Skip to main content

Biosynthesis pathways of expanding carbon chains for producing advanced biofuels

Abstract

Because the thermodynamic property is closer to gasoline, advanced biofuels (C ≥ 6) are appealing for replacing non-renewable fossil fuels using biosynthesis method that has presented a promising approach. Synthesizing advanced biofuels (C ≥ 6), in general, requires the expansion of carbon chains from three carbon atoms to more than six carbon atoms. Despite some specific biosynthesis pathways that have been developed in recent years, adequate summary is still lacking on how to obtain an effective metabolic pathway. Review of biosynthesis pathways for expanding carbon chains will be conducive to selecting, optimizing and discovering novel synthetic route to obtain new advanced biofuels. Herein, we first highlighted challenges on expanding carbon chains, followed by presentation of two biosynthesis strategies and review of three different types of biosynthesis pathways of carbon chain expansion for synthesizing advanced biofuels. Finally, we provided an outlook for the introduction of gene-editing technology in the development of new biosynthesis pathways of carbon chain expansion.

Introduction

Fossil fuels such as gasoline, have increasingly incited a string of ecological disaster, environmental pollution, and looming international policy issues [1]. Developing advanced biofuels to substitute for petrochemical production through biotransformation is regarded as a promising alternative [2]. The production of renewable biofuels through bioengineering has long been executed, converting single-carbon methane to longer biofuels closer to the octane value (C8H18) of gasoline (Fig. 1). Due to various remarkable defects related to its chemical properties, such as high hydroscopicity and low energy density, recent research has attempted to substitute some traditional bioenergy compounds, like ethanol and butanol, with other advanced biofuels that process long carbon chain (C ≥ 6 and ≤ 10) such as fatty alcohol compounds, olefins and alkanes compounds. In fact, recent findings have reported important progress in the expansion of carbon chains via using novel engineering strategy to obtain advanced biofuels with long carbon chain [3].

Fig. 1
figure 1

Development progress of bio-based small molecule compounds for replacing petroleum industry. With the development of bio-based small molecule compounds for substituting non-renewable gasoline, increasing length of carbon chain has been a hallmark of biosynthesis strategy. One of the major future developments of bio-based small molecule compounds for replacing fossil fuel is to exploit advanced biofuels with long carbon chains (C > 6) using appropriate engineering strategy

Some biosynthesis pathways have been proposed for synthesizing advanced biofuels with long carbon chains, such as leveraging the 2-ketonic acid metabolic pathway [4] and the fatty acid metabolic pathway [5]. In the last decade, studies on advanced biofuels with long carbon chains have attracted more attention, however, the problems, such as low efficiency, have kept these pathways from being adopted for industrialization. Biosynthesis pathways directly determine the choice of metabolic pathways of carbon chain expansion and further ascertain the specific key enzymes of the target product. Therefore, it is necessary to review existing biosynthesis pathways and construct new metabolic pathways.

In this paper, we first presented the low efficiency problem of expanding carbon chains and then put forth two biosynthesis strategies to overcome the problem. Based on these biosynthesis strategies, three different biosynthesis pathways were thoroughly reviewed according to classification. Furthermore, some methods including CRISPR/Cas9 gene-editing technology were introduced to improve the low efficiency, and to increase the speed of creating new biosynthesis pathways. This review will be conducive to providing valuable lessons for synthesizing advanced biofuels in future.

Low efficiency of expanding carbon chains

The production of diversified biofuels with short linear carbon chain (C < 5) is generally derived from a biosynthesis pathway that relies on the 2-keto acid pathway (Fig. 2), an intrinsic metabolic pathway in microorganism such as yeast, without requiring the expansion of carbon chain [6]. However, the number of carbon atoms derived from this approach is limited to a maximum of 5 carbons for linear alcohol except for cyclic alcohols such as 2-phenylethanol [19]. Therefore, it is necessary to develop other biosynthesis pathways that would expand carbon chains (C ≥ 6) to obtain linear advanced biofuels that are closer to gasoline.

Fig. 2
figure 2

Engineering strategy for producing short linear alcohols (C2–C5) based on the natural characteristics of microorganisms. Generally, synthesizing bio-based short linear alcohols (C2–C5) is based on the Ehrlich pathway and α-keto acid pathway of the microorganism itself. This engineering strategy is only possible to reach a maximum of five carbons, which cannot meet the necessary for extending carbon chain in generating non-natural bio-based small molecule compounds (C > 6) for substituting non-renewable gasoline. GAPDH: glyceraldehyde 3-phosphate dehydrogenase. PEP phosphoenolpyruvate. KDC 2-keto acid decarboxylases. ADH alcohol dehydrogenase. BAT2 aminotransferase. Double arrow show the oxidation–reduction pathway to biofuel

The majority of native microorganisms isolated from their natural environment are inefficient in synthesizing advanced biofuels. To achieve production of advanced biofuels, exogenetic metabolic pathways need to be successfully incubated in native microorganisms, which usually requires creating an engineering strategy that can overexpress multi-enzyme systems [7].

The process will inevitably produce low efficiency problem. Firstly, it involves more gene expression systems, more metabolic pathways, and more associated intermediates, which will increase the probability of failure, such as overproduction of unnecessary by-products or a low output of targeted products. Secondly, fine-tuned regulation of microbial physiology in the process of controlling the expression systems is complicated by the import of diversiform multi-enzyme combined system. Finally, technical difficulty in identifying a single engineering event from the entirety of the microbial genome also greatly increases the probability of failure. A good synthesis strategy can solve the above problems to some extent. Therefore, biosynthesis strategies are of great significance to guide effective metabolic pathways of biosynthesis. Only by involving sophisticated metabolic pathways can lay the foundation for solving low efficiency problem. So far, however, although a number of biosynthetic pathways have been developed for producing different advanced biofuels, how to choose a reasonable synthesis strategy is rarely summarized for improving the synthesis efficiency.

Biosynthesis strategies on advanced biofuels

Generally, obtaining biosynthetic pathways can be divided into three types according to the source: (1) use only the host's own metabolic pathway. For example, the metabolic pathway of Escherichia coli itself: the synthesis pathway of metabolites such as succinic acid, pyruvate, l-threonine and l-valine already exists in the natural E. coli metabolic pathway [4, 8]. The metabolic pathway of E. coli itself should be modified and regulated appropriately, such as releasing feedback inhibition of key sites, improving the expression of rate-limiting enzymes, regulating the metabolic balance using cofactors, etc., to maximize the metabolic flow to target products. (2) Introduction of foreign metabolic pathways: For example, E. coli has limited genes, so it is often necessary to use foreign genes to complete the metabolic pathway or improve the efficiency of the original metabolic pathway. For example, glycerol-3-phosphate dehydrogenase, glycerol-3-phosphatase from S. cerevisiae and glycerol-3-phosphatase from Klebsiella pneumoniae were introduced into the E. coli cell factory to produce 1, 3-propylene glycol [9, 10]. An alanine dehydrogenase derived from Geobacillus stearothermophilus was introduced to the cell factory to produce L-alanine. Five foreign genes from different sources were integrated into E. coli cell factory to produce 1,4-butanediol [11]. (3) Creating biosynthetic pathways that do not exist in nature microorganism. With the help of technologies such as rational protein modification and de novo design, the design of biosynthetic pathways has also broken through the limitations of natural pathways and gradually developed non-natural biosynthetic pathways.

However, biosynthesis strategies play a fundamental role in the initial exploration of novel biosynthetic pathways. Here, we summarized two biosynthesis strategies based on the above three types of biosynthetic pathways. To achieve advanced biofuels and overcome low efficiency problem of expanding carbon chains for the exploration of biosynthetic pathways, there are two basic biosynthesis strategies to be followed in the synthesis of advanced biofuels. As shown in Fig. 3, the first biosynthesis strategy is a retrospective method (RM) based on the molecular structure of the target product, which involves the reverse inference of substrates with similar structures (Fig. 3a). This strategy relies on reverse deduction and de novo analysis of the whole metabolic network of the target product, as well as the optimization of multi-step metabolic pathways. This strategy has great potential for obtaining new metabolic pathways, however, it requires the identification of multitudinous enzymes. For example, based on RM, through reversely deducing and analyzing analogous molecular structure of the target product 1,4-butanediol, a comparative study was conducted via analyzing potential metabolic pathways [12, 13]. An optimal engineering pathway in E. coli was designed using only four steps with Acetyl-CoA, α-ketoglutarate, glutamic acid, and succinyl Coenzyme A to achieve a maximum production of 18 g/L of 1,4-butanediol [13].

Fig. 3
figure 3

The two biosynthesis strategies of expanding carbon chain in exploring new biosynthesis pathway. RM retrospective method. ED evolutionary design method. n reaction steps

The second biosynthesis strategy is an evolutionary design (ED) approach based on known metabolic pathways or molecular structures of molecules similar to advanced biofuels (Fig. 3b). This strategy takes full advantage of the abundance of biological information in different databases, such as KEGG, NCBI and protein database, and extrapolates the appropriate extrinsic engineering pathway for synthesizing the target products. This strategy could identify the longest and shortest engineering pathway by tracking the metabolic flux of elements. Once a particular chemical structure similar to the target product among the intermediate metabolites, such as carbon metabolic intermediate was determined, the feasibilities of all potential engineering pathways will be validated according to the metabolic flux of the target product. For example, according to the molecular structure of branched-chain amino acids, such as leucine and isoleucine, the metabolic pathways were recombined to produce advanced biofuels hexanol [14, 15] and 3-methyl-butanol [16, 17]. This strategy provides an inspiration for discovering and designing novel biosynthetic pathways based on known metabolic pathways. In addition to using biological information, some prediction tools of synthetic pathways such as network-based pathway analysis tool could also be employed in this strategy, which could speed up the discovery of new biosynthetic pathways for synthesizing advanced biofuels.

Based on the above two biosynthetic strategies, the optimal metabolic pathway can be determined to solve the problem of low efficiency to a large extent in the process of extending the carbon chain, such as reducing the intermediate metabolic reaction as much as possible, and reducing the metabolic tributaries producing by-products as much as possible. Through the above strategies, obtaining the metabolic pathway with the fewest reaction steps can reduce the difficulty in the technical level of gene manipulation, for example, it is easier to knock out the by-product generation pathway, more conducive to release the inhibition of product synthesis, the stability of key enzymes in the overexpression synthesis pathway, the calculation of the minimum metabolic network model, the assembly of metabolic pathway, and the dynamic regulation of gene expression.

Biosynthetic pathways of advanced biofuels via expanding carbon chains

To date, different biosynthetic pathways of expanding carbon chains have been developed based on the above strategies using multi-enzyme expression system to acquire advanced biofuels including C6–C10 alcohols, C6–C18 alkanes, enolates, and C6–C10 olefins [18]. Here, we reviewed three important biosynthetic pathways of expanding carbon chains using 2-keto acid intermediates, fatty acid intermediates and reverse β-oxidation intermediates, and performed the analysis using concrete examples.

Production of advanced biofuels using 2-keto acid intermediates

Some natural precursors derived from different intermediates of 2-keto acids metabolic pathways are transformed into advanced biofuels with molecular structure similar to the intermediates based on the ED strategy (Table 1). In this study, the various kinds of 2-keto acids pathways were divided into three sub-pathways (Type I, II, III). Correspondingly, each of the sub-pathways was reviewed and was able to produce distinct advanced biofuels (Fig. 4).

Table 1 Advanced biofuels using 2-keto acid intermediates
Fig. 4
figure 4

Biosynthesis pathway of expanding carbon chain for producing advanced biofuels using 2-keto acid intermediates. This biosynthesis pathway can be subdivided into three distinct sub-pathways (I, II, III), each of which results in a corresponding and structurally distinct advanced biofuel. a The I sub-pathway is that the carbon chain is extended via adding two carbon atoms (+ 2) with pyruvate as carbon extension unit; the key enzymes are acetohydroxy acid synthase (AHAS) and IlvIHCD. b The II sub-pathway is that the carbon chain is lengthened via adding one carbon atom (+ 1) with CoA-dependent molecule as carbon extension unit. Advanced biofuels with a branched chain are obtained via integrating the following natural biosynthetic pathways: α-keto acid-based pathway and CoA-dependent extension pathway. c Carbon chain extension of the Type III sub-pathway also occurs involving the addition of one carbon atom (+ 1) using CoA-dependent molecule as the carbon extension unit. Advanced biofuels without branched chains were obtained via utilizing this subtype pathway and conducting a condensation reaction with the overexpression of leuABCD. Green arrows represent using pyruvate as carbon extension unit. Blue arrows represent using CoA-dependent molecule as carbon extension unit. Red arrows represent exogenous decarboxylation and reduction. ilvA threonine deaminase; ilvC acetohydroxy acid isomeroreductase; ilvD dihydroxy acid dehydratase; ilvGM acetohydroxybutanoate synthase; kivd ketoisovalerate decarboxylase; leuA, 2-isopropylmalate synthase; leuB 3-isopropylmalate dehydrogenase; leuCD 2-isopropylmalate isomerase; thrA aspartate kinase, homoserine dehydrogenase; thrB homoserine kinase; thrC threonine synthase; Ac-CoA acetylCoA; ACP acyl carrier protein; CoA coenzyme A; OAA oxaloacetate; ADH2 alcohol dehydrogenase; alsS acetolactate synthase; asd aspartate semialdehyde dehydrogenase; aspC aspartate aminotransferase; cimA citramalate synthase. KIC:2-ketoisocaproate, precursor to l-leucine; KMV 2-keto-3-methylvalerate, precursor of l-isoleucine; PaaH1 β-ketothiolase (BKtB)-3-hydroxy-acyl-CoA dehydrogenase; Crt crotonase; Ter trans-enoyl-CoA reductase; adhE alcohol dehydrogenase. The red arrow shows the oxidation–reduction pathway to higher biofuel. The green arrow is the carbon chain extension pathway

The biosynthetic pathways for Type I sub-pathway was featured by the addition of two carbon number (+ 2) using pyruvate molecules as the carbon chain extension factor. Various advanced biofuels were obtained using this metabolic pathway via the conversion of interlinked intermediates of the pathway (Fig. 4a). Carbon chains specifically, two pyruvate molecules were first concatenated, and then converted into a key intermediate 2-ketoisovalerate (KIV, precursor to l-valine), which was then catalyzed by the catalytic reactions of two catalyzing enzymes: IlvC, and IlvD [19, 20]. Further extension of the carbon chains could be achieved using this sub-pathway via a condensation reaction between KIV and pyruvate molecule. For example, 2-keto-3-methyl-valerate (KMV) was synthesized and further converted into 2-methyl-1-butanol by an extrinsic catalytic pathway, which involved an oxidation–reduction reaction responsible for generating decarboxylase and reductase [8, 21, 22].

The biosynthetic pathways for Type II was characterized by the extension of one carbon number (+ 1) using CoA-dependent molecule as the carbon extension factor. The advanced biofuels produced via this pathway were primarily alcohols with branched chains that were the result of comprehensive interlacement with other inherent pathways: a natural α-keto acid metabolic pathway and a metabolic pathway that shared characteristics of the Type I sub-pathway (Fig. 4b). It is worth noting that this metabolic pathway could produce various alcohols. Specifically, the intermediate KMV was further converted into 2-keto-4-methyl hexanoate (K4MH, precursor to 3-methyl-pentanol) via the overexpression of a series of enzymes such as LeuA, LeuB, and LeuCD [23], using acetyl-CoA as the carbon extension unit [24]. Similarly, other intermediates could be obtained from this metabolic pathway, including 2-keto-5-methyl heptanoate (K5MH, precursor to 4-methyl-hexanol) and 2-keto-6-methyl octanoate (KMO, precursor to 5-methyl-heptanol). These studies showed that carbon chain elongation could be implemented by leveraging the catalytic reaction of LeuABCD, and some substrates of advanced biofuels, such as KMV, K4MH, K5MH, and KMO, could be produced by the metabolic pathway. Subsequently, the production of some branched alcohols with a range of 6 to 10 carbon atoms could be achieved via oxidation–reduction reactions.

The biosynthetic pathways for Type III sub-pathway were also defined by the elongation of one carbon atom number (+ 1) using CoA-dependent molecule as carbon chain expanding factor. The similarity between this sub-pathway and the Type II sub-pathway is that they both leveraged acetyl-CoA as the carbon chain elongation factor, and resulted in a condensation reaction by overexpressing LeuABCD. A distinguishing feature between these two metabolic pathways could be found in the advanced biofuels (C ≥ 6) produced from the Type III sub-pathway as it did not contain any branched chains (Fig. 4c). The Type III sub-pathway could be divided into three steps: amino acid synthesis reaction, deammoniation reaction, and condensation reaction. For example, intermediates 2-ketovalerate (2 kV, precursor to norvaline biosynthesis) were first produced from 2-ketobutyrate (2 KB) via the overexpression of LeuABCD [25]. Then, a variety of precursors of advanced biofuels (C ≥ 6) could be obtained by converting 2 kV intermediates. Finally, 2-ketocaproate (2KC, precursor to n-pentanol) were synthesized via a condensation reaction between 2 kV and acetyl-CoA. Based on this strategy, other precursors could produce their corresponding alkane-based alcohols. These precursors include 2-ketoheptanoate (2KH, precursor to n-hexanol), 2-ketooctanoate (2KO, precursor of n-heptanol), and 2-ketononaoate (2KN, precursor to n-octanol), which, similar to 2 kV, could also be obtained via overexpressing LeuABCD [26]. This sub-pathway is significantly less prevalent in natural microbial metabolic systems, with the exception of species from the genus Clostridium, which possess an acetone–butanol–ethanol (ABE) pathway [27, 28]. In this strategy, the expansion of carbon chain is derived from the biological catalytic process in which butyryl-CoA is generated from the condensation reaction between two acetyl-CoA molecules via four enzymatically catalyzed steps: acetyl-CoA acetyltransferase, 3-hydroxybutyryl-CoA dehydrogenase, crotonase, and butyryl-CoA dehydrogenase [29,30,31].

Thus far, researchers have leveraged 2-keto acid intermediates as a biosynthesis strategy for producing various advanced biofuels with longer carbon chain. For example, the metabolic pathway of generating 3-methyl-1-pentanol using E.coli is derived from the Type II sub-pathway and requires two catalytic reactions[3, 32]: a carbon chain expansion reaction featuring catalyzing enzymes LeuABCD and a decarboxylation–reduction reaction involving 2-ketoisovalerate decarboxylase (Kivd) and alcohol dehydrogenase 2 (adh2). Meanwhile, advanced biofuels of unbranched alkane-based alcohols such as n-hexanol [33], which could also be generated from 2-keto acid intermediates through the Type II sub-pathway and it involved two important catalytic reactions featuring high-activity enzymes PaaH1, Crt, Ter in tandem, and an exogenous decarboxylation–reduction reaction: kivd-adhE (Fig. 4).

A large number of studies have proved that 2-isopropylmalate synthase (IPMS) and acetohydroxyacid synthase (AHAS) are two key enzymes involved in the expansion of carbon chains in the biosynthesis strategy that leverages 2-keto acid intermediates. In recent years, in order to improve the efficiency of expanding carbon chains, different carbon–carbon bond transition states catalyzed by LeuA mutants (LeuA*) were simulated using quantum mechanics and protein–substrate complexes to identify the best conformers [34]. A diverse range of advanced biofuels with longer carbon chain ranging from 6 to 9 carbons, such as heptanol and octanol, could be produced using this biosynthesis strategy [35, 36]. Therefore, this biosynthesis strategy is a promising alternative for creating a wider variety of advanced biofuels in place of those generated by the fossil industry.

Production of advanced biofuels using reverse β-oxidation intermediates

Biosynthesis strategy based on reverse β-oxidation intermediates has been demonstrated to show significant promises for expanding carbon chains to produce a variety of advanced biofuels (Table 2), such as various isobutanol and butanol from renewable resources like organic waste [37]. There are some common interactive intermediates between the metabolic pathways that rely on reverse β-oxidation intermediates and those that depend on 2-keto acid intermediates. Especially, the bacterial reverse β-oxidation intermediates have been well studied due to their typical functions in fatty acid catabolism, for which a balance could be maintained with acetyl-CoA as an energy source [38].

Table 2 Advanced biofuels using reverse β-oxidation intermediates

Acetyl-CoA is used as the elongation factor of the reverse β-oxidation intermediates (Fig. 5). The process of expanding carbon chains involves the decomposition of intermediates (Cn+2)-acyl-CoA into two main compounds, acetyl-CoA and (Cn)-acetyl CoA intermediates, via a series of catalytic reactions of the four catalyzing enzymes atoB, fadA, fadB, and fadE. A circulation process in which two carbon molecules from the fatty acid pathway are removed through the β-oxidation cycle, followed by the addition of one acetyl-CoA molecule to the tricarboxylic acid (TCA) cycle (Fig. 5). The carbon chain could be further elongated since the acetyl-CoA is added to another thioester due to the reversibility of the β-oxidation metabolic cycle. Finally, advanced biofuels could be produced after one round of this cyclic process. For example, to produce advanced biofuel 1-butanol, acetoacetyl-CoA is first obtained via a condensation reaction of two acetyl-CoA molecules by acetyl-CoA C-acetyltransferase. Subsequently, 3-hydroxybutyryl-CoA is obtained through 3-hydroxyacyl-CoA dehydrogenase or enoyl-CoA hydratase to reduce the β-keto acid products, which are subsequently converted into crotonyl-CoA. Then, crotonyl-CoA is hydrogenated and converted into butyryl-CoA, thereby, adding a carbon to the carbon chain. The butyryl-CoA is converted into 1-butyraldehyde by a CoA-dependent aldehyde dehydrogenase. Finally, 1-butanol is produced from 1-butyraldehyde via a reduction reaction using adhE. Carbon chain extension could be achieved by consecutive β-oxidation process, from which advanced biofuels with longer carbon chains could be produced [39, 40].

Fig. 5
figure 5

Biosynthesis pathway of expanding carbon chain for producing advanced biofuels using reverse β-oxidation intermediates. This biosynthesis pathway can use acetyl-CoA as a carbon chain elongation factor. Normal carbon chain extension in microorganisms occurs when (Cn + 2)-acyl-CoA molecules are broken down into acetyl-CoA and (Cn)-acetyl CoA molecules under the control of four genes (atoB, fadA, fadB, and fadE). However, chain elongation occurring through the addition of acetyl-CoA to another thioester due to β-oxidation cycle metabolic pathway is essentially reversible. Thus, advanced biofuels could be produced after one turn of this cycle. YqeF acetyl-CoA acetyltransferase; fadA ketoacyl-CoA thiolase; fadB hydroxyacyl-CoA dehydrogenase and enoyl-CoA hydratase; YdiO enoyl-CoA reductase; TES thioesterase; AcoAR acylCoA reductase; ADH alcohol dehydrogenase; aceEF-lpdA pyruvate dehydrogenase multi-enzyme complex; acetyl-CoA acetoacetyl-CoA transferase; atoB acetyl-CoA acyltransferase; atoE putative short-chain fatty acid transporter; dhaKLM PEP-dependent dihydroxyacetone kinase; fadA 3-ketoacyl-CoA thiolase; fadB enoyl-CoA hydratase/3-hydroxyaceyl-CoA dehydrogenase; fadD fatty acyl-CoA synthetase; fadE acyl-CoA dehydrogenase; fadL long-chain fatty acid outer membrane transporter; fbaA and fbaB: fructose-1,6-bisphosphate aldolase; fdhF formate dehydrogenase; galP galactose permease; gldA glycerol dehydrogenase; glk glucokinase; glpF glycerol MIP channel; hycB-1 hydrogenase; pflB pyruvate formate lyase; ptsHI-crr: ptsG glucose specific phosphoenolpyruvate phosphotransferase system; pykA and pykF pyruvate kinase; tpiA triose phosphate isomerase; xylA xyloseisomerase; xylB xylulokinase; xylE xylose MFS transporter; xylFGH: xylose ABC transporter. 2[H]: NAD(P)H = FADH2; DHA dihydroxyacetone, DHAP dihydroxyacetone-phosphate; Fructose-1: 6-BP fructose-1,6-bisphosphate. The red arrow shows the oxidation–reduction pathway to higher biofuel. The blue arrow is the carbon chain extension pathway

Previous studies have demonstrated that advanced biofuel synthesis could be influenced by fatty acid accumulation [41]. Under low fatty acid conditions, the catalytic enzymes in the reverse β-oxidation process could convert some intermediates from the fatty acid metabolic pathway into a diverse collection of alcohols with long carbon chain. Due to the cyclical nature of this biosynthesis strategy, the length of carbon chain and the degree of saturation of the metabolic intermediates could be controlled by selecting the appropriate terminal monospecific enzyme [42,43,44]. In addition, extensive modification of the carbon chain is feasible with this reverse β-oxidation intermediates by reducing fatty acid degradation, removing other carbon metabolic flux in the circuit branch, and avoiding feedback inhibition. Furthermore, reverse β-oxidation intermediates have been used to produce a variety of higher-order advanced biofuels including advanced fatty alcohols with 6–10 carbon atoms such as 1-hexanol, 1-octanol, and 1-decanol [45, 46].

Although the biosynthesis strategy based on reverse β-oxidation intermediates could be used to expand carbon chain in a manner that resembles the sequence of CoA-dependent reactions, the catalytic mechanism involved has yet to be thoroughly studied. Even so, advanced biofuels including some alcoholic compounds with long carbon chain (C6–C10) have been produced using reverse β-oxidation intermediates. For example, 1-hexanol at a yield of 37 mg/L was obtained by expressing alcohol dehydrogenases yiaY, eutG, and betA [15, 47] and expressing open reading frames ydiO and ydiQRST encoding components of the acyl-CoA dehydrogenase complex of anaerobic fatty acid β-oxidation [48]. However, it is unclear whether electron transfer flavoproteins and ferredoxin (YdiQRST) exert any important effects on the catalytic process [49, 50].

Nevertheless, a variety of fatty acids and alcohols with various carbon chain could be achieved by this approach due to the interaction between 2-keto acids and the β-oxidation process [51, 52]. Therefore, this biosynthesis strategy is considered to have great potential for the synthesis of advanced biofuels.

Production of advanced biofuels using fatty acid intermediates

Biodiesel fuel, as an advanced biofuel, has been proposed as a potential substitute for petroleum-based diesel. Biodiesel fuel is typically produced from fatty acid methyl esters (FAMEs) or fatty acid ethyl esters (FAEEs) via the conversion of plant or animal oils. Decarboxylated derivatives, mainly fatty alcohols, with long aliphatic chains or aromatic hydrocarbon molecules can also be synthesized via the transesterification of acylglycerols using this biosynthesis strategy based on fatty acid intermediates. For example, free fatty acids (FFAs) from cyanobacteria can be converted into biodiesel, alcohols, alkanes, and olefins with long carbon chain [53, 54], which are dependent on highly efficient catalytic enzyme in order to achieve esterification. It is well-known that eukaryotic algae, such as photosynthetic microorganisms and yeast S. cerevisiae can also naturally produce a great quantity of acylglycerols, which can be subsequently converted into various advanced biofuels [55,56,57]. However, studies have shown that it is rare for bacteria to naturally generate large quantities of FAMEs and FAEEs. Therefore, using bacteria as the engineering strains for expanding carbon chains to synthesize advanced biofuels based on FAMEs or FAEEs production is of key interest. In this study, two biometabolic pathways using fatty acid intermediates were reviewed according to the production of different kinds of advanced biofuels.

Production of advanced biofuels: fatty alcohols using fatty acid intermediates

Expanding carbon chain to produce non-natural fatty alcohols needs to use acetyl-CoA as the original carbon source in the FADs (Table 3). In some microorganisms, FFAs with long carbon chain from malonyl-CoA are synthesized using ATP-dependent carboxylation. Acyl units could also be added during elongation of carbon chain due to the decarboxylation effect of malonyl-CoA (Fig. 6). For example, acetyl-CoA in E. coli is first converted into malonyl-CoA under the actions of ATP-dependent-acetyl-CoA carboxylase (AccABCD). The FFA compound bound to an acyl carrier protein (ACP) is then produced through a NADPH-dependent reaction in which malonyl-CoA is converted by a multicomponent fatty acid synthase. These fatty acyl-ACPs are utilized as either carbon chain elongation structures or energy storage molecules. The FFAs are generated from these energy stores, and structural components can be reconstructed by overexpressing thioesterases that cleave fatty acyl-ACPs. Cleavage reaction of fatty acyl-ACP can lead to the accumulation of large amounts of fatty acyl-CoAs because the gene cytoplasmic (normally periplasmic) thiolase can hydrolyze fatty acyl-ACPs by fatty acid transport and fadDE gene. Finally, the FFAs with long carbon chain are added to some alcohols with short carbon chains between C8 and C18 under the action of different thioesterases (Fig. 6). For example, the fatty acyl-CoAs obtained from the above reactions can be modified into C12–C18 fatty alcohols in Saccharomyces cerevisiae and Escherichia coli [58, 59] by importation of a reductive pathway using fatty acyl-CoA reductase from other microorganisms such as Acinetobacter calcoaceticus [5, 58, 59].

Table 3 Advanced biofuels: fatty alcohols using fatty acid intermediates
Fig. 6
figure 6

Biosynthesis pathway to produce advanced biofuels of fatty alcohols using fatty acid intermediates. Advanced biofuels: fatty alcohols product from carbon chain extension of fatty acid synthesis. Mechanistically, carbon chain extension relies on acetyl-CoA as the primary carbon source in the fatty acid synthetic pathway via ATP-dependent carboxylation into malonyl-CoA. Acyl units are then provided for carbon elongation by cycles of decarboxylative addition of malonyl-CoA. accABCD acetyl-CoA carboxylase; acrI acyl-CoA reductase; asd aspartate semialdehyde dehydrogenase; atfA acyltransferase; fabA hydroxydecanoyl-ACP dehydrase; fabB ketoacyl-ACP synthase I; fabD malonyl-CoA-ACP transacylase; fabF ketoacyl-ACP synthase II; fabG ketoacyl-ACP reductase; fabH ketoacyl-ACP synthase III; fabI enoyl-ACP reductase; fabZ hydroxyacyl-ACP dehydratase; CER4 fatty acyl-CoA reductase acyl-CoA synthetase; FAEE fatty acid ethyl ester; Ac-CoA acetylCoA; ACP acyl carrier protein; CoA coenzyme A

Previous literature has demonstrated that yield of fatty alcohols can be increased via systemic improvement of metabolic genes involving fatty acid intermediates. For example, some studies have shown that deletion of fadD, overexpression of AccABCD genes, and heterologous expression of ACP thioesterase can successfully increase FFA levels in an Umbellularia californica plant, resulting in enhanced fatty acids production. Some reports show an effective approach that leveraged plasmids containing different copy numbers and collections of acyl-CoA ligase, thioesterase, and acyl-CoA reductase from various biospecies were reported for the optimization of fatty acids production. For example, E. coli and Saccharomyces were optimized using this biosynthesis pathway, which produced fatty alcohol at a yield of 0.81 mg/L with 12-carbon chain when glycerol was used as the substrate. In addition, a series of fatty acids with different lengths of carbon chain (C10–C18) and levels of saturation were also generated based on this biosynthesis pathway using E. coli [60,61,62].

Production of advanced biofuels: alkanes, alkenes and ketones using fatty acid intermediates

Alkanes and alkenes are the most important advanced biofuels that are used as substitutes for hydrocarbon fossil fuel such as jet fuels, diesel, and gasoline. With recent advances in system metabolic engineering, synthetic mechanisms of alkanes and alkenes have been investigated in depth to allow for a better understanding of their underlying metabolic pathways (Table 4). There are currently two metabolic pathways for producing alkanes and alkenes, namely elongation–decarboxylation [63, 64] and head-to-head condensation [65, 66], both of which rely on fatty acid intermediates for carbon chain expansion. The elongation–decarboxylation pathway uses acyl-CoA from fatty acid intermediates as an extension factor via the cyclic addition of the two-carbon molecules from the conversion of malonyl-CoA, followed by subsequent decarboxylation to produce alkanes and alkenes. The head-to-head condensation pathway involves the conjugation of two fatty acid derivatives that are converted into carboxylic acid via Claisen condensation, from which alkenes with odd carbon chain are subsequently generated by decarboxylation and reduction reactions [25, 67,68,69].

Table 4 Advanced biofuels: alkanes, alkenes and ketones using fatty acid intermediates

Previous studies have shown that pathways that convert FFAs or fatty acid intermediates into alkanes and alkenes can be defined by five diversity-oriented sub-pathways, including AAR-ADO, HEAD-TO-HEAD, OLeTJE OLS, and CAR/FAR-ADO (Fig. 7). For AAR-ADO, two very important catalytic enzymes from Synechococcus sp. PCC7002, namely AAR and ADO, participate in the synthesis of alkane [70]. The catalytic process involves Acyl-ACP reductase, which can reduce the conversion of ACP into aldehyde small compounds, followed by the conversion of aldehyde into alkane or alkene via ADO [71]. An engineered prokaryote, such as E. coli and Synechocystis sp. PCC6803, can produce multifarious alkanes and alkenes (C13–C17) that include some advanced biofuels, such as heptadecane, heptadecene, tridecane, pentadecene, pentadecane, heptadecene, pentadecane, and heptadecane, by overexpressing AAR and ADO derived from cyanobacteria strains [72].

Fig. 7
figure 7

Biosynthesis pathway to produce advanced biofuels of alkanes and alkenes using fatty acid intermediates. Increasing microbial synthesis of alka(e)nes via carbon chain extension of five different fatty acid synthesis pathways that can convert free fatty acids or fatty acid derivatives into alka(e)nes, classified as “elongation–decarboxylation” and “head-to-head condensation” biosynthetic metabolic approaches, including AAR-ADO, HEAD-TO-HEAD, OLeTJE, OLS, and CAR/FAR-ADO pathways. The elongation–decarboxylation approach is based on the use of acyl-coenzyme A (CoA) from a fatty acid synthesis pathway as an extension factor for carbon chain elongation via the circular addition of a two-carbon unit, conversion of malonyl-CoA, and subsequent decarboxylation to produce chain alka(e)nes. The second approach is head-to-head condensation, involving the conjugation of two fatty acid derivatives to a carboxylic acid via a Claisen condensation. The odd chain alka(e)ne is formed by decarboxylation and decarbonylation. ACP acyl–acyl carrier protein; ACC acetyl-CoA carboxylase; FabD malonyl-CoA ACP transacylase; FabH β-keto-acyl-ACP synthase III; FabB β-keto-acyl-ACP synthase I; FabG β-keto-acyl-ACP reductase; FabZ: β-hydroxyacyl-ACP dehydratase; FabI enoyl-acyl-ACP reductase; TE thioesterase; FadD acyl-CoA synthase; AAR acyl-ACP reductase; ADO: aldehyde-deformylating oxygenase; OleABCD: a four protein families for long-chain olefin biosynthesis; (OleTJE): fatty acid decarboxylase, a cytochrome P450 enzyme that reduces fatty acids to alkenes; CAR: carboxylic acid reductase; Sfp: A phosphopantetheinyl transferase; FAR: fatty acid reductase; Ols: a type I polyketide synthases for α-olefin biosynthesis. MVA indicates mevalonic pathway for isoprenoid biosynthesis. MEP indicates the methylerythritol phosphate pathway. Red, purple, blue fonts show the oxidation–reduction pathway to higher biofuel

Meanwhile, head-to-head condensation reaction [73]can generate advanced biofuels with long carbon chain via the expansion of carbon chain such as olefins (Fig. 7). For example, in Micrococcus luteus, olefin synthesis allows for the expansion of carbon chain via the action of four catalytic enzymes OleA, OleB, OleC, and OleD [74]. These four enzymes include an α/β-hydrolase, a thiolase, a short-chain dehydrogenase, and an AMP-dependent ligase or synthetase. Some unsaturated alkenes with long carbon chain, such as 27:3, 27:2, 29:3, and 29:2, can be produced using this sub-pathway [75, 76].

The engineering sub-pathway OLeTJE was first identified in the bacteria Jeotgalicoccus sp. ATCC 8456 (Fig. 7). The specific case is terminal n-alkene can be synthesized via conversion of FFAs into terminal olefins using this strategy [77]. For example, the α-olefin (C16–C20) can be produced through the expansion of carbon chain by direct decarboxylation of FFAs with long carbon chain with fatty acid decarboxylase OleTJE, using H2O2 as a source of electrons [78]. For example, 1,10-heptadecadiene and 1-pentadecene have been obtained using this engineering strategy [79].

The sub-pathway OLS pathway was first discovered in marine cyanobacterium Synechococcus sp. PCC 7002 and cyanobacterium Lyngbya majuscula. The ols gene is responsible for encoding type I polyketide synthase (PKS), including multi-domain ketosynthase (KS), acyltransferase (AT), and ketoreductase (KR). The expansion process of carbon chain involves the combination of acyl-ACP with the ACP1 domain to form an acyl-substrate. Then, two carbons from malonyl-CoA are added to the acyl-substrate by the central extension module of KS, AT, and KR. Subsequently, the ACP2 domain and β-keto group are reduced to generate β-hydroxyl, which is then activated by sulfotransferase (ST) via sulfation. Finally, the C-terminal of the thioesterase (TE) domain catalyzes dehydration and decarboxylation reactions to produce the advanced biofuels olefin [80]. For example, 1-nonadecene [81] and 1,14-nonadecadiene can be synthesized according to this engineering strategy using curM-encoding enzyme with ORF ols as the agent responsible for expressing olefin synthase [82, 83]. Some various short-chain ketones (C5–C7) such as ethyl ketone, methyl ketone can be produced by introducing a hybrid polyketide synthase (PKS) via fusing a PKS subunit of lipomycin synthase LipPks1 and a thioesterase (TE) in Streptomyces hosts [84].

The engineering sub-pathway CAR/FAR-ADO is dependent on three very important catalytic enzymes: carboxylic acid reductase (CAR), fatty acid reductase (FAR), and phosphopantetheinyl transferase (Sfp) [85]. The CAR and Sfp catalytic enzymes are derived from two bacteria, Mycobacterium marinum [86] and Bacillus subtilis [87], respectively. The process of expanding carbon chain involves the conversion of FFAs into fatty aldehydes by CAR or FAR complex with the genes luxC, luxE, and luxD isolated from Photorhabdus luminescens [88, 89]. After oxidation, the fatty aldehydes are converted into alkanes and alkenes with straight or branched chains. This conversion of fatty aldehydes is responsible for the ADO catalytic reaction by Sfp from Cyanobacteria in combination with a B. subtilis thioesterase, branched chain α-keto acid dehydrogenase complex, lipase, and β-keto-acyl-ACP synthase III [90]. Some linear alkanes, such as pentadecane, heptadecene, branched tridecane, heptadecane, hexadecane, hexadecane, pentadecane, pentadecene, tridecane, tridecene, and undecane can be obtained via this engineering sub-pathway [91,92,93].

In summary, the biosynthesis pathways based on 2-keto acid intermediates, fatty acid intermediates or reverse β-oxidation intermediates could be used to add “ + 1” or “ + 2” carbon molecules to carbon chain. Addition of one-carbon molecule could agitate the whole metabolic network by influencing the process of catalytic reactions (Fig. 8). Therefore, a mixture including various advanced biofuels with longer branched or unbranched carbon chains, such as alkanes, alkenes and alcohols, can be obtained during carbon chain expansion by leveraging specific biometabolic pathways for the addition of each carbon molecule. These pathways are presented as an important tool for the production of aliphatic acids and advanced biofuels due to their ability to selectively lengthen carbon chain via multiple carbon cycles. This exact level of control in the process of carbon chain expansion has been demonstrated using E. coli to produce many advanced biofuels such as isobutanol at a yield of 22 g/L [19, 94]. Currently, the engineering strategy with 2-keto acid intermediates is one of the most thoroughly researched strategies that combines other engineering technologies derived from protein-substrate, quantum mechanical, metabonomics, and structural protein.

Fig. 8
figure 8

The complex relationship in different biosynthesis pathways of expanding carbon chain. Every one-carbon chain extension generates a global net effect per catalytic cycle regardless of whether the pathway involving 2-keto acid intermediate (“ + 1” or “ + 2” pathway) or a biosynthesis pathway using fatty acid intermediates and reverses β-oxidation. A mixture containing various advanced biofuels namely, longer, branched or unbranched carbon chains (e.g., alcohols) is capable of producing via the per addition of one carbon. Gly-3-P: glyceraldehyde-3-phosphate; PEP phosphoenolpyruvate; Pv pyruvate; MC malonyl-CoA; MA malonyl-ACP; AC acyl-CoA

In the aforementioned biometabolic pathways for expanding carbon chain, CoA molecules are first converted to some intermediates via addition of carbon molecule. For example, synthesis of butanol involves the extension of carbon chain using a series of CoA molecules: acetyl-CoA acetyltransferase, 3-hydroxybutyryl-CoA dehydrogenase, crotonase, and butyryl-CoA dehydrogenase [95]. Therefore, selecting an appropriate engineering strategy is critical for fine-tuning of both the expression of multiple catalytic enzymes and the concurrent adaption of cellular physiology during carbon chain expansion.

Technical approaches for overcoming low efficiency in carbon chain expansion

In order to overcome the problem of low efficiency, from the early traditional modification techniques such as eliminating the generation pathway of by-products, releasing the synthesis inhibition of products, overexpression of key enzymes in the synthesis pathway, to the development of metabolic response database, and machine learning-based network model calculation, optimal assembly of metabolic pathways, dynamic regulation of gene expression and other new metabolic modification techniques. There are usually the following technical approaches for overcoming low efficiency in carbon chain expansion.

DNA fragment assembly technology

The creation and optimization of biosynthetic pathways for carbon chain extension first require a breakthrough in DNA fragment assembly technology. DNA fragment assembly technology is an important prerequisite for the creation of biosynthetic pathways for producing biofuel [96]. Traditional DNA assembly relies mainly on restriction endonuclease and DNA ligase. However, this assembly method has certain limitations, such as the “scar” of enzyme restriction site will be left at the junction, and the corresponding enzyme restriction site cannot exist in the gene fragment to be assembled, thus reducing the range of selective endonuclease [97, 98]. The latest technologies include the following three.

  1. (1)

    Golden Gate assembly technique

Restriction endonuclease Type II, which has features that identify and cut the position inconsistencies of double-stranded DNA, is required. This property enables it to cut DNA outside the recognition site, and the sticky ends generated are connected sequentially under the action of ligase and assembled into DNA fragments without the enzyme cleavage site, realizing the seamless connection of multiple fragments. It maybe provide opportunities in creating a flexible, versatile, and data-driven framework to support biofuel research and development in biofoundries [99, 100].

  1. (B)

    Gibson assembly technique

No restriction enzyme is required, but the ends of each DNA fragment contain homologous sequences of 15-20 bp, according to which the assembly is carried out in the order of homologous sequences. The assembly process involves several enzymes: DNA 5' exonuclease, which first cuts out a sticky terminal, which complicates each other according to homology, and polymerase DNA polymerase, which replicates the missing bases. Finally, the sticky end is attached by DNA ligase. This method allows one or more DNA fragments to be inserted into a linearized vector quickly, efficiently and precisely in a predetermined direction, achieving “seamless” assembly [101]. This technology has been used to design, build, test, learn steps of yeast-based biofuel production [102, 103].

  1. (C)

    CPEC technique

Circular polymerase extension cloning (CPEC) is a short name for circular polymerase extension cloning. The requirements for DNA fragments are similar to Gibson's, and the ends contain homologous sequences. CPEC is a technology to realize the assembly of DNA fragments by PCR: DNA fragments and vectors are denatured and decomposed, and the end homologous sequences complement each other during annealing. The sequences are mutual templates and primers and extend into circular DNA molecules containing gaps under the action of DNA polymerase [104]. The gaps can be repaired in E.coli to obtain complete plasmids. In general, CPEC is a simpler, more efficient and economical DNA assembly technology with more extensive applications, but it is not suitable for the assembly of DNA fragments with high GC content. A standard CPEC protocol 67 was used to assemble the three PCR products to produce biofuel [105, 106].

Gene expression regulation techniques

Gene expression regulation is an important means to optimize biosynthetic pathways and is the core to overcome the low efficiency of target product production. After the pathway is created in the host, it is necessary to regulate the expression intensity of genes at key nodes, including enhancing the product synthesis pathway and cofactor synthesis pathway, or weakening some genes that will lead to non-growth of bacteria and have adverse effects on product production after knockout. According to the technical level, it can be divided into single gene regulation, polygene regulation and gene dynamic regulation.

  1. (1)

    Single gene regulation

Single gene regulation is to regulate the expression of a specific gene of metabolic pathway on chromosomes. In the early stage of metabolic engineering, strong promoters are commonly used for single gene regulation, such as IPTG-induced Tac promoter and T7 promoter. However, strong expression of single genes may not be optimal for metabolic pathways. To achieve optimal expression of a single gene, promoter libraries in E. coli were constructed and to obtain promoters of different strengths for the regulation of glucose transporter gene galP and glucokinase gene glk [107, 108]. The combined regulatory strain GalP93-GIk37 significantly increased the rate of glucose consumption. In addition to the promoter library, the regulation of single genes of the succinic acid transporters DcuB and DcuC by RBS library can also improve the ability to transport succinic acid to the cell and increase the yield of the final product 2,3-butanediol [109,110,111].

  1. (B)

    Multi-gene regulation

Single-gene regulation can only be operated on the single gene dimension. Even if combined regulation, the library capacity is greatly reduced, and it is difficult to screen out the optimal combination of multiple genes. In the optimization of metabolic pathway, to achieve the efficient production of target biofuel with long-carbon chain, the cooperative expression of multiple genes is often required to achieve the optimization of metabolic pathway. Keasling's team developed a tunable intergenic region (TIGR) library technique [112]. Based on the principle that inter-gene sequence changes affect gene expression intensity, this technique achieves the aim of simultaneously regulating the expression intensity of multiple genes within a single operon [113, 114]. Multiple genes of mevalonate pathway in E.coli realized cooperative expression, and mevalonate production was increased by 7 times using this technique [115, 116]. Isaacs et al. developed multiplex automated genome engineering (MAGE) technology, which improves the efficiency of multiplexed gene editing using red homologous recombination system and automatic cycling equipment [117]. Based on the subsequent CRISPR/Cas9 gene-editing technology, three gene libraries of xylose metabolic pathway were regulated on chromosomes, the optimal strain was screened, and the xylose metabolic rate was increased by three times [118, 119].

  1. (C)

    Gene dynamic regulation

Dynamic regulation is one of the most effective strategies in metabolic pathway optimization and an important technique to improve the target product. Although static regulation such as single- and multigene regulation is easy to operate and effective, the intracellular metabolites are dynamic. If the expression level of related genes is too high, the cell resources will be wasted; if the expression level is too low, the metabolic pathway will be restricted, and the yield and output of the final target products will be difficult to improve due to the imbalance of cell metabolism. In order to timely balance the relationship between gene expression and global metabolism required for product synthesis, the introduction of gene dynamic regulation can respond to metabolic signals in real time, and make timely feedback regulation to adapt to changes in cellular metabolism and environment. For example, the S. cerevisiae was engineered to produce and secrete 1-alkenes by manipulation of the fatty acid metabolism, enzyme selection, engineering the electron transfer system and expressing a transporter [120]. Furthermore, a dynamic regulation strategy were implemented to control the expression of membrane enzyme and transporter, which improved 1-alkene production [121,122,123].

Genome editing technique

In order to improving microbial synthesis efficiency, through gene editing, exogenous gene insertion, endogenous gene knockout and key gene expression regulation can be realized for expanding carbon chain. Homologous recombination system is the basis of bacterial and fungal genome editing technology [124, 125]. The most commonly used system is the red homologous recombination system, which is derived from phage and includes three proteins, Exo, Beta and Gam, and achieves homologous recombination of foreign fragments with the coordination of these three proteins [125]. Homologous recombination is widely used in E.coli and other microorganism gene editing for producing biofuels [126]. Based on the Red homologous recombination system and combining different strategies, researchers have developed resistance-free genome editing techniques, which are commonly used in three ways:

  1. (1)

    Two-step homologous recombination based on FIp recombinase

Flp recombinase can recognize short Flippase recognition site sequences and excise the sequences between two flippase recognition targets (FRTS) [127]. Combined with Red homologous recombination system, the resistance genes with FRT sequences at both ends were integrated into the target site on the genome, and then the resistance genes were knocked out by Flp recombinase. The disadvantage of this technique is that it will leave a FRT sequence at the target site, which is not traceless editing and will affect the editing efficiency of genes after multiple rounds of editing.

  1. (B)

    Two-step positive and negative screening method based on sacB gene

The sacB gene encodes secreted sucrose fructanase, which can catalyze the hydrolysis of sucrose into glucose and fructose, and synthesize fructose into high molecular weight fructan, which can produce lethal effect on E. coli, and is a negative screening marker. Under the guidance of Red homologous recombination system, the fragments containing resistance genes and sacB genes were first integrated into the target site [128], then the target fragments were integrated into the target site, and the gene fragments containing resistance genes and sacB genes were replaced, and then cultured in the medium containing sucrose. The correct transformants replacing the resistance gene and sacB gene fragment were obtained by reverse screening strategy.

  1. (C)

    One-step homologous recombination based on CRISPR/Cas9

Genome editing technique, such as CRISPR–Cas, is extensively used in a diverse array of fields, with practical applications in the biological synthesis of new compounds. It is a powerful tool for studying eukaryotic microorganisms and has been reported to be useful for knocking out a specific targeted gene, simultaneously operating multiple genes, disrupting, activating, and repressing genes, and editing the genome in a genetically known or unknown microorganism [129]. Cas9 protein was cleaved at target sites under the mediation of gRNA, which caused the double chain break of E. coli genome and produced biofuels [130, 131]. The Red homologous recombination system can integrate the donor fragment into the target site, repair the broken DNA double strand, or undergo homologous recombination before Cas9 protein splicing, so that Cas9 loses the target site and does not splice.

The integration of synthetic biology, genome-editing technique like CRISPR/Cas9, and high-throughput sequencing technology, would lend feasibility to expanding carbon chain for the production of new advanced biofuels in industrial-scale applications. For example, some studies have been showed lignocellulosic biofuel production can be improved by CRISPR/Cas9-mediated lignin modification [132, 133]. Furthermore, it is highly possible that novel biosynthesis pathway could be discovered using this technique, which could lead to the acquisition of a wide variety of novel synthetic systems with high catalytic performance. This could in turn motivate novel gene expression system for carbon chain expansion to generate undiscovered advanced biofuels. Today, gene knockout and gene cassette insertions employing CRISPR–Cas9 in Saccharomyces cerevisiae and Kluyveromyces marxianus have resulted in enhanced production of bioethanol and 2-phenyl ethanol in these organisms, respectively [134]. Designing new engineering strategy and controlling synthesis processes could be greatly simplified, thus opening up an unprecedented field for the production of advanced biofuels.

One advantage of carbon chain expansion is that it allows refactoring of any potential catalytic reaction to be performed in a simple, convenient, inexpensive, efficient manner. This technology also lends practicality towards transformation of cellular structures of microorganisms and recombination of metabolic networks, especially in changing mitochondria to improve their synthesis efficiency. For example, substantial increase in production of advanced biofuels had been attributed to the close link between genetic improvement of the whole signaling network and recombination of the whole metabolic flux, such as genome editing of algal species by CRISPR–Cas9 for biofuels [135].

The potential application of this technology is continually being explored in different microorganism platforms, along with bioinformatics and large-scale modeling, for analysis at the genome level, transcriptome level, proteome level, metabolome depth, and fluxome-level. Findings from these studies would contribute to better understanding of the biochemical evolution and functional strain during gene function elucidation and epigenetic regulation in loss-of-function (LOF) [136] or gain-of-function (GOF) research [137]. This knowledge would also advance the carbon chain expansion process for producing advanced biofuels [189] and allow for the development of new target production from fungi via CRISPR–Cas9, instead of bacteria such as Streptomyces with fine-tuning the regulation [138,139,140,141,142], Trichoderma reesei with improving cellulase production [143], and Aspergillus species with improving succinic acid production from renewable biomass [144, 145] and yeast with genetic recombination for cellodextrin transport [190].

Outlook

The rapid development of biosynthesizing advanced biofuels, coupled with the demand of newly reconstructed engineering pathway for synthesizing novel chemicals and the need to improve efficiency of existing strategies, require strengthening of previous biometabolic pathways and discovery of novel biosynthesis pathways. In this paper, the low efficiency problem of production in expanding carbon chain was presented, followed by the proposal of two biosynthesis strategies RM and ED, and a review of corresponding biosynthetic pathway for producing advanced biofuels. Our findings contribute to future design of novel catalytic routes to maximize the production of target product. In addition, genome editing technique such as the representative CRISPR/Cas9 gene-editing technology will generate a traction for effective resolution of many technical obstacles in discovering new engineering strategy and increasing production of advanced biofuels.

Availability of data and materials

No data are associated with this article.

References

  1. Martins F, Felgueiras C, Smitkova M, Caetano N. Analysis of fossil fuel energy consumption and environmental impacts in European countries. Energies. 2019;12(6):964.

    Article  CAS  Google Scholar 

  2. Sharma P, Bano A, Singh SP, Atkinson JD, Lam SS, Iqbal HMN, Tong YW. Biotransformation of food waste into biogas and hydrogen fuel—a review. Int J Hydrog Energ. 2022. https://doi.org/10.1016/j.ijhydene.2022.08.081.

    Article  Google Scholar 

  3. Zhang K, Sawaya MR, Eisenberg DS, Liao JC. Expanding metabolism for biosynthesis of nonnatural alcohols. Proc Natl Acad Sci. 2008;105(52):20653–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Atsumi S, Liao JC. Metabolic engineering for advanced biofuels production from Escherichia coli. Curr Opin Biotechnol. 2008;19(5):414–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lennen RM, Pfleger BF. Microbial production of fatty acid-derived fuels and chemicals. Curr Opin Biotechnol. 2013;24(6):1044–53.

    Article  CAS  PubMed  Google Scholar 

  6. Tashiro Y, Rodriguez GM, Atsumi S. 2-Keto acids based biosynthesis pathways for renewable fuels and chemicals. J Ind Microbiol Biotechnol. 2015;42(3):361–73.

    Article  CAS  PubMed  Google Scholar 

  7. Wang S-Z, Zhang Y-H, Ren H, Wang Y-L, Jiang W, Fang B-S. Strategies and perspectives of assembling multi-enzyme systems. Crit Rev Biotechnol. 2017;37(8):1024–37.

    Article  CAS  PubMed  Google Scholar 

  8. Cann AF, Liao JC. Production of 2-methyl-1-butanol in engineered Escherichia coli. Appl Microbiol Biotechnol. 2008;81(1):89–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Vivek N, Pandey A, Binod P. 31—production and applications of 1,3-propanediol. In: Pandey A, Negi S, editors. Current Developments in biotechnology and bioengineering. Soccol CR: Elsevier; 2017. p. 719–38.

    Chapter  Google Scholar 

  10. Wang X, Zhang L, Chen H, Wang P, Yin Y, Jin J, Xu J, Wen J. Rational proteomic analysis of a new domesticated Klebsiella pneumoniae x546 producing 1,3-propanediol. Front Microbiol. 2021. https://doi.org/10.3389/fmicb.2021.770109.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kim DI, Chae TU, Kim HU, Jang WD, Lee SY. Microbial production of multiple short-chain primary amines via retrobiosynthesis. Nat Commun. 2021;12(1):173.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cheng J, Li J, Zheng L. Achievements and perspectives in 1,4-butanediol production from engineered microorganisms. J Agric Food Chem. 2021;69(36):10480–5.

    Article  CAS  PubMed  Google Scholar 

  13. Adi Putra Z, Kurnia JC, Sasmito AP, Muraza O. Process design and techno-economic analysis of ethyl levulinate production from carbon dioxide and 1,4-butanediol as an alternative biofuel and fuel additive. Int J Energy Res. 2019;43(11):5932–45.

    Article  CAS  Google Scholar 

  14. Cheon S, Kim HM, Gustavsson M, Lee SY. Recent trends in metabolic engineering of microorganisms for the production of advanced biofuels. Curr Opin Chem Biol. 2016;35:10–21.

    Article  CAS  PubMed  Google Scholar 

  15. Rabinovitch-Deere CA, Oliver JWK, Rodriguez GM, Atsumi S. Synthetic biology and metabolic engineering approaches to produce biofuels. Chem Rev. 2013;113(7):4611–32.

    Article  CAS  PubMed  Google Scholar 

  16. El-Dalatony MM, Saha S, Govindwar SP, Abou-Shanab RAI, Jeon B-H. Biological conversion of amino acids to higher alcohols. Trends Biotechnol. 2019;37(8):855–69.

    Article  CAS  PubMed  Google Scholar 

  17. Su H, Lin J, Wang G. Metabolic engineering of Corynebacterium crenatium for enhancing production of higher alcohols. Sci Rep. 2016;6(1):39543.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Peralta-Yahya PP, Zhang F, del Cardayre SB, Keasling JD. Microbial engineering for the production of advanced biofuels. Nature. 2012;488(7411):320–8.

    Article  CAS  PubMed  Google Scholar 

  19. Atsumi S, Hanai T, Liao JC. Non-fermentative pathways for synthesis of branched-chain higher alcohols as biofuels. Nature. 2008;451(7174):86–9.

    Article  CAS  PubMed  Google Scholar 

  20. Qiu M, Shen W, Yan X, He Q, Cai D, Chen S, Wei H, Knoshaug EP, Zhang M, Himmel ME, et al. Metabolic engineering of Zymomonas mobilis for anaerobic isobutanol production. Biotechnol Biofuels. 2020;13(1):15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Vogt M, Brüsseler C. Ooyen Jv, Bott M, marienhagen J: production of 2-methyl-1-butanol and 3-methyl-1-butanol in engineered Corynebacterium glutamicum. Metab Eng. 2016;38:436–45.

    Article  CAS  PubMed  Google Scholar 

  22. Zhang Y, Lane S, Chen J-M, Hammer SK, Luttinger J, Yang L, Jin Y-S. Avalos‬ JL: xylose utilization stimulates mitochondrial production of isobutanol and 2-methyl-1-butanol in Saccharomyces cerevisiae. Biotechnol Biofuels. 2019;12(1):223.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Su H, Lin J, Tan F. Progress and perspective of biosynthetic platform for higher-order biofuels. Renew Sustain Energy Rev. 2017;80:801–26.

    Article  Google Scholar 

  24. Dhamankar H, Prather KLJ. Microbial chemical factories: recent advances in pathway engineering for synthesis of value added chemicals. Curr Opin Struct Biol. 2011;21(4):488–94.

    Article  CAS  PubMed  Google Scholar 

  25. Jiang W, Gu P, Zhang F. Steps towards ‘drop-in’ biofuels: focusing on metabolic pathways. Curr Opin Biotechnol. 2018;53:26–32.

    Article  CAS  PubMed  Google Scholar 

  26. Connor MR, Liao JC. Microbial production of advanced transportation fuels in non-natural hosts. Curr Opin Biotechnol. 2009;20(3):307–15.

    Article  CAS  PubMed  Google Scholar 

  27. Abdelaal AS, Yazdani SS. Engineering E. coli to synthesize butanol. Biochem Soc Trans. 2022;50(2):867–76.

    Article  CAS  PubMed  Google Scholar 

  28. Pugazhendhi A, Mathimani T, Varjani S, Rene ER, Kumar G, Kim S-H, Ponnusamy VK, Yoon J-J. Biobutanol as a promising liquid fuel for the future—recent updates and perspectives. Fuel. 2019;253:637–46.

    Article  CAS  Google Scholar 

  29. Branduardi P, de Ferra F, Longo V, Porro D. Microbial n-butanol production from clostridia to non-clostridial hosts. Int J Energy Res. 2014;14(1):16–26.

    CAS  Google Scholar 

  30. Ferreira S, Pereira R, Wahl SA, Rocha I. Metabolic engineering strategies for butanol production in Escherichia coli. Biotechnol Bioeng. 2020;117(8):2571–87.

    Article  CAS  PubMed  Google Scholar 

  31. Li J, Baral NR, Jha AK. Acetone–butanol–ethanol fermentation of corn stover by Clostridium species: present status and future perspectives. World J Microbiol Biotechnol. 2014;30(4):1145–57.

    Article  CAS  PubMed  Google Scholar 

  32. Su H, Jiang J, Lu Q, Zhao Z, Xie T, Zhao H, Wang M. Engineering Corynebacterium crenatum to produce higher alcohols for biofuel using hydrolysates of duckweed (Landoltia punctata) as feedstock. Microb Cell Fact. 2015;14(1):16.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Machado HB, Dekishima Y, Luo H, Lan EI, Liao JC. A selection platform for carbon chain elongation using the CoA-dependent pathway to produce linear higher alcohols. Metab Eng. 2012;14(5):504–11.

    Article  CAS  PubMed  Google Scholar 

  34. Zhang F, Rodriguez S, Keasling JD. Metabolic engineering of microbial pathways for advanced biofuels production. Curr Opin Biotechnol. 2011;22(6):775–83.

    Article  CAS  PubMed  Google Scholar 

  35. Rodriguez M, Atsumi SJCCBG. Synthetic biology approaches to produce C3–C6 alcohols from microorganisms. Curr Chem Biol. 2012;6(1):32–41.

    CAS  Google Scholar 

  36. McEwen JT, Atsumi S. Alternative biofuel production in non-natural hosts. Curr Opin Biotechnol. 2012;23(5):744–50.

    Article  CAS  PubMed  Google Scholar 

  37. Lian J, Zhao H. Reversal of the β-oxidation cycle in Saccharomyces cerevisiae for production of fuels and chemicals. ACS Synth Biol. 2015;4(3):332–41.

    Article  CAS  PubMed  Google Scholar 

  38. Zhang Q, Zeng W, Xu S, Zhou J. Metabolism and strategies for enhanced supply of acetyl-CoA in Saccharomyces cerevisiae. Biores Technol. 2021;342:125978.

    Article  CAS  Google Scholar 

  39. Gulevich AY, Skorokhodova AY, Sukhozhenko AV, Shakulov RS, Debabov VG. Metabolic engineering of Escherichia coli for 1-butanol biosynthesis through the inverted aerobic fatty acid β-oxidation pathway. Biotech Lett. 2012;34(3):463–9.

    Article  CAS  Google Scholar 

  40. Generoso WC, Schadeweg V, Oreb M, Boles E. Metabolic engineering of Saccharomyces cerevisiae for production of butanol isomers. Curr Opin Biotechnol. 2015;33:1–7.

    Article  CAS  PubMed  Google Scholar 

  41. Lei A, Chen H, Shen G, Hu Z, Chen L, Wang J. Expression of fatty acid synthesis genes and fatty acid accumulation in haematococcus pluvialis under different stressors. Biotechnol Biofuels. 2012;5(1):18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Goepfert S, Poirier Y. β-Oxidation in fatty acid degradation and beyond. Curr Opin Plant Biol. 2007;10(3):245–51.

    Article  CAS  PubMed  Google Scholar 

  43. Marella ER, Holkenbrink C, Siewers V, Borodina I. Engineering microbial fatty acid metabolism for biofuels and biochemicals. Curr Opin Biotechnol. 2018;50:39–46.

    Article  CAS  PubMed  Google Scholar 

  44. Janßen HJ, Steinbüchel A. Fatty acid synthesis in Escherichia coli and its applications towards the production of fatty acid based biofuels. Biotechnol Biofuels. 2014;7(1):7.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Wu J, Zhang X, Xia X, Dong M. A systematic optimization of medium chain fatty acid biosynthesis via the reverse beta-oxidation cycle in Escherichia coli. Metab Eng. 2017;41:115–24.

    Article  PubMed  Google Scholar 

  46. Beller HR, Lee TS, Katz L. Natural products as biofuels and bio-based chemicals: fatty acids and isoprenoids. Nat Prod Rep. 2015;32(10):1508–26.

    Article  CAS  PubMed  Google Scholar 

  47. Lamsen E, Atsumi S. Recent progress in synthetic biology for microbial production of C3–C10 alcohols. Front Microbiol. 2012. https://doi.org/10.3389/fmicb.2012.00196.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Gulevich AY, Skorokhodova AY, Debabov VG. Escherichia coli ydiO and ydiQRST genes encode components of acyl-CoA dehydrogenase complex of anaerobic fatty acid β-oxidation pathway. Russ J Genet. 2016;52(10):1101–4.

    Article  CAS  Google Scholar 

  49. Gulevich AY, Skorokhodova AY, Debabov VG. Evaluation of the efficiency of functional reversal of fatty acid Β-oxidation in Escherichia coli upon the action of various native Acyl-CoA dehydrogenases. Appl Biochem Microbiol. 2022;58(4):361–7.

    Article  CAS  Google Scholar 

  50. Dellomonaco C, Clomburg JM, Miller EN, Gonzalez R. Engineered reversal of the β-oxidation cycle for the synthesis of fuels and chemicals. Nature. 2011;476(7360):355–9.

    Article  CAS  PubMed  Google Scholar 

  51. Liao JC, Mi L, Pontrelli S, Luo S. Fuelling the future: microbial engineering for the production of sustainable biofuels. Nat Rev Microbiol. 2016;14(5):288–304.

    Article  CAS  PubMed  Google Scholar 

  52. Liu Y, Cruz-Morales P, Zargar A, Belcher MS, Pang B, Englund E, Dan Q, Yin K, Keasling JD. Biofuels for a sustainable future. Cell. 2021;184(6):1636–47.

    Article  CAS  PubMed  Google Scholar 

  53. Savakis P, Hellingwerf KJ. Engineering cyanobacteria for direct biofuel production from CO2. Curr Opin Biotechnol. 2015;33:8–14.

    Article  CAS  PubMed  Google Scholar 

  54. Machado IMP, Atsumi S. Cyanobacterial biofuel production. J Biotechnol. 2012;162(1):50–6.

    Article  CAS  PubMed  Google Scholar 

  55. Cuellar-Bermudez SP, Garcia-Perez JS, Rittmann BE, Parra-Saldivar R. Photosynthetic bioenergy utilizing CO2: an approach on flue gases utilization for third generation biofuels. J Clean Prod. 2015;98:53–65.

    Article  CAS  Google Scholar 

  56. Białecka-Florjańczyk E, Kapturowska AUJCB: Genetically modified baker’s yeast saccharomyces cerevisiae in chemical synthesis and biotransformations. In book: Chemical Biol. 2012.

  57. Papapetridis I, Goudriaan M, Vázquez Vitali M, de Keijzer NA, van den Broek M, van Maris AJA, Pronk JT. Optimizing anaerobic growth rate and fermentation kinetics in Saccharomyces cerevisiae strains expressing Calvin-cycle enzymes for improved ethanol yield. Biotechnol Biofuels. 2018;11(1):17.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Runguphan W, Keasling JD. Metabolic engineering of Saccharomyces cerevisiae for production of fatty acid-derived biofuels and chemicals. Metab Eng. 2014;21:103–13.

    Article  CAS  PubMed  Google Scholar 

  59. Opgenorth P, Costello Z, Okada T, Goyal G, Chen Y, Gin J, Benites V, de Raad M, Northen TR, Deng K, et al. Lessons from two design–build–test–learn cycles of dodecanol production in Escherichia coli aided by machine learning. ACS Synth Biol. 2019;8(6):1337–51.

    Article  CAS  PubMed  Google Scholar 

  60. Kim S, Cheong S, Chou A, Gonzalez R. Engineered fatty acid catabolism for fuel and chemical production. Curr Opin Biotechnol. 2016;42:206–15.

    Article  CAS  PubMed  Google Scholar 

  61. Sarria S, Kruyer NS, Peralta-Yahya P. Microbial synthesis of medium-chain chemicals from renewables. Nat Biotechnol. 2017;35(12):1158–66.

    Article  CAS  PubMed  Google Scholar 

  62. Yu P, Tai Y-S, Woodruff AP, Xiong M, Zhang K. Engineering artificial metabolic pathways for biosynthesis. Curr Opin Chem Eng. 2012;1(4):373–9.

    Article  Google Scholar 

  63. Liu K, Li S. Biosynthesis of fatty acid-derived hydrocarbons: perspectives on enzymology and enzyme engineering. Curr Opin Biotechnol. 2020;62:7–14.

    Article  CAS  PubMed  Google Scholar 

  64. Lin F-M, Marsh ENG, Lin XN. Recent progress in hydrocarbon biofuel synthesis: pathways and enzymes. Chin Chem Lett. 2015;26(4):431–4.

    Article  CAS  Google Scholar 

  65. Goblirsch BR, Frias JA, Wackett LP, Wilmot CM. Crystal structures of Xanthomonas campestris OleA reveal features that promote head-to-head condensation of two long-chain fatty acids. Biochemistry. 2012;51(20):4138–46.

    Article  CAS  PubMed  Google Scholar 

  66. Madavi TB, Chauhan S, Keshri A, Alavilli H, Choi K-Y, Pamidimarri SDVN. Whole-cell biocatalysis: advancements toward the biosynthesis of fuels. Biofuels, Bioprod Biorefin. 2022;16(3):859–76.

    Article  CAS  Google Scholar 

  67. Kaur D, Singh RP, Gupta S. Screening and characterization of next-generation biofuels producing bacterial strains. Curr Microbiol. 2022;79(3):85.

    Article  CAS  PubMed  Google Scholar 

  68. Wackett LP. Engineering microbes to produce biofuels. Curr Opin Biotechnol. 2011;22(3):388–93.

    Article  CAS  PubMed  Google Scholar 

  69. James OO, Maity S. Chapter 11—carbon-carbon (CC) bond forming reactions for the production of hydrocarbon biofuels from biomass-derived compounds. In: Maity SK, Gayen K, Bhowmick TK, editors. Hydrocarbon biorefinery. Amsterdam: Elsevier; 2022. p. 297–325.

    Chapter  Google Scholar 

  70. Kawahara A, Hihara Y. Biosynthesis of fatty acid derivatives by cyanobacteria: from basics to biofuel production. In: Nielsen Jens, Lee Sang, Stephanopoulos Gregory, Hudson Paul, editors. Cyanobacteria biotechnology. Hoboken: Wiley; 2021. p. 331–67.

    Chapter  Google Scholar 

  71. Wang L, Chen L, Yang S, Tan X. Photosynthetic conversion of carbon dioxide to oleochemicals by cyanobacteria: recent advances and future perspectives. Frontiers in Microbiology. 2020. https://doi.org/10.3389/fmicb.2020.00634.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Kudo H, Hayashi Y, Arai M. Identification of non-conserved residues essential for improving the hydrocarbon-producing activity of cyanobacterial aldehyde-deformylating oxygenase. Biotechnol Biofuels. 2019;12(1):89.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Sukovich DJ, Seffernick JL, Richman JE, Gralnick JA, Wackett LP. Widespread head-to-head hydrocarbon biosynthesis in bacteria and role of OleA. Appl Environ Microbiol. 2010;76(12):3850–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Beller HR, Goh E-B, Keasling JD. Genes involved in long-chain alkene biosynthesis in Micrococcus luteus. Appl Environ Microbiol. 2010;76(4):1212–23.

    Article  CAS  PubMed  Google Scholar 

  75. Herman NA, Zhang W. Enzymes for fatty acid-based hydrocarbon biosynthesis. Curr Opin Chem Biol. 2016;35:22–8.

    Article  CAS  PubMed  Google Scholar 

  76. Surger M, Angelov A, Liebl W. Distribution and diversity of olefins and olefin-biosynthesis genes in Gram-positive bacteria. Biotechnol Biofuels. 2020;13(1):70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Belcher J, McLean KJ, Matthews S, Woodward LS, Fisher K, Rigby SEJ, Nelson DR, Potts D, Baynham MT, Parker DA, et al. Structure and biochemical properties of the alkene producing cytochrome P450 OleT (CYP152L1) from the Jeotgalicoccus sp. 8456 bacterium *. J Biol Chem. 2014;289(10):6535–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Jiang Y, Li Z, Wang C, Zhou YJ, Xu H, Li S. Biochemical characterization of three new α-olefin-producing P450 fatty acid decarboxylases with a halophilic property. Biotechnol Biofuels. 2019;12(1):79.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Tian T, Lee TSJSYLE. Advanced Biodiesel and Biojet Fuels from Lignocellulosic Biomas. In book: Consequences of Microbial Interactions with Hydrocarbons, Oils, and Lipids: Product Fuels Chem. 2017. https://doi.org/10.1007/978-3-319-50436-0_372.

    Article  Google Scholar 

  80. Yoshino T, Liang Y, Arai D, Maeda Y, Honda T, Muto M, Kakunaka N, Tanaka T. Alkane production by the marine cyanobacterium Synechococcus sp. NKBG15041c possessing the α-olefin biosynthesis pathway. Applied Microbiol Biotechnol. 2015;99(3):1521–9.

    Article  CAS  Google Scholar 

  81. Wise CE, Hsieh CH, Poplin NL, Makris TM. Dioxygen activation by the biofuel-generating cytochrome P450 OleT. ACS Catal. 2018;8(10):9342–52.

    Article  CAS  Google Scholar 

  82. Shobana R, Ranjitha J, Anand M, Mahboob S, Vijayalakshmi S. Chapter 8—microbial production of hydrocarbon and its derivatives using different kinds of microorganisms. In: Gupta VK, Tuohy M, Ramteke P, Nguyen Q, editors. Valorization of biomass to bioproducts. Bhat R: Elsevier; 2023. p. 137–49.

    Chapter  Google Scholar 

  83. Sattayawat P, Yunus IS, Jones PR. Production of fatty acids and derivatives using cyanobacteria. Berlin: Springer, Berlin Heidelberg; 2018. p. 1–25.

    Google Scholar 

  84. Yuzawa S, Mirsiaghi M, Jocic R, Fujii T, Masson F, Benites VT, Baidoo EEK, Sundstrom E, Tanjore D, Pray TR, et al. Short-chain ketone production by engineered polyketide synthases in Streptomyces albus. Nat Commun. 2018;9(1):4569.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Wang W, Lu X. Microbial synthesis of Alka(e)nes. Front Bioeng Biotechnol. 2013. https://doi.org/10.3389/fbioe.2013.00010.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Zhou YJ, Buijs NA, Zhu Z, Qin J, Siewers V, Nielsen J. Production of fatty acid-derived oleochemicals and biofuels by synthetic yeast cell factories. Nat Commun. 2016;7(1):11709.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Akhtar MK, Dandapani H, Thiel K, Jones PR. Microbial production of 1-octanol: a naturally excreted biofuel with diesel-like properties. Metab Eng Commun. 2015;2:1–5.

    Article  PubMed  Google Scholar 

  88. Howard TP, Middelhaufe S, Moore K, Edner C, Kolak DM, Taylor GN, Parker DA, Lee R, Smirnoff N, Aves SJ, et al. Synthesis of customized petroleum-replica fuel molecules by targeted modification of free fatty acid pools in Escherichia coli. Proc Natl Acad Sci. 2013;110(19):7636–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Molloy S. Designer biofuels? Nat Rev Microbiol. 2013;11(6):361–361.

    Article  Google Scholar 

  90. Choi Y-N, Lee JW, Kim JW, Park JM. Acetyl-CoA-derived biofuel and biochemical production in cyanobacteria: a mini review. J Appl Phycol. 2020;32(3):1643–53.

    Article  CAS  Google Scholar 

  91. Arai M, Hayashi Y, Kudo H. Cyanobacterial Enzymes for Bioalkane Production. In: Zhang W, Song X, editors. Synthetic biology of cyanobacteria. Singapore: Springer Singapore; 2018. p. 119–54.

    Chapter  Google Scholar 

  92. Wang J, Zhu K. Microbial production of alka(e)ne biofuels. Curr Opin Biotechnol. 2018;50:11–8.

    Article  CAS  PubMed  Google Scholar 

  93. Choi YJ, Lee SY. Microbial production of short-chain alkanes. Nature. 2013;502(7472):571–4.

    Article  CAS  PubMed  Google Scholar 

  94. Keasling J, Garcia Martin H, Lee TS, Mukhopadhyay A, Singer SW, Sundstrom E. Microbial production of advanced biofuels. Nat Rev Microbiol. 2021;19(11):701–15.

    Article  CAS  PubMed  Google Scholar 

  95. Yan Q, Cordell WT, Jindra MA, Courtney DK, Kuckuk MK, Chen X, Pfleger BF. Metabolic engineering strategies to produce medium-chain oleochemicals via acyl-ACP:CoA transacylase activity. Nat Commun. 2022;13(1):1619.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Wang H, Hart DJ, An Y. Functional metagenomic technologies for the discovery of novel enzymes for biomass degradation and biofuel production. Bio Energ Res. 2019;12(3):457–70.

    Google Scholar 

  97. Liu Z, Wang J, Nielsen J. Yeast synthetic biology advances biofuel production. Curr Opin Microbiol. 2022;65:33–9.

    Article  CAS  PubMed  Google Scholar 

  98. Gibson DG, Young L, Chuang R-Y, Venter JC, Hutchison CA, Smith HO. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat Methods. 2009;6(5):343–5.

    Article  CAS  PubMed  Google Scholar 

  99. Zhang J, Chen Y, Fu L, Guo E, Wang B, Dai L, Si T. Accelerating strain engineering in biofuel research via build and test automation of synthetic biology. Curr Opin Biotechnol. 2021;67:88–98.

    Article  CAS  PubMed  Google Scholar 

  100. Jones AT: Engineering Subcellular Fluorescence for High-Throughput Microscopy Screening in Microbial Biofuels Production. MiCodes: Fluorescent Cell Barcodes, 2013.

  101. Suástegui M, Gao M, Shao Z. Chapter 6—pathway assembly and optimization. In: Eckert CA, Trinh CT, editors. Biotechnology for biofuel production and optimization. Amsterdam: Elsevier; 2016. p. 139–64.

    Chapter  Google Scholar 

  102. Liu H, Zhou P, Qi M, Guo L, Gao C, Hu G, Song W, Wu J, Chen X, Chen J, et al. Enhancing biofuels production by engineering the actin cytoskeleton in Saccharomyces cerevisiae. Nat Commun. 2022;13(1):1886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Afrasiab I, Keawsompong S, Kongsaeree P, Parakulsuksatid P. Formulation of an efficient combinatorial cellulase cocktail by comparative analysis of gibson assembly and NEBuilder HiFi DNA assembly Modus Operandi. Int J Emerg Technol. 2020;11:490–5.

    Google Scholar 

  104. Kahn A, Bayer EA, Moraïs S. Advanced cloning tools for construction of designer cellulosomes. In: Lübeck M, editor. Cellulases: methods and protocols. New York: Springer, New York; 2018. p. 135–51.

    Chapter  Google Scholar 

  105. Liu R, Liang L, Freed EF, Choudhury A, Eckert CA, Gill RT. Engineering regulatory networks for complex phenotypes in E. coli. Nat Commun. 2020;11(1):4050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Zhan T, Chen Q, Zhang C, Bi C, Zhang X. Constructing a novel biosynthetic pathway for the production of glycolate from glycerol in Escherichia coli. ACS Synth Biol. 2020;9(9):2600–9.

    Article  CAS  PubMed  Google Scholar 

  107. Clomburg JM, Gonzalez R. Biofuel production in Escherichia coli: the role of metabolic engineering and synthetic biology. Appl Microbiol Biotechnol. 2010;86(2):419–34.

    Article  CAS  PubMed  Google Scholar 

  108. Yao R, Shimizu K. Recent progress in metabolic engineering for the production of biofuels and biochemicals from renewable sources with particular emphasis on catabolite regulation and its modulation. Process Biochem. 2013;48(9):1409–17.

    Article  CAS  Google Scholar 

  109. Sengupta S, Sahasrabuddhe D, Wangikar PP. Transporter engineering for the development of cyanobacteria as cell factories: a text analytics guided survey. Biotechnol Adv. 2022;54: 107816.

    Article  CAS  PubMed  Google Scholar 

  110. Szczerba H, Dudziak K, Krawczyk M, Targoński Z. A Genomic perspective on the potential of wild-type rumen bacterium enterobacter sp. LU1 as an industrial platform for bio-based succinate production. Int J Mol Sci. 2020;21(14):4835.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Sun W, Jiang B, Zhang Y, Guo J, Zhao D, Pu Z, Bao Y. Enabling the biosynthesis of malic acid in Lactococcus lactis by establishing the reductive TCA pathway and promoter engineering. Biochem Eng J. 2020;161:107645.

    Article  CAS  Google Scholar 

  112. Pfleger BF, Pitera DJ, Smolke CD, Keasling JD. Combinatorial engineering of intergenic regions in operons tunes expression of multiple genes. Nat Biotechnol. 2006;24(8):1027–32.

    Article  CAS  PubMed  Google Scholar 

  113. Liu W, Jiang R. Combinatorial and high-throughput screening approaches for strain engineering. Appl Microbiol Biotechnol. 2015;99(5):2093–104.

    Article  CAS  PubMed  Google Scholar 

  114. You J, Du Y, Pan X, Zhang X, Yang T, Rao Z. Increased production of riboflavin by coordinated expression of multiple genes in operons in Bacillus subtilis. ACS Synth Biol. 2022;11(5):1801–10.

    Article  CAS  PubMed  Google Scholar 

  115. Niu F-X, He X, Wu Y-Q, Liu J-Z. Enhancing production of pinene in Escherichia coli by using a combination of tolerance, evolution, and modular co-culture engineering. Front Microbiol. 2018. https://doi.org/10.3389/fmicb.2018.01623.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Shen Y-P, Fong LS, Yan Z-B. Liu J-ZJBfb: combining directed evolution of pathway enzymes and dynamic pathway regulation using a quorum-sensing circuit to improve the production of 4-hydroxyphenylacetic acid in Escherichia coli. Biotechnol Biofuels. 2019;12(1):1–11.

    Article  Google Scholar 

  117. Wang HH, Isaacs FJ, Carr PA, Sun ZZ, Xu G, Forest CR, Church GM. Programming cells by multiplex genome engineering and accelerated evolution. Nature. 2009;460(7257):894–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Zhu X, Zhao D, Qiu H, Fan F, Man S, Bi C, Zhang X. The CRISPR/Cas9-facilitated multiplex pathway optimization (CFPO) technique and its application to improve the Escherichia coli xylose utilization pathway. Metab Eng. 2017;43:37–45.

    Article  CAS  PubMed  Google Scholar 

  119. Utomo JC, Hodgins CL, Ro D-K. Multiplex genome editing in yeast by CRISPR/Cas9—a potent and agile tool to reconstruct complex metabolic pathways. Front Plant Sci. 2021. https://doi.org/10.3389/fpls.2021.719148.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Zhu Z, Zhou YJ, Kang M-K, Krivoruchko A, Buijs NA, Nielsen J. Enabling the synthesis of medium chain alkanes and 1-alkenes in yeast. Metab Eng. 2017;44:81–8.

    Article  CAS  PubMed  Google Scholar 

  121. Zhou YJ, Hu Y, Zhu Z, Siewers V, Nielsen J. Engineering 1-alkene biosynthesis and secretion by dynamic regulation in yeast. ACS Synth Biol. 2018;7(2):584–90.

    Article  CAS  PubMed  Google Scholar 

  122. Hu Y, Zhu Z, Nielsen J, Siewers V. Engineering Saccharomyces cerevisiae cells for production of fatty acid-derived biofuels and chemicals. Open Biol. 2019;9(5):190049.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Gast V, Sandegren A, Dunås F, Ekblad S, Güler R, Thorén S, Tous Mohedano M, Molin M, Engqvist MKM, Siewers V. Engineering Saccharomyces cerevisiae for the production and secretion of affibody molecules. Microb Cell Fact. 2022;21(1):36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Sharan SK, Thomason LC, Kuznetsov SG, Court DL. Recombineering: a homologous recombination-based method of genetic engineering. Nat Protoc. 2009;4(2):206–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Bernardi B, Wendland J. Homologous recombination: a GRAS yeast genome editing tool. Fermentation. 2020;6(2):57.

    Article  CAS  Google Scholar 

  126. Zhang Y, Muyrers JPP, Testa G, Stewart AF. DNA cloning by homologous recombination in Escherichia coli. Nat Biotechnol. 2000;18(12):1314–7.

    Article  CAS  PubMed  Google Scholar 

  127. Lin Y-Y, Zhao S, Lin X, Zhang T, Li C-X, Luo X-M, Feng J-X. Improvement of cellulase and xylanase production in Penicillium oxalicum under solid-state fermentation by flippase recombination enzyme/ recognition target-mediated genetic engineering of transcription repressors. Biores Technol. 2021;337:125366.

    Article  CAS  Google Scholar 

  128. Hmelo LR, Borlee BR, Almblad H, Love ME, Randall TE, Tseng BS, Lin C, Irie Y, Storek KM, Yang JJ, et al. Precision-engineering the Pseudomonas aeruginosa genome with two-step allelic exchange. Nat Protoc. 2015;10(11):1820–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Jiang W, Zhao X, Gabrieli T, Lou C, Ebenstein Y, Zhu TF. Cas9-assisted targeting of chromosome segments CATCH enables one-step targeted cloning of large gene clusters. Nat Commun. 2015;6(1):8101.

    Article  PubMed  Google Scholar 

  130. Shanmugam S, Ngo H-H, Wu Y-R. Advanced CRISPR/Cas-based genome editing tools for microbial biofuels production: a review. Renewable Energy. 2020;149:1107–19.

    Article  CAS  Google Scholar 

  131. Lakhawat SS, Malik N, Kumar V, Kumar S, Sharma PKJCP, Science P. Implications of CRISPR-Cas9 in developing next generation biofuel: a mini-review. Curr Protein Peptid Sci. 2022;23(9):574–84.

    Article  CAS  Google Scholar 

  132. Lee JH, Won HJ, Hoang Nguyen Tran P, Lee S-m, Kim H-Y, Jung JH. Improving lignocellulosic biofuel production by CRISPR/Cas9-mediated lignin modification in barley. GCB Bioenerg. 2021;13(4):742–52.

    Article  CAS  Google Scholar 

  133. Noman M, Shahid M, Ahmed T, Javed MR, Manzoor N, Mazoor I, Shah AA, Maqsood A, Arshad M. Lignocellulosic biomass and microbial genome engineering for sustainable ethanol production: an overview. In: Arshad M, editor. Sustainable ethanol and climate change: sustainability assessment for ethanol distilleries. Cham: Springer International Publishing; 2021. p. 87–112.

    Chapter  Google Scholar 

  134. Liao B, Chen X, Zhou X, Zhou Y, Shi Y, Ye X, Liao M, Zhou Z, Cheng L, Ren B. Applications of CRISPR/Cas gene-editing technology in yeast and fungi. Arch Microbiol. 2021;204(1):79.

    Article  PubMed  Google Scholar 

  135. Tanwar A, Kumar S. Chapter 15—genome editing of algal species by CRISPR Cas9 for biofuels. In: Singh V, editor. Genome engineering via CRISPR-Cas9 system. Dhar PK: Academic Press; 2020. p. 163–76.

    Chapter  Google Scholar 

  136. Housden BE, Muhar M, Gemberling M, Gersbach CA, Stainier DYR, Seydoux G, Mohr SE, Zuber J, Perrimon N. Loss-of-function genetic tools for animal models: cross-species and cross-platform differences. Nat Rev Genet. 2017;18(1):24–40.

    Article  CAS  PubMed  Google Scholar 

  137. Duprex WP, Fouchier RAM, Imperiale MJ, Lipsitch M, Relman DA. Gain-of-function experiments: time for a real debate. Nat Rev Microbiol. 2015;13(1):58–64.

    Article  CAS  PubMed  Google Scholar 

  138. Mihajlovski K, Buntić A, Milić M, Rajilić-Stojanović M, Dimitrijević-Branković S. From agricultural waste to biofuel: enzymatic potential of a bacterial isolate Streptomyces fulvissimus CKS7 for bioethanol production. Waste Biomass Valoriz. 2021;12(1):165–74.

    Article  CAS  Google Scholar 

  139. Tao W, Yang A, Deng Z, Sun Y. CRISPR/Cas9-based editing of streptomyces for discovery, characterization, and production of natural products. Front Microbiol. 2018. https://doi.org/10.3389/fmicb.2018.01660.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Zhang Y, Yun K, Huang H, Tu R, Hua E, Wang M. Antisense RNA interference-enhanced CRISPR/Cas9 base editing method for improving base editing efficiency in streptomyces lividans 66. ACS Synth Biol. 2021;10(5):1053–63.

    Article  CAS  PubMed  Google Scholar 

  141. Pšeničnik A, Reberšek R, Slemc L, Godec T, Kranjc L, Petković H. Simple and reliable in situ CRISPR-Cas9 nuclease visualization tool is ensuring efficient editing in Streptomyces species. J Microbiol Methods. 2022;200:106545.

    Article  PubMed  Google Scholar 

  142. Tong Y, Whitford CM, Blin K, Jørgensen TS, Weber T, Lee SY. CRISPR–Cas9, CRISPRi and CRISPR-BEST-mediated genetic manipulation in streptomycetes. Nat Protoc. 2020;15(8):2470–502.

    Article  CAS  PubMed  Google Scholar 

  143. Pant S, Ritika NP, Ghati A, Chakraborty D, Maximiano MR, Franco OL, Mandal AK, Kuila A. Employment of the CRISPR/Cas9 system to improve cellulase production in Trichoderma reesei. Biotechnol Adv. 2022;60:108022.

    Article  CAS  PubMed  Google Scholar 

  144. Yang L, Henriksen MM, Hansen RS, Lübeck M, Vang J, Andersen JE, Bille S, Lübeck PS. Metabolic engineering of Aspergillus niger via ribonucleoprotein-based CRISPR–Cas9 system for succinic acid production from renewable biomass. Biotechnol Biofuels. 2020;13(1):206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Chang P-K. A simple CRISPR/Cas9 system for efficiently targeting genes of Aspergillus section <i>Flavi</i> species Aspergillus nidulans, Aspergillus fumigatus, Aspergillus terreus, and Aspergillus niger. Microbiol Spectr. 2023;11(1):e04648-e4622.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Blombach B, Eikmanns BJ. Current knowledge on isobutanol production with Escherichia coli, Bacillus subtilis and Corynebacterium glutamicum. Bioengineered Bugs. 2011;2(6):346–50.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Su H, Lin J, Wang Y, Chen Q, Wang G, Tan F. Engineering Brevibacterium flavum for the production of renewable bioenergy: C4–C5 advanced alcohols. Biotechnol Bioeng. 2017;114(9):1946–58.

    Article  CAS  PubMed  Google Scholar 

  148. Branduardi P, Longo V, Berterame NM, Rossi G, Porro D. A novel pathway to produce butanol and isobutanol in Saccharomyces cerevisiae. Biotechnol Biofuels. 2013;6(1):68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Connor MR, Liao JC. Engineering of an Escherichia coli strain for the production of 3-methyl-1-butanol. Appl Environ Microbiol. 2008;74(18):5769–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Kobayashi S, Atsumi S, Ikebukuro K, Sode K, Asano R. Light-induced production of isobutanol and 3-methyl-1-butanol by metabolically engineered cyanobacteria. Microb Cell Fact. 2022;21(1):7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Park S-H, Kim S, Hahn J-S. Metabolic engineering of Saccharomyces cerevisiae for the production of isobutanol and 3-methyl-1-butanol. Appl Microbiol Biotechnol. 2014;98(21):9139–47.

    Article  CAS  PubMed  Google Scholar 

  152. Wang C, Pfleger BF, Kim S-W. Reassessing Escherichia coli as a cell factory for biofuel production. Curr Opin Biotechnol. 2017;45:92–103.

    Article  CAS  PubMed  Google Scholar 

  153. Kong C-L, Li A-H, Su J, Wang X-C, Chen C-Q, Tao Y-S. Flavor modification of dry red wine from Chinese spine grape by mixed fermentation with Pichia fermentans and S. cerevisiae. LWT. 2019;109:83–92.

    Article  CAS  Google Scholar 

  154. Chen L, Li D, Ren L, Song S, Ma X, Rong Y. Effects of simultaneous and sequential cofermentation of Wickerhamomyces anomalus and Saccharomyces cerevisiae on physicochemical and flavor properties of rice wine. Food Sci Nutr. 2021;9(1):71–86.

    Article  CAS  PubMed  Google Scholar 

  155. Jun Choi Y, Hwan Park J, Yong Kim T, Yup Lee S. Metabolic engineering of Escherichia coli for the production of 1-propanol. Metab Eng. 2012;14(5):477–86.

    Article  CAS  Google Scholar 

  156. Siebert D, Wendisch VF. Metabolic pathway engineering for production of 1,2-propanediol and 1-propanol by Corynebacterium glutamicum. Biotechnol Biofuels. 2015;8(1):91.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Wang Y-P, Sun Z-g, Wei X-Q, Guo X-W, Xiao D-G. Identification of core regulatory genes and metabolic pathways for the n-propanol synthesis in Saccharomyces cerevisiae. J Agric Food Chem. 2021;69(5):1637–46.

    Article  CAS  PubMed  Google Scholar 

  158. Azambuja SPH, Goldbeck R. Butanol production by Saccharomyces cerevisiae: perspectives, strategies and challenges. World J Microbiol Biotechnol. 2020;36(3):48.

    Article  CAS  PubMed  Google Scholar 

  159. Ebrahimian F, Denayer JFM, Karimi K. Efficient coproduction of butanol, ethanol, and biohydrogen from municipal solid waste through a cocultivated biorefinery. Energy Convers Manage. 2022;255:115303.

    Article  CAS  Google Scholar 

  160. Wang X, Li J, Zhang C, Zhang Y, Meng J. Self-assembly of CdS@C. Beijerinckii hybrid system for efficient lignocellulosic butanol production. Chem Eng J. 2021;424:130458.

    Article  CAS  Google Scholar 

  161. Cann AF, Liao JC. Pentanol isomer synthesis in engineered microorganisms. Appl Microbiol Biotechnol. 2010;85(4):893–9.

    Article  CAS  PubMed  Google Scholar 

  162. Duarte WF, Amorim JC, Schwan RF. The effects of co-culturing non-Saccharomyces yeasts with S. cerevisiae on the sugar cane spirit (cachaça) fermentation process. Antonie van Leeuwenhoek. 2013;103(1):175–94.

    Article  CAS  PubMed  Google Scholar 

  163. Ingram LO, Vreeland NS. Differential effects of ethanol and hexanol on the Escherichia coli cell envelope. J Bacteriol. 1980;144(2):481–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Dekishima Y, Lan EI, Shen CR, Cho KM, Liao JC. Extending carbon chain length of 1-butanol pathway for 1-hexanol synthesis from glucose by engineered Escherichia coli. J Am Chem Soc. 2011;133(30):11399–401.

    Article  CAS  PubMed  Google Scholar 

  165. Fisher MA, Boyarskiy S, Yamada MR, Kong N, Bauer S, Tullman-Ercek D. Enhancing tolerance to short-Chain alcohols by engineering the Escherichia coli AcrB efflux pump to secrete the non-native substrate n-butanol. ACS Synth Biol. 2014;3(1):30–40.

    Article  CAS  PubMed  Google Scholar 

  166. Verma S, Ray AK, De BK. Bioconversion of heptanal to heptanol by Saccharomyces cerevisiae. Yeast. 2010;27(5):269–75.

    Article  CAS  PubMed  Google Scholar 

  167. Hamilton-Kemp T, Newman M, Collins R, Elgaali H, Yu K, Archbold D. Production of the long-chain alcohols octanol, decanol, and dodecanol by Escherichia coli. Curr Microbiol. 2005;51(2):82–6.

    Article  CAS  PubMed  Google Scholar 

  168. Hernández Lozada NJ, Simmons TR, Xu K, Jindra MA, Pfleger BF. Production of 1-octanol in Escherichia coli by a high flux thioesterase route. Metab Eng. 2020;61:352–9.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Henritzi S, Fischer M, Grininger M, Oreb M, Boles E. An engineered fatty acid synthase combined with a carboxylic acid reductase enables de novo production of 1-octanol in Saccharomyces cerevisiae. Biotechnol Biofuels. 2018;11(1):150.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Lauer I, Philipps G, Jennewein S. Metabolic engineering of Clostridium ljungdahlii for the production of hexanol and butanol from CO2 and H2. Microb Cell Fact. 2022;21(1):85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Liu P-T, Duan C-Q, Yan G-L. Comparing the effects of different unsaturated fatty acids on fermentation performance of Saccharomyces cerevisiae and aroma compounds during red wine fermentation. Molecules. 2019;24(3):538.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Kataoka N, Vangnai AS, Pongtharangkul T, Yakushi T, Matsushita K. Production of 1,3-diols in Escherichia coli. Biores Technol. 2017;245:1538–41.

    Article  CAS  Google Scholar 

  173. Steen EJ, Chan R, Prasad N, Myers S, Petzold CJ, Redding A, Ouellet M, Keasling JD. Metabolic engineering of Saccharomyces cerevisiae for the production of n-butanol. Microb Cell Fact. 2008;7(1):36.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Huang T, Ma Y. Advances in biosynthesis of higher alcohols in Escherichia coli. World J Microbiol Biotechnol. 2023;39(5):125.

    Article  CAS  PubMed  Google Scholar 

  175. Petrov VV, Okorokov LA. Increase of anion and proton permeability of Saccharomyces carlsbergensis plasmalemma by n-alcohols as a possible cause of its de-energization. Yeast. 1990;6(4):311–8.

    Article  CAS  PubMed  Google Scholar 

  176. Fatma Z, Jawed K, Mattam AJ, Yazdani SS. Identification of long chain specific aldehyde reductase and its use in enhanced fatty alcohol production in E. coli. Metab Eng. 2016;37:35–45.

    Article  CAS  PubMed  Google Scholar 

  177. Zheng Y-N, Li L-L, Liu Q, Yang J-M, Wang X-W, Liu W, Xu X, Liu H, Zhao G, Xian M. Optimization of fatty alcohol biosynthesis pathway for selectively enhanced production of C12/14 and C16/18 fatty alcohols in engineered Escherichia coli. Microb Cell Fact. 2012;11(1):65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Wenning L, Yu T, David F, Nielsen J, Siewers V. Establishing very long-chain fatty alcohol and wax ester biosynthesis in Saccharomyces cerevisiae. Biotechnol Bioeng. 2017;114(5):1025–35.

    Article  CAS  PubMed  Google Scholar 

  179. Fillet S, Adrio JL. Microbial production of fatty alcohols. World J Microbiol Biotechnol. 2016;32(9):152.

    Article  PubMed  Google Scholar 

  180. Jin Z, Wong A, Foo JL, Ng J, Cao Y-X, Chang MW, Yuan Y-J. Engineering Saccharomyces cerevisiae to produce odd chain-length fatty alcohols. Biotechnol Bioeng. 2016;113(4):842–51.

    Article  CAS  PubMed  Google Scholar 

  181. Vivek N, Hazeena SH, Alphy MP, Kumar V, Magdouli S, Sindhu R, Pandey A, Binod P. Recent advances in microbial biosynthesis of C3–C5 diols: genetics and process engineering approaches. Biores Technol. 2021;322:124527.

    Article  CAS  Google Scholar 

  182. Gupta P, Mahajan N, Taneja SC. Recent advances in the stereoselective synthesis of 1,3-diols using biocatalysts. Catal Sci Technol. 2013;3(10):2462–80.

    Article  CAS  Google Scholar 

  183. Yunus IS, Anfelt J, Sporre E, Miao R, Hudson EP, Jones PR. Synthetic metabolic pathways for conversion of CO2 into secreted short-to medium-chain hydrocarbons using cyanobacteria. Metab Eng. 2022;72:14–23.

    Article  CAS  PubMed  Google Scholar 

  184. Song X, Yu H, Zhu K. Improving alkane synthesis in Escherichia coli via metabolic engineering. Appl Microbiol Biotechnol. 2016;100(2):757–67.

    Article  CAS  PubMed  Google Scholar 

  185. Ruffing A: Synechococcus sp. PCC 7002: A Cyanobacterial Chassis for Advanced Biofuel Production. In: Conference: Proposed for presentation at the Bioscience External Review Board held March 4–6, 2014 in Livermore, CA; United States. AC04–94AL85000 2020–11–19: 2014: Medium: ED; Size: 18 p.

  186. Rajneesh SSP, Pathak J, Sinha RP. Cyanobacterial factories for the production of green energy and value-added products: an integrated approach for economic viability. Renew Sustain Energ Rev. 2017;69:578–95.

    Article  CAS  Google Scholar 

  187. Liu Y, Wang C, Yan J, Zhang W, Guan W, Lu X, Li S. Hydrogen peroxide-independent production of α-alkenes by OleTJE P450 fatty acid decarboxylase. Biotechnol Biofuels. 2014;7(1):28.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Zhu T, Scalvenzi T, Sassoon N, Lu X, Gugger M. Terminal olefin profiles and phylogenetic analyses of olefin synthases of diverse cyanobacterial species. Appl Environ Microbiol. 2018;84(13):e00425-e418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Huo YX, Cho KM, Rivera JGL, Monte E, Shen CR, Yan Y, Liao JC. Conversion of proteins into biofuels by engineering nitrogen flux. Nat Biotechnol. 2011;29(4):346–51.

    Article  CAS  PubMed  Google Scholar 

  190. Galazka JM, Tian C, Beeson WT, Martinez B, Glass NL, Cate JH. Cellodextrin transport in yeast for improved biofuel production. Science. 2010;330(6000):84–6.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to express their gratitude to the National Natural Science Foundations of China (Grant Nos. 21502186), National Natural Science Foundation and frontier research program of Chongqing (Grant Nos. cstc2017jcyjA02138), Chongqing Science Committee, and Key laboratory of Degraded and Unused Land Consolidation Engineering, the Ministry of Natural and Resources (Grant No. SXDJ2019) for supporting this work.

Funding

This work was supported by Key laboratory of Degraded and Unused Land Consolidation Engineering, the Ministry of Natural and Resources (Grant No. SXDJ2019) Grant.

Author information

Authors and Affiliations

Authors

Contributions

HFS participated in the conception, design, data collection and analysis, drafted the manuscript, and provided fund support. JFL provided some ideas. All authors read and approved the final manuscript.

Corresponding author

Correspondence to JiaFu Lin.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

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

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

Su, H., Lin, J. Biosynthesis pathways of expanding carbon chains for producing advanced biofuels. Biotechnol Biofuels 16, 109 (2023). https://doi.org/10.1186/s13068-023-02340-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13068-023-02340-0

Keywords