: a review. Bioresour Technol. 2018;258:335–44.

    Article  PubMed  CAS  Google Scholar 

  95. Artamonova EY, Vasskog T, Eilertsen HC. Lipid content and fatty acid composition of Porosira glacialis and Attheya longicornis in response to carbon dioxide (CO2) aeration. PLoS ONE. 2017;12(5): e0177703.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Helping carbon dependent industries reduce emissions through proven and scalable microalgal carbon capture technology. 2022.https://carbonbiocapture.com/technology/. Accessed 20 Oct 2023.

  97. Srinivasan R, Mageswari A, Subramanian P, Suganthi C, Chaitanyakumar A, Aswini V, Gothandam KM. Bicarbonate supplementation enhances growth and biochemical composition of Dunaliella salina V-101 by reducing oxidative stress induced during macronutrient deficit conditions. Sci Rep. 2018;8(1):6972.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Huang JJ, Bunjamin G, Teo ES, Ng DB, Lee YK. An enclosed rotating floating photobioreactor (RFP) powered by flowing water for mass cultivation of photosynthetic microalgae. Biotechnol Biofuels. 2016;9(1):218.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Singh RP, Yadav P, Kumar A, Hashem A, Al-Arjani A-BF, Abd_Allah EF, Rodríguez Dorantes A, Gupta RK. Physiological and Biochemical Responses of Bicarbonate Supplementation on Biomass and Lipid Content of Green Algae Scenedesmus sp. BHU1 Isolated From Wastewater for Renewable Biofuel Feedstock. Front Microbiol. 2022. Doi: https://doi.org/10.3389/fmicb.2022.839800

  100. Parichehreh R, Gheshlaghi R, Mahdavi MA, Kamyab H. Investigating the effects of eleven key physicochemical factors on growth and lipid accumulation of Chlorella sp. as a feedstock for biodiesel production. J Biotechnol. 2021;340:64–74.

    Article  PubMed  CAS  Google Scholar 

  101. Wang M, Ye X, Wang Y, Su D, Liu S, Bu Y. Transcriptome dynamics and hub genes of green alga Nannochloris sp. JB17 under NaHCO3 stress. Algal Res. 2021;54:102185.

    Article  Google Scholar 

  102. Peng X, Liu S, Zhang W, Zhao Y, Chen L, Wang H, Liu T. Triacylglycerol accumulation of Phaeodactylum tricornutum with different supply of inorganic carbon. J Appl Phycol. 2014;26(1):131–9.

    Article  CAS  Google Scholar 

  103. Alishah Aratboni H, Rafiei N, Garcia-Granados R, Alemzadeh A, Morones-Ramírez JR. Biomass and lipid induction strategies in microalgae for biofuel production and other applications. Microb Cell Fact. 2019;18(1):178.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Feng P, Xu Z, Qin L, Asraful Alam M, Wang Z, Zhu S. Effects of different nitrogen sources and light paths of flat plate photobioreactors on the growth and lipid accumulation of Chlorella sp. GN1 outdoors. Bioresour Technol. 2020;301:122762.

    Article  PubMed  CAS  Google Scholar 

  105. Negi S, Barry AN, Friedland N, Sudasinghe N, Subramanian S, Pieris S, Holguin FO, Dungan B, Schaub T, Sayre R. Impact of nitrogen limitation on biomass, photosynthesis, and lipid accumulation in Chlorella sorokiniana. J Appl Phycol. 2016;28(2):803–12.

    Article  CAS  Google Scholar 

  106. Chokshi K, Pancha I, Ghosh A, Mishra S. Nitrogen starvation-induced cellular crosstalk of ROS-scavenging antioxidants and phytohormone enhanced the biofuel potential of green microalga Acutodesmus dimorphus. Biotechnol Biofuels. 2017;10(1):60.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Li X, Chen K, He Y. In situ and non-destructive detection of the lipid concentration of Scenedesmus obliquus using hyperspectral imaging technique. Algal Res. 2020;45: 101680.

    Article  Google Scholar 

  108. Maltsev Y, Maltseva I, Maltseva S, Kociolek JP, Kulikovskiy M. A new species of freshwater algae Nephrochlamys yushanlensis sp. Nov. (Selenastraceae, Sphaeropleales) and its lipid accumulation during nitrogen and phosphorus starvation. J Phycol. 2021;57(2):606–18.

    Article  PubMed  CAS  Google Scholar 

  109. Liu T, Li Y, Liu F, Wang C. The enhanced lipid accumulation in oleaginous microalga by the potential continuous nitrogen-limitation (CNL) strategy. Bioresour Technol. 2016;203:150–9.

    Article  PubMed  CAS  Google Scholar 

  110. Anand J, Arumugam M. Enhanced lipid accumulation and biomass yield of Scenedesmus quadricauda under nitrogen starved condition. Bioresour Technol. 2015;188:190–4.

    Article  PubMed  CAS  Google Scholar 

  111. Jia J, Han D, Gerken HG, Li Y, Sommerfeld M, Hu Q, Xu J. Molecular mechanisms for photosynthetic carbon partitioning into storage neutral lipids in Nannochloropsis oceanica under nitrogen-depletion conditions. Algal Res. 2015;7:66–77.

    Article  Google Scholar 

  112. Dong H-P, Williams E, Wang D-Z, Xie Z-X, Hsia R-C, Jenck A, Halden R, Li J, Chen F, Place AR. Responses of Nannochloropsis oceanica IMET1 to long-term nitrogen starvation and recovery. Plant Physiol. 2013;162:1110–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Maltsev Y, Krivova Z, Maltseva S, Maltseva K, Gorshkova E, Kulikovskiy M. Lipid accumulation by Coelastrella multistriata (Scenedesmaceae, Sphaeropleales) during nitrogen and phosphorus starvation. Sci Rep. 2021;11(1):19818.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Yu Z, Pei H, Jiang L, Hou Q, Nie C, Zhang L. Phytohormone addition coupled with nitrogen depletion almost tripled the lipid productivities in two algae. Bioresour Technol. 2018;247:904–14.

    Article  PubMed  CAS  Google Scholar 

  115. Jouhet J, Lupette J, Clerc O, Magneschi L, Bedhomme M, Collin S, Roy S, MareÂchal E, ReÂbeille F. LC-MS/MS versus TLC plus GC methods: consistency of glycerolipid and fatty acid profiles in microalgae and higher plant cells and effect of a nitrogen starvation. PLoS ONE. 2017;12(8): e0182423.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Maltsev Y, Maltseva I, Maltseva S, Kociolek JP, Kulikovskiy M. Fatty acid content and profile of the novel strain of Coccomyxa elongata (Trebouxiophyceae, Chlorophyta) cultivated at reduced nitrogen and phosphorus concentrations. J Phycol. 2019;55(5):1154–65.

    Article  PubMed  CAS  Google Scholar 

  117. Lamers PP, Janssen M, De Vos RCH, Bino RJ, Wijffels RH. Carotenoid and fatty acid metabolism in nitrogen-starved Dunaliella salina, a unicellular green microalga. J Biotechnol. 2012;162(1):21–7.

    Article  PubMed  CAS  Google Scholar 

  118. Cheng J, Lu H, Huang Y, Li K, Huang R, Zhou J, Cen K. Enhancing growth rate and lipid yield of Chlorella with nuclear irradiation under high salt and CO2 stress. Bioresour Technol. 2016;203:220–7.

    Article  PubMed  CAS  Google Scholar 

  119. Shrestha N, Dandinpet KK, Schneegurt MA. Effects of nitrogen and phosphorus limitation on lipid accumulation by Chlorella kessleri str. UTEX 263 grown in darkness. J Appl Phycol. 2020;32(5):2795–805.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Li J, Han D, Wang D, Ning K, Jia J, Wei L, Jing X, Huang S, Chen J, Li Y, et al. Choreography of transcriptomes and lipidomes of nannochloropsis reveals the mechanisms of oil synthesis in microalgae. Plant Cell. 2014;26(4):1645–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Wang D, Lai Y-C, Karam AL, De los Reyes FL, Ducoste JJ. Dynamic modeling of microalgae growth and lipid production under transient light and nitrogen conditions. Environ Sci Technol. 2019;53(19):11560–8.

    Article  PubMed  CAS  Google Scholar 

  122. Longworth J, Wu D, Huete-Ortega M, Wright PC, Vaidyanathan S. Proteome response of Phaeodactylum tricornutum, during lipid accumulation induced by nitrogen depletion. Algal Res. 2016;18:213–24.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Arguelles E, Laurena A, Monsalud R, Martinez-Goss M. Fatty acid profile and fuel-derived physico-chemical properties of biodiesel obtained from an indigenous green microalga, Desmodesmus sp. (I-AU1), as potential source of renewable lipid and high quality biodiesel. J Appl Phycol. 2017;30(1):411–9.

    Article  Google Scholar 

  124. Yang L, Chen J, Qin S, Zeng M, Jiang Y, Hu L, Xiao P, Hao W, Hu Z, Lei A, et al. Growth and lipid accumulation by different nutrients in the microalga Chlamydomonas reinhardtii. Biotechnol Biofuels. 2018;11(1):40.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Feng P, Deng Z, Fan L, Hu Z. Lipid accumulation and growth characteristics of Chlorella zofingiensis under different nitrate and phosphate concentrations. J Biosci Bioeng. 2012;114(4):405–10.

    Article  PubMed  CAS  Google Scholar 

  126. Hounslow E, Kapoore RV, Vaidyanathan S, Gilmour DJ, Wright PC. The search for a lipid trigger: the effect of salt stress on the lipid profile of the model microalgal species Chlamydomonas reinhardtii for biofuels production. Curr Biotechnol. 2016;5(4):305–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Chokshi K, Pancha I, Ghosh A, Mishra S. Salinity induced oxidative stress alters the physiological responses and improves the biofuel potential of green microalgae Acutodesmus dimorphus. Bioresour Technol. 2017;244:1376–83.

    Article  PubMed  CAS  Google Scholar 

  128. Pandit PR, Fulekar MH, Karuna MSL. Effect of salinity stress on growth, lipid productivity, fatty acid composition, and biodiesel properties in Acutodesmus obliquus and Chlorella vulgaris. Environ Sci Pollut Res. 2017;24(15):13437–51.

    Article  CAS  Google Scholar 

  129. Hounslow E, Evans CA, Pandhal J, Sydney T, Couto N, Pham TK, Gilmour DJ, Wright PC. Quantitative proteomic comparison of salt stress in Chlamydomonas reinhardtii and the snow alga Chlamydomonas nivalis reveals mechanisms for salt-triggered fatty acid accumulation via reallocation of carbon resources. Biotechnol Biofuels. 2021;14(1):121.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Srivastava G, Goud VV. Salinity induced lipid production in microalgae and cluster analysis (ICCB 16-BR_047). Bioresour Technol. 2017;242:244–52.

    Article  PubMed  CAS  Google Scholar 

  131. You Z, Zhang Q, Peng Z, Miao X. Lipid droplets mediate salt stress tolerance in Parachlorella kessleri. Plant Physiol. 2019;181(2):510–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Mao X, Zhang Y, Wang X, Liu J. Novel insights into salinity-induced lipogenesis and carotenogenesis in the oleaginous astaxanthin-producing alga Chromochloris zofingiensis: a multi-omics study. Biotechnol Biofuels. 2020;13(1):73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Atikij T, Syaputri Y, Iwahashi H, Praneenararat T, Sirisattha S, Kageyama H, Waditee-Sirisattha R. Enhanced lipid production and molecular dynamics under salinity stress in green microalga Chlamydomonas reinhardtii (137C). Mar Drugs. 2019;17(8):484.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Graef M. Lipid droplet-mediated lipid and protein homeostasis in budding yeast. FEBS Lett. 2018;592(8):1291–303.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Mansour MMF, Salama KHA, Allam HYH. Role of the plasma membrane in saline conditions: lipids and proteins. Bot Rev. 2015;81(4):416–51.

    Article  Google Scholar 

  136. Guo X, Su G, Li Z, Chang J, Zeng X, Sun Y, Lu Y, Lin L. Light intensity and N/P nutrient affect the accumulation of lipid and unsaturated fatty acids by Chlorella sp. Bioresour Technol. 2015;191:385–90.

    Article  PubMed  CAS  Google Scholar 

  137. Atta M, Idris A, Bukhari A, Wahidin S. Intensity of blue LED light: a potential stimulus for biomass and lipid content in fresh water microalgae Chlorella vulgaris. Bioresour Technol. 2013;148:373–8.

    Article  PubMed  CAS  Google Scholar 

  138. Difusa A, Talukdar J, Kalita MC, Mohanty K, Goud VV. Effect of light intensity and pH condition on the growth, biomass and lipid content of microalgae Scenedesmus species. Biofuels. 2015;6(1–2):37–44.

    Article  CAS  Google Scholar 

  139. Liu J, Mao X, Zhou W, Guarnieri MT. Simultaneous production of triacylglycerol and high-value carotenoids by the astaxanthin-producing oleaginous green microalga Chlorella zofingiensis. Bioresour Technol. 2016;214:319–27.

    Article  PubMed  CAS  Google Scholar 

  140. Liao Q, Sun Y, Huang Y, Xia A, Fu Q, Zhu X. Simultaneous enhancement of Chlorella vulgaris growth and lipid accumulation through the synergy effect between light and nitrate in a planar waveguide flat-plate photobioreactor. Bioresour Technol. 2017;243:528–38.

    Article  PubMed  CAS  Google Scholar 

  141. Ruangsomboon S. Effect of light, nutrient, cultivation time and salinity on lipid production of newly isolated strain of the green microalga, Botryococcus braunii KMITL 2. Bioresour Technol. 2012;109:261–5.

    Article  PubMed  CAS  Google Scholar 

  142. Cuellar-Bermudez SP, Romero-Ogawa MA, Vannela R, Lai YS, Rittmann BE, Parra-Saldivar R. Effects of light intensity and carbon dioxide on lipids and fatty acids produced by Synechocystis sp. PCC6803 during continuous flow. Algal Res. 2015;12:10–6.

    Article  Google Scholar 

  143. Yaisamlee C, Sirikhachornkit A. Characterization of Chlamydomonas very high light-tolerant mutants for enhanced lipid production. J Oleo Sci. 2020;69(4):359–68.

    Article  PubMed  CAS  Google Scholar 

  144. Bialevich V, Zachleder V, Bišová K. The effect of variable light source and light intensity on the growth of three algal species. Cells. 2022;11(8):1293.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Zhang P, Xin Y, He Y, Tang X, Shen C, Wang Q, Lv N, Li Y, Hu Q, Xu J. Exploring a blue-light-sensing transcription factor to double the peak productivity of oil in Nannochloropsis oceanica. Nat Commun. 2022;13(1):1664.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Poliner E, Busch AWU, Newton L, Kim YU, Clark R, Gonzalez-Martinez SC, Jeong B-R, Montgomery BL, Farré EM. Aureochromes maintain polyunsaturated fatty acid content in Nannochloropsis oceanica. Plant Physiol. 2022;189(2):906–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Hultberg M, Jönsson HL, Bergstrand K-J, Carlsson AS. Impact of light quality on biomass production and fatty acid content in the microalga Chlorella vulgaris. Bioresour Technol. 2014;159:465–7.

    Article  PubMed  CAS  Google Scholar 

  148. Ra CH, Kang C-H, Jung J-H, Jeong G-T, Kim S-K. Enhanced biomass production and lipid accumulation of Picochlorum atomus using light-emitting diodes (LEDs). Bioresour Technol. 2016;218:1279–83.

    Article  PubMed  CAS  Google Scholar 

  149. Li D, Yuan Y, Cheng D, Zhao Q. Effect of light quality on growth rate, carbohydrate accumulation, fatty acid profile and lutein biosynthesis of Chlorella sp. AE10. Bioresour Technol. 2019;291:121783.

    Article  PubMed  CAS  Google Scholar 

  150. Kim SH, Sunwoo IY, Hong HJ, Awah CC, Jeong G-T, Kim S-K. Lipid and unsaturated fatty acid productions from three microalgae using nitrate and light-emitting diodes with complementary LED wavelength in a two-phase culture system. Bioprocess Biosyst Eng. 2019;42(9):1517–26.

    Article  PubMed  CAS  Google Scholar 

  151. Ma R, Thomas-Hall SR, Chua ET, Eltanahy E, Netzel ME, Netzel G, Lu Y, Schenk PM. LED power efficiency of biomass, fatty acid, and carotenoid production in Nannochloropsis microalgae. Bioresour Technol. 2018;252:118–26.

    Article  PubMed  CAS  Google Scholar 

  152. Calhoun S, Bell TS, Dahlin LR, Kunde Y, LaButti K, Louie KB, Kuftin A, Treen D, Dilworth D, Mihaltcheva S, et al. A multi-omic characterization of temperature stress in a halotolerant Scenedesmus strain for algal biotechnology. Communicat Bio. 2021;4:333.

    CAS  Google Scholar 

  153. Wei L, Huang X, Huang Z. Temperature effects on lipid properties of microalgae Tetraselmis subcordiformis and Nannochloropsis oculata as biofuel resources. Chin J Oceanol Limnol. 2015;33(1):99–106.

    Article  CAS  Google Scholar 

  154. Zachleder V, Kselíková V, Ivanov IN, Bialevich V, Vítová M, Ota S, Takeshita T, Kawano S, Bišová K. Supra-optimal temperature: an efficient approach for overaccumulation of starch in the green Alga Parachlorella kessleri. Cells. 2021;10(7):1806.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Chadova O, Skriptsova A, Velansky P. Effect of temperature and light intensity on the polar lipidome of endophytic brown algae Streblonema corymbiferum and Streblonema sp. In Vitro Mar Drugs. 2022;20(7):428.

    Article  PubMed  CAS  Google Scholar 

  156. Zhu S, Gu D, Lu C, Zhang C, Chen J, Yang R, Luo Q, Wang T, Zhang P, Chen H. Cold stress tolerance of the intertidal red alga Neoporphyra haitanensis. BMC Plant Biol. 2022;22(1):114.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Chua ET, Dal’Molin C, Thomas-Hall S, Netzel ME, Netzel G, Schenk PM. Cold and dark treatments induce omega-3 fatty acid and carotenoid production in Nannochloropsis oceanica. Algal Res. 2020;51: 102059.

    Article  Google Scholar 

  158. Los DA, Mironov KS, Allakhverdiev SI. Regulatory role of membrane fluidity in gene expression and physiological functions. Photosynth Res. 2013;116(2):489–509.

    Article  PubMed  CAS  Google Scholar 

  159. Bharathiraja B, Chakravarthy M, Ranjith Kumar R, Yogendran D, Yuvaraj D, Jayamuthunagai J, Praveen Kumar R, Palani S. Aquatic biomass (algae) as a future feed stock for bio-refineries: a review on cultivation, processing and products. Renew Sustain Energy Rev. 2015;47:634–53.

    Article  CAS  Google Scholar 

  160. Scranton MA, Ostrand JT, Fields FJ, Mayfield SP. Chlamydomonas as a model for biofuels and bio-products production. Plant J. 2015;82(3):523–31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Lee OK, Lee EY. Sustainable production of bioethanol from renewable brown algae biomass. Biomass Bioenergy. 2016;92:70–5.

    Article  CAS  Google Scholar 

  162. Biomass and Biofuels from Microalgae. Cham, Switzerland Springer Cham. 2015.

  163. Dogaris I, Welch M, Meiser A, Walmsley L, Philippidis G. A novel horizontal photobioreactor for high-density cultivation of microalgae. Bioresour Technol. 2015;198:316–24.

    Article  PubMed  CAS  Google Scholar 

  164. Schoepp NG, Stewart RL, Sun V, Quigley AJ, Mendola D, Mayfield SP, Burkart MD. System and method for research-scale outdoor production of microalgae and cyanobacteria. Bioresour Technol. 2014;166:273–81.

    Article  PubMed  CAS  Google Scholar 

  165. Chen C-Y, Nagarajan D, Cheah WY. Eicosapentaenoic acid production from Nannochloropsis oceanica CY2 using deep sea water in outdoor plastic-bag type photobioreactors. Bioresour Technol. 2018;253:1–7.

    Article  PubMed  CAS  Google Scholar 

  166. Yang Z-Y, Gao F, Liu J-Z, Yang J-S, Liu M, Ge Y-M, Chen D-Z, Chen J-M. Improving sedimentation and lipid production of microalgae in the photobioreactor using saline wastewater. Bioresour Technol. 2022;347: 126392.

    Article  PubMed  CAS  Google Scholar 

  167. Yu Z, Hou Q, Liu M, Xie Z, Ma M, Chen H, Pei H. From lab to application: Cultivating limnetic microalgae in seawater coupled with wastewater for biodiesel production on a pilot scale. Water Res. 2023;229: 119471.

    Article  PubMed  CAS  Google Scholar 

  168. Sheng ALK, Bilad MR, Osman NB, Arahman N. Sequencing batch membrane photobioreactor for real secondary effluent polishing using native microalgae: process performance and full-scale projection. J Clean Prod. 2017;168:708–15.

    Article  CAS  Google Scholar 

  169. Kim ZH, Park H, Hong S-J, Lim S-M, Lee C-G. Development of a floating photobioreactor with internal partitions for efficient utilization of ocean wave into improved mass transfer and algal culture mixing. Bioprocess Biosyst Eng. 2016;39(5):713–23.

    Article  PubMed  CAS  Google Scholar 

  170. Chang H, Quan X, Zhong N, Zhang Z, Lu C, Li G, Cheng Z, Yang L. High-efficiency nutrients reclamation from landfill leachate by microalgae Chlorella vulgaris in membrane photobioreactor for bio-lipid production. Bioresour Technol. 2018;266:374–81.

    Article  PubMed  CAS  Google Scholar 

  171. Chang H, Fu Q, Zhong N, Yang X, Quan X, Li S, Fu J, Xiao C. Microalgal lipids production and nutrients recovery from landfill leachate using membrane photobioreactor. Bioresour Technol. 2019;277:18–26.

    Article  PubMed  CAS  Google Scholar 

  172. Yu Z, Pei H, Li Y, Yang Z, Xie Z, Hou Q, Nie C. Inclined algal biofilm photobioreactor (IABPBR) for cost-effective cultivation of lipid-rich microalgae and treatment of seawater-diluted anaerobically digested effluent from kitchen waste with the aid of phytohormones. Bioresour Technol. 2020;315: 123761.

    Article  PubMed  CAS  Google Scholar 

  173. Wang L, Zhao R, Wang Q, Han Z, Mao X-Z. Novel bioreactor with inclined baffles in cost-efficiently increasing algal biomass and carbon fixation. Energy. 2022;247:123453.

    Article  CAS  Google Scholar 

  174. Hassanpour M, Abbasabadi M, Ebrahimi S, Hosseini M, Sheikhbaglou A. Gravimetric enrichment of high lipid and starch accumulating microalgae. Bioresour Technol. 2015;196:17–21.

    Article  PubMed  CAS  Google Scholar 

  175. Gao X, Kong B, Vigil RD. Multiphysics simulation of algal growth in an airlift photobioreactor: effects of fluid mixing and shear stress. Bioresour Technol. 2018;251:75–83.

    Article  PubMed  CAS  Google Scholar 

  176. Banerjee S, Dasgupta S, Das D, Atta A. Influence of photobioreactor configuration on microalgal biomass production. Bioprocess Biosyst Eng. 2020;43(8):1487–97.

    Article  PubMed  CAS  Google Scholar 

  177. Huang J, Li Y, Wan M, Yan Y, Feng F, Qu X, Wang J, Shen G, Li W, Fan J, et al. Novel flat-plate photobioreactors for microalgae cultivation with special mixers to promote mixing along the light gradient. Bioresour Technol. 2014;159:8–16.

    Article  PubMed  CAS  Google Scholar 

  178. Lim YA, Ilankoon IMSK, Chong MN, Foo SC. Improving microalgae growth and carbon capture through micro-size bubbles generation in flat-panel photobioreactors: impacts of different gas sparger designs on mixing performance. Renew Sustain Energy Rev. 2023;171: 113001.

    Article  CAS  Google Scholar 

  179. Guo X, Yao L, Huang Q. Aeration and mass transfer optimization in a rectangular airlift loop photobioreactor for the production of microalgae. Bioresour Technol. 2015;190:189–95.

    Article  PubMed  CAS  Google Scholar 

  180. Pham H-M, Kwak HS, Hong M-E, Lee J, Chang WS, Sim SJ. Development of an X-Shape airlift photobioreactor for increasing algal biomass and biodiesel production. Bioresour Technol. 2017;239:211–8.

    Article  PubMed  CAS  Google Scholar 

  181. Vo HNP, Ngo HH, Guo W, Nguyen TMH, Liu Y, Liu Y, Nguyen DD, Chang SW. A critical review on designs and applications of microalgae-based photobioreactors for pollutants treatment. Sci Total Environ. 2019;651:1549–68.

    Article  PubMed  CAS  Google Scholar 

  182. Benvenuti G, Bosma R, Klok AJ, Ji F, Lamers PP, Barbosa MJ, Wijffels RH. Microalgal triacylglycerides production in outdoor batch-operated tubular PBRs. Biotechnol Biofuels. 2015;8(1):100.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Jagadevan S, Banerjee A, Banerjee C, Guria C, Tiwari R, Baweja M, Shukla P. Recent developments in synthetic biology and metabolic engineering in microalgae towards biofuel production. Biotechnol Biofuels. 2018;11(1):185.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Fayyaz M, Chew KW, Show PL, Ling TC, Ng IS, Chang J-S. Genetic engineering of microalgae for enhanced biorefinery capabilities. Biotechnol Adv. 2020;43: 107554.

    Article  PubMed  CAS  Google Scholar 

  185. Shin YS, Jeong J, Nguyen THT, Kim JYH, Jin E, Sim SJ. Targeted knockout of phospholipase A2 to increase lipid productivity in Chlamydomonas reinhardtii for biodiesel production. Bioresour Technol. 2019;271:368–74.

    Article  PubMed  CAS  Google Scholar 

  186. Chen J-W, Liu W-J, Hu D-X, Wang X, Balamurugan S, Alimujiang A, Yang W-D, Liu J-S, Li H-Y. Identification of a malonyl CoA-acyl carrier protein transacylase and its regulatory role in fatty acid biosynthesis in oleaginous microalga Nannochloropsis oceanica. Biotechnol Appl Biochem. 2017;64(5):620–6.

    Article  PubMed  CAS  Google Scholar 

  187. Fukuda S, Hirasawa E, Takemura T, Takahashi S, Chokshi K, Pancha I, Tanaka K, Imamura S. Accelerated triacylglycerol production without growth inhibition by overexpression of a glycerol-3-phosphate acyltransferase in the unicellular red alga Cyanidioschyzon merolae. Sci Rep. 2018;8(1):12410.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Mund NK, Liu Y, Chen S. Advances in metabolic engineering of cyanobacteria for production of biofuels. Fuel. 2022;322: 124117.

    Article  CAS  Google Scholar 

  189. Afrin S, Khan MRI, Zhang W, Wang Y, Zhang W, He L, Ma G. Membrane-located expression of thioesterase from acinetobacter baylyi enhances free fatty acid production with decreased toxicity in Synechocystis sp. PCC6803. Front Microbiol. 2018. https://doi.org/10.3389/fmicb.2018.02842.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Yunus IS, Wichmann J, Wördenweber R, Lauersen KJ, Kruse O, Jones PR. Synthetic metabolic pathways for photobiological conversion of CO2 into hydrocarbon fuel. Metab Eng. 2018;49:201–11.

    Article  PubMed  CAS  Google Scholar 

  191. Kato A, Takatani N, Ikeda K, Maeda S-I, Omata T. Removal of the product from the culture medium strongly enhances free fatty acid production by genetically engineered Synechococcus elongatus. Biotechnol Biofuels. 2017;10(1):141.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Ruffing AM. Improved free fatty acid production in cyanobacteria with Synechococcus sp. PCC 7002 as Host. Front Bioeng Biotechnol. 2014. https://doi.org/10.3389/fbioe.2014.00017.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Jia B, Xie X, Wu M, Lin Z, Yin J, Lou S, Huang Y, Hu Z. Understanding the functions of endogenous DOF transcript factor in Chlamydomonas reinhardtii. Biotechnol Biofuels. 2019;12(1):67.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Salas-Montantes CJ, González-Ortega O, Ochoa-Alfaro AE, Camarena-Rangel R, Paz-Maldonado LMT, Rosales-Mendoza S, Rocha-Uribe A, Soria-Guerra RE. Lipid accumulation during nitrogen and sulfur starvation in Chlamydomonas reinhardtii overexpressing a transcription factor. J Appl Phycol. 2018;30(3):1721–33.

    Article  CAS  Google Scholar 

  195. Bajhaiya AK, Dean AP, Zeef LAH, Webster RE, Pittman JK. PSR1 is a global transcriptional regulator of phosphorus deficiency responses and carbon storage metabolism in Chlamydomonas reinhardtii. Plant Physiol. 2015;170(3):1216–34.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Ngan CY, Wong C-H, Choi C, Yoshinaga Y, Louie K, Jia J, Chen C, Bowen B, Cheng H, Leonelli L, et al. Lineage-specific chromatin signatures reveal a regulator of lipid metabolism in microalgae. Nature Plants. 2015;1(8):15107.

    Article  PubMed  CAS  Google Scholar 

  197. Kao P-H, Ng IS. CRISPRi mediated phosphoenolpyruvate carboxylase regulation to enhance the production of lipid in Chlamydomonas reinhardtii. Bioresour Technol. 2017;245:1527–37.

    Article  PubMed  CAS  Google Scholar 

  198. Tan KWM, Lee YK. Expression of the heterologous Dunaliella tertiolecta fatty acyl-ACP thioesterase leads to increased lipid production in Chlamydomonas reinhardtii. J Biotechnol. 2017;247:60–7.

    Article  PubMed  CAS  Google Scholar 

  199. Rengel R, Smith RT, Haslam RP, Sayanova O, Vila M, León R. Overexpression of acetyl-CoA synthetase (ACS) enhances the biosynthesis of neutral lipids and starch in the green microalga Chlamydomonas reinhardtii. Algal Res. 2018;31:183–93.

    Article  Google Scholar 

  200. Singh P, Kumari S, Guldhe A, Singh G, Bux F. ACCase and rbcL gene expression as a function of nutrient and metal stress for enhancing lipid productivity in Chlorella sorokiniana. Energy Convers Manage. 2017;148:809–19.

    Article  CAS  Google Scholar 

  201. Lin W-R, Lai Y-C, Sung P-K, Tan S-I, Chang C-H, Chen C-Y, Chang J-S, Ng IS. Enhancing carbon capture and lipid accumulation by genetic carbonic anhydrase in microalgae. J Taiwan Inst Chem Eng. 2018;93:131–41.

    Article  CAS  Google Scholar 

  202. Shi H, Chen H, Gu Z, Zhang H, Chen W, Chen YQ. Application of a delta-6 desaturase with α-linolenic acid preference on eicosapentaenoic acid production in Mortierella alpina. Microb Cell Fact. 2016;15(1):117.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Li D-W, Balamurugan S, Yang Y-F, Zheng J-W, Huang D, Zou L-G, Yang W-D, Liu J-S, Guan Y, Li H-Y. Transcriptional regulation of microalgae for concurrent lipid overproduction and secretion. Sci Adv. 2019;5(1):3795.

    Article  Google Scholar 

  204. Poliner E, Pulman JA, Zienkiewicz K, Childs K, Benning C, Farré EM. A toolkit for Nannochloropsis oceanica CCMP1779 enables gene stacking and genetic engineering of the eicosapentaenoic acid pathway for enhanced long-chain polyunsaturated fatty acid production. Plant Biotechnol J. 2018;16(1):298–309.

    Article  PubMed  CAS  Google Scholar 

  205. Zienkiewicz K, Zienkiewicz A, Poliner E, Du Z-Y, Vollheyde K, Herrfurth C, Marmon S, Farré EM, Feussner I, Benning C. Nannochloropsis, a rich source of diacylglycerol acyltransferases for engineering of triacylglycerol content in different hosts. Biotechnol Biofuels. 2017;10(1):8.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Wei H, Shi Y, Ma X, Pan Y, Hu H, Li Y, Luo M, Gerken H, Liu J. A type-I diacylglycerol acyltransferase modulates triacylglycerol biosynthesis and fatty acid composition in the oleaginous microalga, Nannochloropsis oceanica. Biotechnol Biofuels. 2017;10(1):174.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Li D-W, Cen S-Y, Liu Y-H, Balamurugan S, Zheng X-Y, Alimujiang A, Yang W-D, Liu J-S, Li H-Y. A type 2 diacylglycerol acyltransferase accelerates the triacylglycerol biosynthesis in heterokont oleaginous microalga Nannochloropsis oceanica. J Biotechnol. 2016;229:65–71.

    Article  PubMed  CAS  Google Scholar 

  208. Xin Y, Lu Y, Lee Y-Y, Wei L, Jia J, Wang Q, Wang D, Bai F, Hu H, Hu Q, et al. Producing designer oils in industrial microalgae by rational modulation of co-evolving type-2 diacylglycerol acyltransferases. Mol Plant. 2017;10(12):1523–39.

    Article  PubMed  CAS  Google Scholar 

  209. Kwon S, Kang NK, Koh HG, Shin S-E, Lee B, Jeong B-R, Chang YK. Enhancement of biomass and lipid productivity by overexpression of a bZIP transcription factor in Nannochloropsis salina. Biotechnol Bioeng. 2018;115(2):331–40.

    Article  PubMed  CAS  Google Scholar 

  210. Kang NK, Kim EK, Kim YU, Lee B, Jeong W-J, Jeong B-R, Chang YK. Increased lipid production by heterologous expression of AtWRI1 transcription factor in Nannochloropsis salina. Biotechnol Biofuels. 2017;10(1):231.

    Article  PubMed  PubMed Central  Google Scholar 

  211. Kang NK, Jeon S, Kwon S, Koh HG, Shin S-E, Lee B, Choi G-G, Yang J-W, Jeong B-R, Chang YK. Effects of overexpression of a bHLH transcription factor on biomass and lipid production in Nannochloropsis salina. Biotechnol Biofuels. 2015;8(1):200.

    Article  PubMed  PubMed Central  Google Scholar 

  212. 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(1):318.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. 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.

    Article  PubMed  Google Scholar 

  214. Xue J, Balamurugan S, Li D-W, Liu Y-H, Zeng H, Wang L, Yang W-D, Liu J-S, Li H-Y. Glucose-6-phosphate dehydrogenase as a target for highly efficient fatty acid biosynthesis in microalgae by enhancing NADPH supply. Metab Eng. 2017;41:212–21.

    Article  PubMed  CAS  Google Scholar 

  215. Chen C-Y, Kao A-L, Tsai Z-C, Chow T-J, Chang H-Y, Zhao X-Q, Chen P-T, Su H-Y, Chang J-S. Expression of type 2 diacylglycerol acyltransferse gene DGTT1 from Chlamydomonas reinhardtii enhances lipid production in Scenedesmus obliquus. Biotechnol J. 2016;11(3):336–44.

    Article  PubMed  CAS  Google Scholar 

  216. Wang F, Bi Y, Diao J, Lv M, Cui J, Chen L, Zhang W. Metabolic engineering to enhance biosynthesis of both docosahexaenoic acid and odd-chain fatty acids in Schizochytrium sp. S31. Biotechnol Biofuels. 2019;12(1):141.

    Article  PubMed  PubMed Central  Google Scholar 

  217. Li Z, Meng T, Ling X, Li J, Zheng C, Shi Y, Chen Z, Li Z, Li Q, Lu Y, et al. Overexpression of Malonyl-CoA: ACP transacylase in Schizochytrium sp. to improve polyunsaturated fatty acid production. J Agri Food Chem. 2018;66(21):5382–91.

    Article  CAS  Google Scholar 

  218. Mishra A, Medhi K, Malaviya P, Thakur IS. Omics approaches for microalgal applications: prospects and challenges. Bioresour Technol. 2019;291: 121890.

    Article  PubMed  CAS  Google Scholar 

  219. Kaneko T, Sato S, Kotani H, Tanaka A, Asamizu E, Nakamura Y, Miyajima N, Hirosawa M, Sugiura M, Sasamoto S, et al. Sequence analysis of the genome of the unicellular cyanobacterium Synechocystis sp. strain PCC6803. II. Sequence determination of the entire genome and assignment of potential protein-coding regions. DNA Res. 1996;3(3):109–36.

    Article  PubMed  CAS  Google Scholar 

  220. Fu W, Nelson DR, Mystikou A, Daakour S, Salehi-Ashtiani K. Advances in microalgal research and engineering development. Curr Opin Biotechnol. 2019;59:157–64.

    Article  PubMed  CAS  Google Scholar 

  221. Blaby-Haas CE, Merchant SS. Comparative and functional algal genomics. Annu Rev Plant Biol. 2019;70(1):605–38.

    Article  PubMed  CAS  Google Scholar 

  222. Guarnieri MT, Pienkos PT. Algal omics: unlocking bioproduct diversity in algae cell factories. Photosynth Res. 2015;123(3):255–63.

    Article  PubMed  CAS  Google Scholar 

  223. Daboussi F, Leduc S, Maréchal A, Dubois G, Guyot V, Perez-Michaut C, Amato A, Falciatore A, Juillerat A, Beurdeley M, et al. Genome engineering empowers the diatom Phaeodactylum tricornutum for biotechnology. Nat Commun. 2014;5(1):3831.

    Article  PubMed  CAS  Google Scholar 

  224. Cheng S, Melkonian M, Smith SA, Brockington S, Archibald JM, Delaux P-M, Li F-W, Melkonian B, Mavrodiev EV, Sun W, et al. 10KP: a phylodiverse genome sequencing plan. GigaScience. 2018. https://doi.org/10.1093/gigascience/giy013.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Vandepoele K, Van Bel M, Richard G, Van Landeghem S, Verhelst B, Moreau H, Van de Peer Y, Grimsley N, Piganeau G. pico-PLAZA, a genome database of microbial photosynthetic eukaryotes. Environ Microbiol. 2013;15(8):2147–53.

    Article  PubMed  CAS  Google Scholar 

  226. Zheng H-Q, Chiang-Hsieh Y-F, Chien C-H, Hsu B-KJ, Liu T-L, Chen C-NN, Chang W-C. AlgaePath: comprehensive analysis of metabolic pathways using transcript abundance data from next-generation sequencing in green algae. BMC Genomics. 2014;15:196.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Aoki Y, Okamura Y, Ohta H, Kinoshita K, Obayashi T. ALCOdb: gene coexpression database for microalgae. Plant Cell Physiol. 2016;57(1): e3.

    Article  PubMed  Google Scholar 

  228. Khatodia S, Bhatotia K, Passricha N, Khurana SMP, Tuteja N. The CRISPR/Cas genome-editing tool: application in improvement of crops. Front Plant Sci. 2016. https://doi.org/10.3389/fpls.2016.00506.

    Article  PubMed  PubMed Central  Google Scholar 

  229. Keeling PJ, Burki F, Wilcox HM, Allam B, Allen EE, Amaral-Zettler LA, Armbrust EV, Archibald JM, Bharti AK, Bell CJ, et al. The Marine Microbial Eukaryote Transcriptome Sequencing Project (MMETSP): illuminating the functional diversity of eukaryotic life in the oceans through transcriptome sequencing. PLoS Biol. 2014;12(6): e1001889.

    Article  PubMed  PubMed Central  Google Scholar 

  230. Van Vlierberghe M, Di Franco A, Philippe H, Baurain D. Decontamination, pooling and dereplication of the 678 samples of the Marine Microbial Eukaryote Transcriptome Sequencing Project. BMC Res Notes. 2021;14(1):306.

    Article  PubMed  PubMed Central  Google Scholar 

  231. Yang F, Xiang W, Li T, Long L. Transcriptome analysis for phosphorus starvation-induced lipid accumulation in Scenedesmus sp. Sci Rep. 2018;8(1):16420.

    Article  PubMed  PubMed Central  Google Scholar 

  232. Cecchin M, Marcolungo L, Rossato M, Girolomoni L, Cosentino E, Cuine S, Li-Beisson Y, Delledonne M, Ballottari M. Chlorella vulgaris genome assembly and annotation reveals the molecular basis for metabolic acclimation to high light conditions. Plant J. 2019;100(6):1289–305.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  233. Bai F, Yu L, Shi J, Li-Beisson Y, Liu J. Long-chain acyl-CoA synthetases activate fatty acids for lipid synthesis, remodeling and energy production in Chlamydomonas. New Phytol. 2022;233(2):823–37.

    Article  PubMed  CAS  Google Scholar 

  234. Wu D, Hou Y, Cheng J, Han T, Hao N, Zhang B, Fan X, Ji X, Chen F, Gong D, et al. Transcriptome analysis of lipid metabolism in response to cerium stress in the oleaginous microalga Nannochloropsis oculata. Sci Total Environ. 2022;838: 156420.

    Article  PubMed  CAS  Google Scholar 

  235. Anand V, Kashyap M, Samadhiya K, Kiran B. Strategies to unlock lipid production improvement in algae. Int J Environ Sci Technol. 2019;16(3):1829–38.

    Article  Google Scholar 

  236. Tran N-AT, Padula MP, Evenhuis CR, Commault AS, Ralph PJ, Tamburic B. Proteomic and biophysical analyses reveal a metabolic shift in nitrogen deprived Nannochloropsis oculata. Algal Res. 2016;19:1–11.

    Article  Google Scholar 

  237. Nonoyama T, Nojima D, Maeda Y, Noda M, Yoshino T, Matsumoto M, Bowler C, Tanaka T. Proteomics analysis of lipid droplets indicates involvement of membrane trafficking proteins in lipid droplet breakdown in the oleaginous diatom Fistulifera solaris. Algal Res. 2019;44: 101660.

    Article  Google Scholar 

  238. Karthikaichamy A, Beardall J, Coppel R, Noronha S, Bulach D, Schittenhelm RB, Srivastava S. Data-independent-acquisition-based proteomic approach towards understanding the acclimation strategy of oleaginous microalga Microchloropsis gaditana CCMP526 in hypersaline conditions. ACS Omega. 2021;6(34):22151–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  239. Yoneda K, Ishibashi Y, Yoshida M, Watanabe MM, Ito M, Suzuki I. Proteomic and lipidomic analyses of lipid droplets in Aurantiochytrium limacinum ATCC MYA-1381. Algal Res. 2022;67: 102844.

    Article  Google Scholar 

  240. Hulatt CJ, Smolina I, Dowle A, Kopp M, Vasanth GK, Hoarau GG, Wijffels RH, Kiron V. Proteomic and transcriptomic patterns during lipid remodeling in Nannochloropsis gaditana. Int J Mol Sci. 2020;21(18):6946.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  241. Xing G, Yuan H, Yang J, Li J, Gao Q, Li W, Wang E. Integrated analyses of transcriptome, proteome and fatty acid profilings of the oleaginous microalga Auxenochlorella protothecoides UTEX 2341 reveal differential reprogramming of fatty acid metabolism in response to low and high temperatures. Algal Res. 2018;33:16–27.

    Article  Google Scholar 

  242. Wu M, Zhang H, Sun W, Li Y, Hu Q, Zhou H, Han D. Metabolic plasticity of the starchless mutant of Chlorella sorokiniana and mechanisms underlying its enhanced lipid production revealed by comparative metabolomics analysis. Algal Res. 2019;42: 101587.

    Article  Google Scholar 

  243. Chu H, Ren L, Yang L, Chen J, Zhou X, Zhang Y. Metabolomics reveals a lipid accumulation mechanism involving carbon allocation in Scenedesmus obliquus under norfloxacin stress. Renewable Energy. 2020;157:585–92.

    Article  CAS  Google Scholar 

  244. Zhao Y, Wang H-P, Yu C, Ding W, Han B, Geng S, Ning D, Ma T, Yu X. Integration of physiological and metabolomic profiles to elucidate the regulatory mechanisms underlying the stimulatory effect of melatonin on astaxanthin and lipids coproduction in Haematococcus pluvialis under inductive stress conditions. Bioresour Technol. 2021;319: 124150.

    Article  PubMed  CAS  Google Scholar 

  245. Trovão M, Schüler LM, Machado A, Bombo G, Navalho S, Barros A, Pereira H, Silva J, Freitas F, Varela J. Random mutagenesis as a promising tool for microalgal strain improvement towards industrial production. Mar Drugs. 2022;20(7):440.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Cecchin M, Berteotti S, Paltrinieri S, Vigliante I, Iadarola B, Giovannone B, Maffei ME, Delledonne M, Ballottari M. Improved lipid productivity in Nannochloropsis gaditana in nitrogen-replete conditions by selection of pale green mutants. Biotechnol Biofuels. 2020;13(1):78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  247. Cecchin M, Cazzaniga S, Martini F, Paltrinieri S, Bossi S, Maffei ME, Ballottari M. Astaxanthin and eicosapentaenoic acid production by S4, a new mutant strain of Nannochloropsis gaditana. Microb Cell Fact. 2022;21(1):117.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  248. Wan Razali WA, Evans CA, Pandhal J. Comparative proteomics reveals evidence of enhanced EPA trafficking in a mutant strain of Nannochloropsis oculata. Front Bioeng Biotechnol. 2022. https://doi.org/10.3389/fbioe.2022.838445.

    Article  PubMed  PubMed Central  Google Scholar 

  249. Moha-León JD, Pérez-Legaspi IA, Ortega-Clemente LA, Rubio-Franchini I, Ríos-Leal E. Improving the lipid content of Nannochloropsis oculata by a mutation-selection program using UV radiation and quizalofop. J Appl Phycol. 2019;31(1):191–9.

    Article  Google Scholar 

  250. Ma C, Ren H, Xing D, Xie G, Ren N, Liu B. Mechanistic understanding towards the effective lipid production of a microalgal mutant strain Scenedesmus sp. Z-4 by the whole genome bioinformation. J Hazard Mater. 2019;375:115–20.

    Article  PubMed  CAS  Google Scholar 

  251. Liu B, Ma C, Xiao R, Xing D, Ren H, Ren N. The screening of microalgae mutant strain Scenedesmus sp. Z-4 with a rich lipid content obtained by 60Co γ-ray mutation. RSC Adv. 2015;5(64):52057–61.

    Article  CAS  Google Scholar 

  252. Jakhwal P, Kumar Biswas J, Tiwari A, Kwon EE, Bhatnagar A. Genetic and non-genetic tailoring of microalgae for the enhanced production of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)–A review. Bioresour Technol. 2022;344: 126250.

    Article  PubMed  CAS  Google Scholar 

  253. Le TG, Tran D-T, Van Do TC, Nguyen VT. Design Considerations of Microalgal Culture Ponds and Photobioreactors for Wastewater Treatment and Biomass Cogeneration. In: Alam MA, Wang Z, editors. Microalgae Biotechnology for Development of Biofuel and Wastewater Treatment. Singapore: Springer Singapore; 2019. p. 535–67.

    Chapter  Google Scholar 

  254. Ryu AJ, Kang NK, Jeon S, Hur DH, Lee EM, Lee DY, Jeong B-R, Chang YK, Jeong KJ. Development and characterization of a Nannochloropsis mutant with simultaneously enhanced growth and lipid production. Biotechnol Biofuels. 2020;13(1):46.

    Article  CAS  Google Scholar 

  255. Südfeld C, Hubáček M, Figueiredo D, Naduthodi MIS, van der Oost J, Wijffels RH, Barbosa MJ, D’Adamo S. High-throughput insertional mutagenesis reveals novel targets for enhancing lipid accumulation in Nannochloropsis oceanica. Metab Eng. 2021;66:239–58.

    Article  PubMed  Google Scholar 

  256. Cheng X, Liu G, Ke W, Zhao L, Lv B, Ma X, Xu N, Xia X, Deng X, Zheng C, et al. Building a multipurpose insertional mutant library for forward and reverse genetics in Chlamydomonas. Plant Methods. 2017;13(1):36.

    Article  PubMed  PubMed Central  Google Scholar 

  257. Park S, Nguyen THT, Jin E. Improving lipid production by strain development in microalgae: strategies, challenges and perspectives. Bioresour Technol. 2019;292: 121953.

    Article  PubMed  CAS  Google Scholar 

  258. Adeniyi OM, Azimov U, Burluka A. Algae biofuel: current status and future applications. Renew Sustain Energy Rev. 2018;90:316–35.

    Article  Google Scholar 

  259. Kindle KL. High-frequency nuclear transformation of Chlamydomonas reinhardtii. Proc Natl Acad Sci. 1990;87(3):1228–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  260. Rismani-Yazdi H, Haznedaroglu BZ, 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 Genomics. 2011;12(1):148.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  261. Zhu B-H, Zhang R-H, Lv N-N, Yang G-P, Wang Y-S, Pan K-H. The role of malic enzyme on promoting total lipid and fatty acid production in Phaeodactylum tricornutum. Front Plant Sci. 2018. https://doi.org/10.3389/fpls.2018.00826.

    Article  PubMed  PubMed Central  Google Scholar 

  262. Xin Y, Shen C, She Y, Chen H, Wang C, Wei L, Yoon K, Han D, Hu Q, Xu J. Biosynthesis of triacylglycerol molecules with a tailored PUFA profile in industrial microalgae. Mol Plant. 2019;12(4):474–88.

    Article  PubMed  CAS  Google Scholar 

  263. Levitan O, Dinamarca J, Zelzion E, Lun DS, Guerra LT, Kim MK, Kim J, Van Mooy BAS, Bhattacharya D, Falkowski PG. Remodeling of intermediate metabolism in the diatom Phaeodactylum tricornutum under nitrogen stress. Proc Natl Acad Sci. 2015;112(2):412–7.

    Article  PubMed  CAS  Google Scholar 

  264. Trentacoste EM, Shrestha RP, Smith SR, Glé C, Hartmann AC, Hildebrand M, Gerwick WH. Metabolic engineering of lipid catabolism increases microalgal lipid accumulation without compromising growth. Proc Natl Acad Sci. 2013;110(49):19748–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  265. 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. https://doi.org/10.3389/fpls.2021.650820.

    Article  PubMed  PubMed Central  Google Scholar 

  266. Sizova I, Greiner A, Awasthi M, Kateriya S, Hegemann P. Nuclear gene targeting in Chlamydomonas using engineered zinc-finger nucleases. Plant J. 2013;73(5):873–82.

    Article  PubMed  CAS  Google Scholar 

  267. Greiner A, Kelterborn S, Evers H, Kreimer G, Sizova I, Hegemann P. Targeting of photoreceptor genes in Chlamydomonas reinhardtii via zinc-finger nucleases and CRISPR/Cas9. Plant Cell. 2017;29(10):2498–518.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  268. 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(1):312.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  269. Abdallah NA, Prakash CS, McHughen AG. Genome editing for crop improvement: challenges and opportunities. GM Crops & Food. 2015;6(4):183–205.

    Article  Google Scholar 

  270. Boettcher M, McManus MT. Choosing the Right Tool for the Job: RNAi, TALEN, or CRISPR. Mol Cell. 2015;58(4):575–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  271. Thurtle-Schmidt DM, Lo T-W. Molecular biology at the cutting edge: a review on CRISPR/CAS9 gene editing for undergraduates. Biochem Mol Biol Educ. 2018;46(2):195–205.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  272. Jeon S, Lim J-M, Lee H-G, Shin S-E, Kang NK, Park Y-I, Oh H-M, Jeong W-J, Jeong B-R, Chang YK. Current status and perspectives of genome editing technology for microalgae. Biotechnol Biofuels. 2017;10(1):267.

    Article  PubMed  PubMed Central  Google Scholar 

  273. Wang Q, Lu Y, Xin Y, Wei L, Huang S, Xu J. Genome editing of model oleaginous microalgae Nannochloropsis spp. by CRISPR/Cas9. Plant J. 2016;88(6):1071–81.

    Article  PubMed  CAS  Google Scholar 

  274. Ajjawi I, Verruto J, Aqui M, Soriaga LB, Coppersmith J, Kwok K, Peach L, Orchard E, Kalb R, Xu W, et al. Lipid production in Nannochloropsis gaditana is doubled by decreasing expression of a single transcriptional regulator. Nat Biotechnol. 2017;35(7):647–52.

    Article  PubMed  CAS  Google Scholar 

  275. Chang KS, Kim J, Park H, Hong S-J, Lee C-G, Jin E. Enhanced lipid productivity in AGP knockout marine microalga Tetraselmis sp. using a DNA-free CRISPR-Cas9 RNP method. Bioresour Technol. 2020;303:122932.

    Article  PubMed  CAS  Google Scholar 

  276. Hao X, Chen W, Amato A, Jouhet J, Maréchal E, Moog D, Hu H, Jin H, You L, Huang F, et al. Multiplexed CRISPR/Cas9 editing of the long-chain acyl-CoA synthetase family in the diatom Phaeodactylum tricornutum reveals that mitochondrial ptACSL3 is involved in the synthesis of storage lipids. New Phytol. 2022;233(4):1797–812.

    Article  PubMed  CAS  Google Scholar 

  277. Beacham TA, Sweet JB, Allen MJ. Large scale cultivation of genetically modified microalgae: A new era for environmental risk assessment. Algal Res. 2017;25:90–100.

    Article  Google Scholar 

  278. Campbell ML. Assessing biosecurity risk associated with the importation of non-indigenous microalgae. Environ Res. 2011;111(7):989–98.

    Article  PubMed  CAS  Google Scholar 

  279. Tabatabaei M, Tohidfar M, Jouzani GS, Safarnejad M, Pazouki M. Biodiesel production from genetically engineered microalgae: future of bioenergy in Iran. Renew Sustain Energy Rev. 2011;15(4):1918–27.

    Article  CAS  Google Scholar 

  280. Nethravathy MU, Jitendra GM, Sandeep NM, Shekh AY. Recent advances in microalgal bioactives for food, feed, and healthcare products: commercial potential, market space, and sustainability. Comprehensive Rev Food Sci Food Safety. 2019;18(6):1882–97.

    Article  Google Scholar 

  281. Society CER. China Energy Outlook 2030. Beijing, China: Energy and Management Press; 2016.

    Google Scholar 

  282. Sun M-T, Fan X-L, Zhao X-X, Fu S-F, He S, Manasa MRK, Guo R-B. Effects of organic loading rate on biogas production from macroalgae: performance and microbial community structure. Bioresour Technol. 2017;235:292–300.

    Article  PubMed  CAS  Google Scholar 

  283. He S, Fan X, Luo S, Katukuri NR, Guo R. Enhanced the energy outcomes from microalgal biomass by the novel biopretreatment. Energy Convers Manage. 2017;135:291–6.

    Article  CAS  Google Scholar 

  284. Rastogi RP, Pandey A, Larroche C, Madamwar D. Algal green energy – R&D and technological perspectives for biodiesel production. Renew Sustain Energy Rev. 2018;82:2946–69.

    Article  CAS  Google Scholar 

  285. Chong JWR, Tang DYY, Leong HY, Khoo KS, Show PL, Chew KW. Bridging artificial intelligence and fucoxanthin for the recovery and quantification from microalgae. Bioengineered. 2023;14(1):2244232.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This research was funded by Scientific Research Funds for Heilongjiang Provincial Institutes (Grant number: CZKYF2022-2-C001), Design and Breeding of New Varieties of Cold-resistant and high-yield alfalfa in Beidahuang Reclamation Area (Grant number: 2022ZD040120403), and Heilongjiang Academy of Agricultural Sciences Postdoctoral Programme.

Author information

Authors and Affiliations

Authors

Contributions

MW wrote the main manuscript text. XY prepared figures. HB and ZS provided conceptualization and supervision. HB improved language. MW and ZS provide Funding. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Min Wang or Zhongbao Shen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1

. Fatty acids composition of green algae (percentage of total fatty acids).

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, M., Ye, X., Bi, H. et al. Microalgae biofuels: illuminating the path to a sustainable future amidst challenges and opportunities. Biotechnol Biofuels 17, 10 (2024). https://doi.org/10.1186/s13068-024-02461-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13068-024-02461-0

Keywords