You are on page 1of 11

Bioresource Technology 384 (2023) 129280

Contents lists available at ScienceDirect

Bioresource Technology
journal homepage: www.elsevier.com/locate/biortech

Review

Microbial itaconic acid bioproduction towards sustainable development:


Insights, challenges, and prospects
Priskila Adjani Diankristanti 1, I-Son Ng 2, *
Department of Chemical Engineering, National Cheng Kung University, Tainan 701, Taiwan

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• Biocatalyst improvement is essential for


industrial production of itaconic acid.
• Titers, productivities, and yields vary
widely but improved in the last 10
years.
• Ustilago maydis, aside from A. terreus,
shows potentials as native IA producer.
• In vitro IA production obtained remark­
able yield in shorter reaction time.
• Biomass waste and one-carbon com­
pounds can be an economical produc­
tion pathway.

A R T I C L E I N F O A B S T R A C T

Keywords: Microbial biomanufacturing is a promising approach to produce high-value compounds with low-carbon foot­
Itaconic acid print and significant economic benefits. Among twelve “Top Value-Added Chemicals from Biomass”, itaconic
Aspergillus terreus acid (IA) stands out as a versatile platform chemical with numerous applications. IA is naturally produced by
Cis-aconitic acid decarboxylase
Aspergillus and Ustilago species through a cascade enzymatic reaction between aconitase (EC 4.2.1.3) and cis-
Bioproduction
aconitic acid decarboxylase (EC 4.1.1.6). Recently, non-native hosts such as Escherichia coli, Corynebacterium
Sustainable development goals
glutamicum, Saccharomyces cerevisiae, and Yarrowia lipolytica have been genetically engineered to produce IA
through the introduction of key enzymes. This review provides an up-to-date summary of the progress made in IA
bioproduction, from native to engineered hosts, covers in vivo and in vitro approaches, and highlights the
prospects of combination tactics. Current challenges and recent endeavors are also addressed to envision
comprehensive strategies for renewable IA production in the future towards sustainable development goals
(SDGs).

* Corresponding author.
E-mail address: yswu@mail.ncku.edu.tw (I.-S. Ng).
1
ORCID: 0000-0002-2006-5747.
2
ORCID: 0000-0003-1659-5814.

https://doi.org/10.1016/j.biortech.2023.129280
Received 3 May 2023; Received in revised form 30 May 2023; Accepted 1 June 2023
Available online 7 June 2023
0960-8524/© 2023 Elsevier Ltd. All rights reserved.
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Fig. 1. Metabolic pathway of IA production. Blue color illustrates the pathway existing only in A. terreus, pink color illustrates the pathway existing only in U. maydis,
orange color illustrates the pathway existing only in E. coli, and black color illustrates the pathway existing in all hosts. Glucose as substrate undergoes glycolysis
pathway in cytosol to produce pyruvate and begin the TCA cycle in mitochondria. Other possible substrates, xylose and arabinose, undergo the pentose phosphate
pathway to produce pyruvate and begin the TCA cycle. CAA will be transported back to the cytosol and initiate IA synthesis.

1. Introduction substitute acrylic acid and methacrylic acid. Furthermore, it can be used
as a building block in producing essential polymers, such as resins, fi­
Overdependency on fossil fuels accounts for 85% of global primary bers, and rubber (Okabe et al., 2009), and even applied for medical
energy consumption (Kabeyi and Olanrewaju, 2022), given that they industry and agriculture sector. Widespread application of IA is ranging
currently are the major aspects to produce chemicals, electricity, and from dietary supplement in animal feed (Zhu et al., 2022), drug-delivery
transportation fuels. Shifting to renewable sources is certainly a way to system (Veerabagu et al., 2019), intelligent food packaging (Cottet et al.,
combat climate change, leading to the awakening of renewable chemical 2021), dye removal agent (Gnanasekaran et al., 2019), and slow-release
production. In 2015, the United Nation set up the 17 Sustainable fertilizer system (Bora and Karak, 2022). One of the most valuable de­
Development Goals (SDGs) as a perspective of protecting the planet by rivatives of IA is methyltetrahydrofuran (MTHF), an emerging compo­
interconnecting environment protection, economic growth, and social nent in biofuel synthesis and an eco-friendly solvent for chemical
integration (Olabi et al., 2023). Nevertheless, to completely substitute reactions (Ashokkumar et al., 2022). The market size of IA in 2021 is
fossil-based resources and achieve the goals, it is mandatory to provide 80,000 tons with an estimated price up to $2 per kilogram. Global
an economically viable way to convert renewable resources into high- market value is expected to rise from $96 million to $116.6 million in
titer and high-value chemicals. 2026 (Narisetty et al., 2021), indicating the importance of IA for in­
In 2004, the United States Department of Energy introduced twelve dustrial usage.
promising bio-based building blocks as “Top Value-Added Chemicals IA-producing pathway relies on the conversion of citric acid into the
from Biomass” (Werpy and Petersen, 2004), in which itaconic acid (IA) intermediate cis-aconitic acid (CAA) by aconitase (acn, EC 4.2.1.3) and
is listed. Among all, IA is the only one that provides possibilities to finally into IA by cis-aconitic acid decarboxylase (cadA, EC 4.1.1.6)

2
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

(Klement and Büchs, 2013), as pointed by bold lines in Fig. 1. Two-step (Klement and Büchs, 2013; Zhu et al., 2021). IA production pathway
enzymatic conversion requires right compartmentalization of both en­ involves two key enzymes, aconitase (acn, EC 4.2.1.3) which converts
zymes to obtain high itaconic acid accumulation. Although Aspergillus is citrate into CAA, and cis-aconitic acid decarboxylase (cadA, EC 4.1.16)
known as the most prevalent microbial cell factory for IA production, which transforms CAA into IA. CAA is synthesized in the mitochondria
further optimization of production process remains critical, as it has and transported into the cytosol by mitochondrial tricarboxylate trans­
been reported that fermentation using filamentous fungi undergoes a porter (mttA), where the following decarboxylation step and IA forma­
few drawbacks. Being highly aerobic, Aspergillus demands sufficient tion take place.
oxygen supply for itaconic acid production, which is considered as cost Complete gene cluster of IA bioconversion in A. terreus has been
ineffective (Gopaliya et al., 2021; Narisetty et al., 2023). Therefore, encoded through transcriptome analysis (Li et al., 2011; Deng et al.,
engineered heterologous hosts, such as Escherichia coli (Harder et al., 2020), where cadA is proven to be an essential enzyme in IA biocon­
2018; Harder et al., 2016), Saccharomyces cerevisiae (Blazeck et al., version. Compartmentalization of the key enzymes and intermediates
2014), Corynebacterium glutamicum (Merkel et al., 2022), and Yarrowia plays a vital role as IA bioproduction requires the shuttling of CAA from
lipolytica (Blazeck et al., 2015), are used to solve deficiencies of native mitochondria to cytosol in the presence of mttA, a native mitochondrial
hosts in the view of tight oxygen supply. Overexpression of cadA from transporter in A. terreus. Attempts to maximize the power of mttA have
A. terreus has started to take place ever since its first comprehensive been reported, which the high copy number of mttA in A. terreus showed
identification and characterization (Li et al., 2022; Kanamasa et al., no improvement (Wierckx et al., 2020), while introduction of mttA in
2008), allowing non-native hosts to obtain high level IA through A. niger boosted IA yield significantly (Wang et al., 2022). To elevate the
metabolic engineering and synthetic biology. quality of downstream processing and diminish product inhibition ef­
This review provides a summary of the recent advancements in IA fect, Kreyenschulte et al. (2018) carried out an in situ reactive extrac­
bioconversion, including the utilization of native and engineered hosts. tion, where IA is extracted using trioctylamine as solvent, and glucose is
It also covers the benefits of employing in vivo processes using commonly supplemented to maintain substrate availability. It showed a way to
available substrates (such as glucose or glycerol) and waste materials, as integrate production and recovery steps of IA, yielding up to 105 g/L IA.
well as the application of whole-cell bioconversion of citrate to IA. Concerning further development of IA production in A. terreus as well
Additionally, the review discusses the current challenges and limitations as cost reduction, Huang et al. (2014) overexpressed glucoamylase gene
associated with IA bioconversion, providing clear insight of future from A. niger to allow direct conversion from liquefied starch into IA,
expansion in a comprehensive direction in achieving the renewable preventing gelatinization on high-temperature sterilization and omit­
material toward low carbon emission. ting the saccharification step. Xylose, another alternative sugar, has
been reported to give comparable result to glucose-grown microbial cell
2. In vivo IA production from native hosts: Aspergillus or factories. Magalhães Jr et al. (2020) assessed the use of xylose in
Ustilago? A. terreus NRRL 1960 as an alternative substrate, although the final IA
titer was lower compared to that used by glucose. Aside from sugar
Microbial fermentation using native hosts is the most predominant compounds, other possibilities of substrate such as biomass hydrolysate
way of IA production, initiated by substrate uptake and conversion into (Krull et al., 2017; Gnanasekaran et al., 2019; Narisetty et al., 2021) and
two pyruvate molecules through glycolysis or pentose phosphate agricultural waste (Huang et al., 2014; Bafana and Pandey, 2019; Yang
pathway. One pyruvate molecule undergoes an anaplerotic reaction in et al., 2020) are reported.
the cytosol to replenish the TCA intermediates (oxaloacetate and ma­ In contrast to IA production pathway in A. terreus, IA production in
late), while the other one is further decarboxylated into acetyl-CoA and U. maydis involves the decarboxylation of trans-aconitic acid, the ther­
directly enters TCA cycle to produce citrate, the precursor of IA modynamic isomer of CAA (Kuenz and Krull, 2018; Hosseinpour Tehrani

Table 1
Summary of in vivo IA production in Aspergillus terreus and Ustilago maydis.
Strains Carbon source IA titer (g/ Time Yield (g/ Strategy References
L) (h) g)

A. terreus XH86-8 Liquefied corn 77.6 72 0.55 - Expression of glucoamylase gene under citA promoter Huang et al., 2014
starch - Expression cassette using strong signal peptide
A. terreus DSM WCHa 27.7 146 0.41 - Utilization of WCH Krull et al., 2017
23081 - Detoxification via ion exchanger
A. terreus DSM Glucose 105 115 0.58 - In situ product recovery using trioctylamine as substrate Kreyenschulte et al., 2018
23801 - Glucose supplementation
A. terreus C1 Potato starch 29.7 168 0.2 - Utilization of potato starch waste under optimized physical Bafana and Pandey, 2019
waste properties
A. niveus Glycerol, ABH b 31.55 168 0.45 - Utilization of ABHb and purified glycerol Gnanasekaran et al., 2019
A. niger AB1.13 Glucose 42.7 240 0.26 - Overexpression of acl12 to improve glycolytic flux Hossain et al., 2019
- Nitrogen source feeding
A. terreus NRRL Xylose, glucose 25.96 240 0.35 - Utilization of xylose for glucose-grown biocatalyst Magalhães Jr et al., 2020
1960
A. terreus 2433 Bamboo residue 41.54 240 N/D - Utilization of bamboo residue in fed-batch fermentation Yang et al., 2020
A. terreus BD Glucose, FWH c 41.1 216 0.27 - Utilization of FWHc for itaconic acid production in a Narisetty et al., 2021
thermal-tolerant strain
U. maydis MB215 Glycerol, glucose 220 480 0.33 - Oversxpression of ria1 and AtMttA Hosseinpour Tehrani et al.,
- fuz7 and cyp3 deletion 2019a
- Optimized fermentation and in situ product crystallization
U. maydis MB215 Cellulose, glucose 33.8 480 0.16 - Co-culture with T. reesei RUT C-30, a cellulose producer Schlembach et al., 2020
- By-product reduction through cyp3, MEL, and UA deletion
U. rabenhorstiana Glucose 50.3 360 0.4 - Optimization of physical properties Krull et al., 2020
U. maydis MB215 Glucose 75.7 140 0.66 - By-product reduction through cyp3, MEL, and UA deletion Becker et al., 2021
- fuz7 deletion
- Overexpression of ria1 & AtMttA
a
wheat chaff hydrolysate; b algal biomass hydrolysate; c food waste hydrolysate.

3
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

et al., 2019b). As shown in Fig. 1, CAA shuttling from mitochondria to engineering tools (Wang et al., 2021; Feng et al., 2022). Since cadA is not
cytosol is existed in U. maydis, in the presence of mitochondrial trans­ naturally present in E. coli, overexpression of cadA from A. terreus is an
porter mtt1. This step is essential for IA bioproduction, proven by a essential step in utilizing E. coli to synthesize IA (Kim et al., 2017; Xu and
strong increase of IA titer with the overexpression of mtt1. Mitochondrial Li, 2023). In addition to acn and cadA, IA production also relies on the
transporter mtt1 also facilitates the shuttling of cytosolic oxaloacetate activity of gltA which converts oxaloacetate into citric acid. Vuoristo
into mitochondria, which is closely related to intracellular anaplerotic et al. (2015) obtained 690 mg/L IA through the overexpression of gltA
reaction to replenish TCA intermediates (Wierckx et al., 2020). and acnA from C. glutamicum along with cadA from A. terreus, and
Furthermore, heterologous expression of mitochondrial transporter mttA deletion of pta and ldhA to accumulate pyruvate, which is the precursor
from A. terreus in U. maydis showed increased IA production, with the for IA bioproduction. Utilization of synonymous codon variants (scv) is a
overexpression of the transcriptional regulator gene ria1 and deletion of method to upgrade functional expression level of cadA in E. coli. Jeon
fuz7 and cyp3 (Geiser et al., 2016b). Hosseinpour Tehrani et al. (2019) et al. (2016) screened scv of cadA in the first 10 codons to engineer
integrated optimized fermentation using engineered strain and in situ E. coli. Among all positive clones, one expressed more than 95% of cadA
product crystallization to reduce product inhibition. This approach in soluble form, leading to threefold improvement of IA production
successfully increased the productivity up to three folds, leading to 220 under the condition of nitrogen limitation and glycerol feeding. Ye et al.
g/L IA, the current highest titer achieved through fermentation in native (2022) introduced an unnatural chassis to mimic spatial compartmen­
host. Another strategy worth mentioning to boost IA bioproduction is talization in E. coli, where instead of acn, prpD is employed to convert
blocking the formation of downstream product. In U. maydis, IA is citrate to cis-aconitate. This strategy allowed intermediates release
further being oxidized into (S)-2-hydroxyparaconate by cyp3, a cyto­ without physical barrier, resulting in 5.06 g/L IA from acetate and a
chrome P450 monooxygenase (Geiser et al., 2016a). Deletion of cyp3 is yield of 0.33 g/g.
reported to be beneficial for IA bioproduction, while the overexpression Beside existing as the intermediate product from the conversion of
of it resulted in drastic decrease of IA (Schlembach et al., 2020). Aside citric acid into IA, CAA is also the intermediate between the reversible
from cyp3 deletion, Becker et al. (2021) reported that deletion of the conversion of citrate and isocitrate catalyzed by aconitase (acnB).
whole gene cluster for glycolipid biosynthesis, mannosylerythritol lipids However, the presence of isocitrate dehydrogenase (icd) prevents aco­
(MEL) and ustilagic acid (UA), is beneficial for IA production, as it re­ nitase from converting back into CAA and drags it away from the IA
duces the amount of side reactions and byproducts accumulation. Under production pathway (Chang et al., 2017; Harder et al., 2018). As a result,
this condition, U. maydis produced IA by 1.3-fold higher and increased the irreversible conversion of isocitrate decarboxylates into α-ketoglu­
the production rate by 12%. tarate must be inhibited to conserve CAA. Nevertheless, high IA pro­
Commercial production of IA nowadays is relying on A. terreus as the ductivity might be obtained through icd inhibition, while glutamate
most impressive native producer with high yield of end-product up to auxotrophy is inevitable.
146 g/L (Hevekerl et al., 2014). With A. terreus being the sole host for To minimize trade-off effect between cell growth and IA production,
commercial production of IA, industries are undoubtedly facing several dynamic control of icd is examined, as it allows the cell to grow and
limitations: high initial sugar concentration and oxygen supply, low pH accumulate sufficient biomass for the production phase. Harder et al.
condition, and strictly controlled manganese content (Wu et al., 2017). (2018) applied cI857, a heat-inducible repressor used in the production
Although it was reported that phosphate limitation attenuates the of recombinant protein in E. coli, to construct a temperature-sensitive
inhibitory effect of manganese (Saha and Kennedy, 2019), a manganese- two-stage process. Enabling fast formation of biomass and augmented
tolerant host seems more beneficial. Being capable of producing IA at a productivity, they obtained 47 g/L IA. However, precise temperature
more neutral pH range (4.5 to 6.5) and tolerant to manganese, U. maydis control in a large-scale bioreactor is undoubtedly a hassle, thus it is
offers a better opportunity for IA production, especially in a non-purified necessary to dynamically regulate metabolic flux. YpItcR/Pccl, an IA-
lignocellulose-based fermentation. Low pH might inhibit growth and responsive promoters from from Yersinia pseudotuberculosis, has been
reduce carbon intake, resulting in the drop of biomass accumulation and reported to effectively regulate cellular activity between biomass phase
final IA titer. Schlembach et al. (2020) proposed a consolidated bio­ and production phase without deleting any vital gene for growth (Hanko
process through the co-culture fermentation of U. maydis as the host for et al., 2018). The presence of itaconic acid activated YpItcR/Pccl, thus
IA bioproduction and T. reesei RUT C-30 which converts cellulose repressed the growth phase and further directed the metabolic flux into
feedstock into glucose, to be consumed by U. maydis for IA production. the IA synthesis pathway. Zhao et al. (2022) designed a CRISPRi-
This study has shown a breakthrough by proving the compatibility of mediated self-inducible system (CiMS) using YpItcR/Pccl, which then
U. maydis and T. reesei to undergo an integrated saccharification- was used to regulate the expression of icd, pykA, and sucCD. By utilizing
fermentation process, yielding up to 33.8 g/L IA in 480 h fed-batch CiMS, increase in itaconic acid titer by 23% was observed. A
cultivation. Other Ustilago species, such as U. rabenhorstiana (Krull fluorescence-based biosensor has also been developed using YpItcR/Pccl
et al., 2020), U. vetiveriae (Zambanini et al., 2017), U. cynodontis (Hos­ and is effective to identify the optimum expression level of cadA. This
seinpour Tehrani et al., 2019b) have also been explored as alternatives, system can also detect the presence of mesaconate, cis-, and trans-aco­
reported to obtain more than 50 g/L IA. Taken together, these findings nitate (Hanko et al., 2018).
present solution to overcome drawbacks from A. terreus and expand the Another photocontrol strategy to regulate icd expression level was
prospects of utilizing Ustilago species in large-scale IA production. All reported, where AsLOV2, a light-sensing photoreceptor of Avena sativa is
the results have been summarized in Table 1. Moreover, compared to the inserted into different sites of icd, allowing precise control of TCA cycle
filamentous Aspergillus, the yeast-like morphology of Ustilago is showing flux and redirection into IA-producing pathway (Li et al., 2022b). Out of
advantages for large-scale production, not to mention the insensitivity to other strategies of knocking down or inhibiting icd, the recombinant
impurities. Comprehensive understanding on fungal morphology is Weimberg pathway from Bacillus xenovorans was introduced to over­
essential to fully exploit promising potentials, as fungi undergoes sig­ come glutamate auxotrophy by producing ketoglutarate needed in TCA
nificant change under fluctuation in oxygen content and shearing force cycle. Due to the independency of existing native pathways in E. coli,
(Nemestóthy et al., 2019; Cairns et al., 2019). Weimberg pathway is able supply ketoglutarate using xylose as sub­
strate without competing against IA production pool (Lu et al., 2021).
3. Engineered E. coli for in vivo IA production Two major long-overdue problems in engineered hosts with heter­
ologous protein expression are inclusion body formation due to the low
Known as a superior host for recombinant protein expression, E. coli protein solubility (Vuoristo et al., 2015), and metabolic flux imbalance
stands out as a promising non-native host for IA production, owing to the which leads to low product titer (Cha et al., 2020). Several attempts to
rapid growth, well-characterized genome, and accessible genetic balance cellular pathway flux have been reported, namely modulation of

4
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Table 2
Summary of in vivo IA production in non-native hosts.
Strains Carbon source IA titer Time Yield (g/ Strategy References
(g/L) (h) g)

In vivo
E. coli BW25113(DE3) Glucose 0.69 72 0.06 - Overexpression of AtCadA and CgGltA Vuoristo et al.,
- Deletion of pta and ldhA 2015
E. coli XL1-Blue Glycerol 7.2 90 0.1 - Expression of codon variants (scv) of AtCadA Jeon et al., 2016
- Feed glycerol and citric acid
E. coli Rosetta(DE3) N/D 0.22 24 N/D - Self-assembled reaction of Acn and AtCadA via protein-peptide Yang et al., 2017
interaction
E. coli BW25113 Glycerol 43.0 32 0.6 - Overexpression of gltA, acnB, CgPyc, AtCadA Chang et al.,
- icd deletion 2017
E. coli W3110 Acetate 3.6 88 0.09 - Overexpression of aceA, gltA (ACS pathway) Noh et al., 2018
- iclR deletion (glyoxylate shunt pathway)
E. coli MG1655 Glucose 46.9 120 0.45 - Dynamic regulation of icd expression through heat-inducible Harder et al.,
repressor (cI857) 2018
E. coli XL1-Blue Glucose 6.6 48 0.66 - Construction of synthetic protein scaffold between gltA, acnA, Tran et al., 2019
and AtCadA
E. coli BW25113 Glycerol, xylose 20 100 0.32 - Expression of Weimberg pathway from B. xenovorans Lu et al., 2021
- High aeration and fine-tuning of xylose supply
E. coli BL21(DE3) Glucose 3.3 24 0.33 - Dynamic regulation of icd using AsLOV2, a light-sensitive Li et al., 2022b
biosensor
E. coli MG1655 Glucose 3.9 N/D N/D - Dynamic regulation of icd expression using itaconic acid- Zhao et al., 2022
responding biosensor (YpItcR)
E. coli W3110 Acetate 5.06 80 0.33 - Utilization of prpD for kinetic compartmentalization under Ye et al., 2022
strong promoter
Corynebacterium glutamicum Acetate 29.2 46 0.16 - Overexpression of codon optimized AtCadA Merkel et al.,
ATCC 13032 - Coupled pH and DO control 2022
Halomonas bluephagenesis TD01 Citrate 63.6 36 0.63 - Expression of CadA and Can Zhang et al.,
- Assistance of GroESL 2021
- Downregulation of icd
Methylorubrum extorquens AM1 Methanol 0.03 192 N/D - Overexpression of codon optimized AtCadA Lim et al., 2019
- Impediment of PHB through mutation of phaR
Pichia kudriavzevii Glucose 1.2 24 0.03 - Overexpression of PkMttA and AtCadA Sun et al., 2020
- Downregulation of icd
Pseudomonas putida JE3719 Alkali-treated 1.4 48 0.79 - Dynamic two-stage bioconversion using nitrogen-limiting Elmore et al.,
lignin biosensor cassette 2021
Yarrowia lipolytica Glucose 22.0 460 0.22 - Expression of mitochondrial transporter AtMttA Zhao et al., 2019
Yarrowia lipolytica WCO a 54.6 96 0.3 - Overexpression of AtCadA and POT1 Rong et al., 2022
- icl deletion
Saccharomyces cerevisiae Ethanol 0.14 72 0.01 - Overexpression of AAC2 Xu et al., 2021
- Utilization of ethanol
Saccharomyces cerevisiae Glucose 0.17 72 0.01 - Construction of synthetic hybrid promoter PGPD Blazeck et al.,
2014
Schizosaccharomyces pombe Glucose 1.55 72 0.03 - Overexpression of AtCadA Fujie et al., 2022
- Deletion of idh2, mae1, acl2, mce1
- High cell density culture

enzyme expression level (Noh et al., 2017), directed evolution to in­ through the utilization of acetyl-CoA (Szenk et al., 2017). E. coli owns
crease enzymatic expression level or turnover activities (Lv et al., 2016), the capacity to ingest acetate when there is no other carbon source in the
cell surface display to enhance stability (Guo et al., 2018), fusion tag culture medium (Sakuntala and Kim, 2022). The uptake and conversion
protein (Lin et al., 2022), and protein scaffold mechanism to colocalize of acetate from acetyl-CoA involves either ACK-PTA (acetate kinase-
multiple enzymes (Xu and Li, 2021). Tran et al. (2019) developed a phosphotransacetylase), or ACS (acetyl-CoA synthase) pathway which
synthetic protein scaffold between gltA, acnA, and cadA by channeling is driven by ATP (Li et al., 2016). Nevertheless, acetate significantly
three protein–protein ligands to the C-terminus of each enzyme, which inhibits cell growth despite the potential it holds to achieve economi­
are GBD-gltA, SH3-acnA, and PDZ-cadA. Another innovative strategy cally viable microbial process (Pinhal et al., 2019; Kiefer et al., 2021). To
was proposed by Yang et al. (2017), where acnA and cadA are fused overcome this limitation, E. coli W, an acetate-tolerant strain, is pro­
through the protein-peptide interactional force of PDZ domain and PDZ posed as a host through the activation of ACS pathway along with
ligand, allowing a self-assembly interaction to take place. Chang et al. glyoxylate shunt pathway (Noh et al., 2018). ACS pathway allows cell to
(2017) supplemented citrate upon induction to increase precursor sup­ grow on acetate as the sole carbon source via anaplerosis reaction, while
ply for IA pool and minimize CAA loss from side reactions. Combined glyoxylate shunt pathway enables the conversion of acetyl-coA into
with high aeration and pH maintenance in fed-batch reactor, 43 g/L IA malate and succinate (C4 compounds), which further yield oxaloacetate,
was obtained in 32 h of fermentation using glycerol, reaching a final the precursor of IA pool (Dolan and Welch, 2018). By utilizing this
yield of 0.6 g IA/g glycerol. strategy, Noh et al. (2018) obtained 3.57 g/L IA in 88 h fermentation.
Among all other carbon sources, glucose serves as the most beneficial The aforementioned study also demonstrated excellent acetate tolerance
carbon source in fermentation, which is then dissimilated into pyruvate. and assimilation as shown by 38.7 g/L acetate consumed during culti­
When a substrate dissimilation involves PEP-dependent phosphoryla­ vation. Although the final titer is considered low (5.31 mol IA/mol ac­
tion, such as glucose and glycerol, overexpression of pyruvate carbox­ etate, 0.09 g IA/g acetate), this finding is a proof of acetate feasibility for
ylase from C. glutamicum (CgPyc) is more beneficial than the native PEP- low-cost IA production.
carboxylase from E. coli (EcPpc) (Chang et al., 2017).Acetate is the Employing E. coli as a non-native host is a propitious strategy, owing
predominant product of overflow metabolism in E. coli, generated to the comprehensive genetic background, rapid growth rate, and the

5
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Fig. 2. Scheme of in vitro whole-cell IA bioconversion. Four critical concerns are (1) genetic design, (2) biocatalyst cultivation, (3) bioconversion optimization, and
(4) biocatalyst recycle.

feasibility to overexpress heterologous protein, albeit still limited by the S. cerevisiae has gained increasing attention due to the “fully sequenced
solubility issue. Reflecting from all the aforementioned potentials, in­ genome” as a potential host for IA production, either using glucose or
vestigations are going further to explore other prospects from a wide ethanol as carbon source. Blazeck et al. (2014) constructed a synthetic
range of non-native hosts. Other prospects, such as new biosynthesis hybrid promoter PGPD, which is the modification of the fastest consti­
route, unconventional substrate, or novel hosts, are worth exploring tutive promoter in yeast. Under the control of galactose that enabled
(Table 2). fine-tuning, they obtained 0.17 g/L IA. However, yeast respiration is
substantially inhibited by the presence of glucose in high concentration,
4. Exploration of diverse non-native hosts and various carbon resulting in insufficient ATP for cellular metabolism, a phenomenon
sources known as the Crabtree effect (Malina et al., 2021; Rothman et al., 2021).
Intending to suppress the Crabtree effect, Xu and Li (2021) attempted to
To investigate the prospect of using methanol as carbon source, en­ use ethanol which then is converted into acetyl-CoA, the precursor of
gineering methylotrophic hosts is considerably favorable. Methyloru­ citric acid synthesis. To increase ethanol uptake rate and accelerate
brum sp, which is capable of converting C-1 molecules such as methanol acetyl-CoA synthesis, AAC2, the major mitochondrial ADP/ATP trans­
and formate, was reported as a heterologous host for IA production. Lim porter, was overexpressed. Upregulation of AAC2 reduced acetate
et al. (2019) utilized methanol to produce IA in M. extorquens and accumulation by enhanced final IA titer from 77 mg/L to 142 mg/L.
attempted to regulate carbon flux through phaR mutation, resulting in Yarrowia lipolytica, an oleaginous yeast, has been explored as a great
0.03 g/L IA. Although further study to characterize related gene and industrial platform for its potential to produce lipid (Wang et al., 2020),
enhance final titer is still required, this report shows a comprehensive terpenoids (Ma et al., 2019), and IA (Rong et al., 2022). By over­
possibility of both host and substrate. Alkali-treated lignin was reported expressing cadA from A. terreus and blocking downstream pathway,
by Elmore et al. (2021) as carbon source, along a nitrogen-limiting Rong et al. (2022) reported the production of 54.55 g/L IA in stirred-
biosensor to control nitrogen starvation phase. This strategy was re­ tank reactor. As the compartmentalization of CAA between mitochon­
ported to obtain 1.4 g/L IA in 48 h. Acetate, which severely inhibits cell dria and cytosol plays a crucial role, a mitochondrial transporter mttA
growth in E. coli, was used as the sole carbon sole in IA production using from A. terreus was introduced into Y. lipolytica along with cadA.
C. glutamicum ATCC 13032, where 29.2 g/L IA was achieve in 46 h Augmentation of CAA synthesis greatly affected IA production by
(Merkel et al., 2022). Compared to acetate-based fermentation using providing overflowing supply of precursor, leading to about 60-fold
E. coli which was reported to reach 3.57 g/L IA in 88 h (Noh et al., 2018), increase in final IA titer. Schizosaccharomyces pombe, as known as
C. glutamicum holds a higher potential as a model organism in chemical fission yeast, was reported to be able in producing up to 1.55 g/L IA from
production from acetate, as shown by higher IA titer and acetate glucose (Fujie et al., 2022). Moreover, P. kudriavzevii, as known as
consumption. I. orientalis, was engineered as a host owing to better ability to effectively
Yeast is another alternative for cell factory aside from bacteria, maintain its membrane integrity and higher tolerance to acid. Sun et al.
which owns more robust growth in extreme environment, such as low (2020) overexpressed cadA and mttA from A. terreus in P. kudriavzevii for
pH, low temperature, high oxygen concentration, and high shear stress efficient transport of CAA to cytosol, along with icd deletion, and ob­
(Cheng et al., 2017). Yeast is capable of utilizing a wide range of sub­ tained 1.2 g/L IA from glucose in 24 h with a yield of 29 mg IA/g
strates, varying from sugar such as glucose and glycerol (Zeng et al., glucose.
2020; Bansal et al., 2020), up to other nonfermentable substrates such as Another prospective host comes from alkaliphiles, H. bluephagenesis,
acetate, lactate, and ethanol (Xu and Li, 2021). Among other yeast, which has shown rapid growth in alkaline environment (pH > 8.5) as

6
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Table 3
Summary of in vitro IA production using citrate as substrate.
Strains IA titer (g/L) Time (h) Yield (g/ Strategy References
g)

E. coli Rosetta(DE3) 8.7 30 0.1 - Self-assembled reaction of Acn and AtCadA via protein-peptide Yang et al., 2017
interaction
E. coli BL21(DE3) 41.6 24 0.48 - Increasing AtCadA copy number Kim et al., 2017
E. coli BL21(DE3) 47.5 24 0.55 - Increase AtCadA copy number Moon et al., 2018
- Immobilization using Na-alginate and BaCl2
E. coli BL21(DE3) 67.0 8 0.35 - Overexpression of AtCadA and CgAcnA under dual T7 promoters Hsiang et al., 2022
E. coli Lemo21(DE3) 98.7 10 0.51 - Cold treatment for 24 h Diankristanti et al., 2023
- Integration of GroELS
Shewanella livingstonensis 1.41 1 0.16 - Heat at 45 ◦ C to increase membrane permeability Luo et al., 2020
Ac10

well as in open nonsterile condition. Its high tolerance to organic salt genetic background, and inexpensive culture medium, E. coli is a
including IA is advantageous to diminish negative effect of product in­ preferred host for recombinant protein production, specifically as
hibition. With the overexpression of cadA from A. terreus and acnA from whole-cell biocatalysts in IA production. Expression of acnA from
C. glutamicum, icd deletion, and co-expression of GroESL, Zhang et al. C. glutamicum ATCC 13032 and codon-optimized cadA from A. terreus
(2021) attained 63.6 g/L IA from 500 mM citrate in 36 h, which is the into E. coli BL21(DE3) under dual T7 promoter is reported for further
highest IA titer obtained from a heterologous host (Table 2). Nonethe­ investigations (Kim et al., 2017; Hsiang et al., 2022). The effect of gene
less, in vivo IA production is still holding a few drawbacks, long culti­ copy number was found out to increasing cadA copy number up to three
vation time to reach favorable titer remains a challenge. On the lookout times, IA production was enhanced by 25.7% (Kim et al. 2017). A self-
to shorten fermentation period, in vitro strategy is gaining attraction, by assembly enzymatic cascade was designed through the fusion of cadA
utilizing whole-cell biocatalysts where the bioproduction pathway is with PDZ ligand and acnA with PDZ domain Yang et al. (2017), there­
engineered. fore, the protein-peptide interactional force enables these two enzymes
to co-localize and connect, leading to lower steric hindrance and higher
5. Whole-cell bioconversion for high-level IA production IA titer. Aside from E. coli as whole-cell biocatalysts, Luo et al. (2020)
investigated the potential of Shewanella livingstonensis Ac10 by
In vitro approach, in contrast to in vivo, employs cell post-cultivation expressing acnB from E. coli and cadA from A. terreus under the tac
as biocatalyst carrying related genes to undergo the reaction outside the promoter. This study generates a psychrophile-based simple biocatalyst
cell itself (Honda et al., 2017). Cells are cultivated and maintained in (PSCats), in which desired pathways from mesophilic or thermophilic
suitable medium with sufficient nutrient to ensure growth and metabolic bacteria are overexpressed. By increasing the temperature, intrinsic
activity. After cultivation stage, cells are harvested and exposed to metabolic pathways are inactivated, while the targeted recombinant
desired substrate (in this case, citric acid) to initiate the bioconversion of genes remain (Mojarrad et al., 2021).
IA. Utilization of whole-cell biocatalysts in in vitro production offers There are two major stages of in vitro IA production: the growth when
trouble-free control over physical parameters (i.e., enzyme content, pH, the metabolic flux is used for replication, and the production when
temperature), faster reaction rate, simpler product isolation, and cellular energy is driven into IA-producing pathway. Rich medium such
excellent reusability. Being highly specific by eliminating competing as Terrific Broth (TB) medium (Kim et al., 2017) and Triptic Soy Broth
pathway, whole-cell biocatalysts possess the capability of sidestepping (Luo et al., 2020) are used to elevate biomass. However, it is quite
off-target reactions, thus allowing the theoretical yield to approach inapplicable commercially, thus it is necessary to formulate an economic
100%. On top of that, through metabolic engineering, synthetic pathway culture media without compromising on cell, such as by employing
and heterologous enzymes can be introduced to hosts, enabling high-end lignocellulosic feedstock (Nieder-Heitmann et al., 2018). Utilization of
chemical production from low-cost feedstock (Lin et al., 2017). glycerol instead of glucose is also an attempt to deal with acetate
Numerous value-added chemicals have been produced by using whole- overflow which usually occurs when glucose is being consumed.
cell biocatalysts, namely cadaverine (Ting et al., 2021), gamma- At the end of cultivation, biocatalysts are washed using deionized
aminobutyric acid (GABA) (Xue et al., 2021), putrescine (Yang et al., water or phosphate-buffered saline (PBS) to remove remaining compo­
2022), p-coumaric acid (Effendi et al., 2021), and IA (Diankristanti nents of culture medium. Citric acid, the sole substrate, is added into the
et al., 2023). The scheme of in vitro IA production is illustrated in Fig. 2, cell pellet to let the reaction begin at defined incubation temperature. To
from the design step, biocatalysts production and optimization, and the attain an industrially favored and economically feasible process, opti­
recycling attempts to reach an economically viable system. mizing titer, productivity, and yield of the desired product is indis­
As IA bioconversion is involving two enzymes, enzymatic balance, pensable. Hence, it is favorable to optimize related parameters, namely
efficacy, and effectivity of the cascade reaction must be taken into pH, temperature, reaction time, cell density, and substrate concentra­
consideration when designing the biocatalysts. In the aspect of enzyme tion. Kim et al. (2017) found the optimum pH and temperature for in
kinetics, deciphering kinetic constants of enzyme is essential to under­ vitro IA production are 5.5 and 35 ◦ C, respectively. Hsiang et al. (2022)
stand how quickly the enzyme can convert the substrate and how easily carried out a Taguchi L9 array to further optimize biocatalyst density in
the enzyme can get saturated. Hsiang et al. (2022) reported that cadA, term of OD600 and found that biocatalysts activity increased along with
compared to acnA, has a much higher substrate affinity and lower re­ the increasing cell density from 40 to 90, but it started to diminish when
action rate, implying the conversion of CAA to IA is the rate-limiting the value of OD600 elevated from 90 to 130. This phenomenon is related
step. In view of recombinant protein production, accessing data from to the availability of enzyme-substrate active binding site which allow
enzyme database such as BRENDA (BRaunschweig ENzyme DAtabase) IA bioconversion to take place.
and employing computational codon optimization strategies are essen­ Cell permeability is a factor which influences the rate of substrate
tial to accentuate the structural and functional design, since they enable uptake and product release through the cell membrane (Osire et al.,
deeper understanding of cellular mechanism and phenotype-to- 2021), thus closely affects IA conversion rate and final titer. Surfactant,
genotype correlations. such as hexadecyltrimethylammonium bromide, polyvinyl alcohol, and
Possessing the combination of rapid growth rate, well-studied Tween 80, has been reported to enhance membrane permeability of

7
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Fig. 3. Comparison of IA production in vivo and in vitro, considering three critical factors which are feedstock cost, reaction time, and purification cost.

E. coli thus allow rapid mass transfer. On the other hand, Luo et al. mutants of cadA successfully enhanced IA production by 1.86 folds.
(2020) applied heat treatment at 45 ◦ C for 15 min in prior to the reac­ Owing to the capacity to expand the number of engineered enzyme
tion, boosting IA productivity by 6 folds (Table 3). properties, machine learning might be an invaluable strategy to control
In addition to high level of IA, commercial production also calls for heterologous protein expression and beat the roadblocks. To further
highly stable and reusable whole-cell biocatalysts. A certain level of enhance cellular robustness, modification of global metabolic regulatory
whole-cell stability and reusability can be approached through enzyme in fungi is worth exploring. Microbes often alter its gene expression and
immobilization, which also mitigates product separation process for carbon flux as a respond to non-ideal bioconversion process, leading to a
continuous production. Moon et al. (2018) immobilized engineered failure in IA production pathway. Pomraning et al. (2022) examined
E. coli JY001 with barium-alginate beads and optimized various related laeA gene, putative methyltransferase gene which exists in secondary
conditions, such as components proportion in the beads, permeabiliza­ metabolite clusters, and found out that it could enhance IA production.
tion agent (i.e., CTAB), temperature, and pH. Immobilized biocatalysts Higher yield was observed as a result of increased enzymatic activity in
could maintain initial activity even after four times of repeated used, IA gene cluster, but phosphate limitation led to decreased growth.
while the activity of free cells dramatically decreased after 2 cycles. However, higher activity of secondary metabolites might also lead to
Heat-treated S. livingstonensis could also remained stable in four cycles troublesome purification process, even contamination.
(Luo et al., 2020), confirming an excellent cell membrane integrity In vitro whole-cell bioconversion strives to mitigate the drawbacks
which is a critical parameter in whole-cell bioconversion. It is clear that present in in vivo process, such as competing side reaction, by-product
in vitro bioconversion is an advantage with maximum productivity of formation, complicated product extraction, and intractable down­
9.8 g/L/h (Diankristanti et al., 2023), while in vivo approach using stream process. Nonetheless, in vitro IA production is facing the problem
A. terreus could only reach around 1 g/L/h (Huang et al., 2014). of restricted mass transport due to the impediment from cell membrane.
Xue et al. (2021) demonstrated the effect of cold treatment in acceler­
6. Opportunities, challenges, and perspectives ating substrate uptake rate and productivity through the migration of
glutamate decarboxylase (GadB) in whole-cell E. coli, and further
Microbial cell factories sometimes suffer from low productivity, improving cell reusability up to 10 times of repeated use. Diankristanti
limited native pathways, and denaturation as a result of withstanding et al. (2023) exploited the aforementioned cold treatment to develop a
environmental tensions. Oxygen and pH controls are essential to main­ seeding strategy, where biocatalyst is kept in rich medium for quality
tain growth and ensure stable productivity, as cellular metabolism often maintenance and stored in low temperature for long-term usage, and
leads to acid generation and oxygen depletion. When chemical is pro­ finally obtained 98.7 g/L IA, which is the highest record for IA pro­
duced inside the cell, the production pool competes with growth-related duction when using in vitro method. To maximize the performance of
pathway, leading to a trade-off between growth and yield, even to severe whole-cell biocatalysts, Sakkos et al. (2019) constructed a microbial
cellular imbalance and crashes. exoskeleton consisting of silica and poly-diallyldimethylammonium
Heterologous expression of recombinant genes faces the difficulties chloride. Layer-by-layer nanocoating on cell surface was found to
of protein misfolding and inclusion body formation, that can be over­ elevate catalysis rate up to 5 folds. This finding offers an insight on how
come by utilizing the proper promoter of vector, optimizing codon usage a microbial exoskeleton could protect cell from environmental stresses
of DNA sequence, and controlling the cultural temperature. To upgrade such as extreme heat and cold, osmotic shock, and desiccation, while at
enzymatic properties, protein engineering serves as an approach the same time enhance reactivity. From industrial point of view, in vitro
through the modification of amino acid sequences. Kim et al. (2022) IA production is restricted by the relatively high cost of citric acid, which
performed an activity-enhancing mutation using SCANEER, a web-based is only half of IA market price. Revealing alternative cascade pathways
tool which identifies the evolution of amino acid sequence, to amplify to utilize cheaper substrate in the whole-cell bioconversion is a major
the activity of cadA. Based on the evolutionary information, three focus of future research in this field.

8
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Holding a great lot of potentials for high-end application, commer­ Blazeck, J., Miller, J., Pan, A., Gengler, J., Holden, C., Jamoussi, M., Alper, H.S., 2014.
Metabolic engineering of Saccharomyces cerevisiae for itaconic acid production. Appl.
cial production of IA is still gaining attention. However, the barricade of
Microbiol. Biotechnol. 98 (19), 8155–8164.
purification process is a challenge to conquer. Although in vivo strategy Blazeck, J., Hill, A., Jamoussi, M., Pan, A., Miller, J., Alper, H.S., 2015. Metabolic
requires lower price of feedstock since it allows the utilization of various engineering of Yarrowia lipolytica for itaconic acid production. Metab. Eng. 32,
carbon source, the purification process will definitely cost a pretty 66–73.
Bora, A., Karak, N., 2022. Starch and itaconic acid-based superabsorbent hydrogels for
penny, given that multiple side reactions exist alongside the desired IA agricultural application. Eur. Polym. J. 176, 111430.
pathway. While in vitro is highly dependent on citric acid whose price is Cairns, T.C., Feurstein, C., Zheng, X., Zheng, P., Sun, J., Meyer, V., 2019. A quantitative
relatively high, it offers little to no purification process as the conversion image analysis pipeline for the characterization of filamentous fungal morphologies
as a tool to uncover targets for morphology engineering: a case study using aplD in
from citric acid to IA is the only reaction taking place. The trade-off of IA Aspergillus niger. Biotechnol. Biofuels 12, 1–17.
production through in vivo and in vitro approaches is shown in Fig. 3. All Cha, D., Ha, H.S., Lee, S.K., 2020. Metabolic engineering of Pseudomonas putida for the
untangled considerations provide blunt perspectives when it comes to production of various types of short-chain-length polyhydroxyalkanoates from
levulinic acid. Bioresour. Technol. 309, 123332.
choosing the direction of IA bioproduction from total consideration of Chang, P., Chen, G.S., Chu, H.Y., Lu, K.W., Shen, C.R., 2017. Engineering efficient
feedstock cost, reaction time, and purification cost. production of itaconic acid from diverse substrates in Escherichia coli. J. Biotechnol.
249, 73–81.
Cheng, C., Zhang, M., Xue, C., Bai, F., Zhao, X., 2017. Development of stress tolerant
7. Conclusion Saccharomyces cerevisiae strains by metabolic engineering: New aspects from cell
flocculation and zinc supplementation. J. Biosci. Bioeng. 123 (2), 141–146.
Cottet, C., Salvay, A.G., Peltzer, M.A., Fernández-García, M., 2021. Incorporation of poly
Producing IA in vivo or using whole-cell biocatalyst (in vitro) has its
(Itaconic acid) with quaternized thiazole groups on gelatin-based films for
own pain and gain, either dodging competing side reactions and long antimicrobial-active food packaging. Polymers 13, 200.
cultivation time, or sidestepping barricade of cell wall and mass trans­ Deng, S., Dai, Z., Swita, M., Pomraning, K.R., Hofstad, B., Panisko, E., Baker, S.,
Magnuson, J., 2020. Deletion analysis of the itaconic acid biosynthesis gene cluster
port limitation. Stability and reactivity are vital to meet industrial needs,
components in Aspergillus pseudoterreus ATCC32359. Appl. Microbiol., Biootechnol.
where microbes are expected to work under dynamic conditions, steady 104 (9), 3981–3992.
supply chain, and well-organized recycle set-up. Engineering de novo Diankristanti, P.A., Effendi, S.S.W., Hsiang, C.-C., Ng, I.-S., 2023. High-level itaconic acid
strains through computational modeling and machine learning paves a (IA) production using engineered Escherichia coli Lemo21 (DE3) toward sustainable
biorefinery. Enzyme Microb. Technol. 167, 110231.
way to maximize performance of microorganisms. By tailoring good Dolan, S.K., Welch, M., 2018. The glyoxylate shunt, 60 years on. Annu. Rev. Microbiol.
genetic design and reaction chassis, microbial and low-carbon IA bio­ 72 (1), 309–330.
production is strikingly moving forward to circular economy and Effendi, S.S.W., Xue, C., Tan, S.I., Ng, I.S., 2021. Whole-cell biocatalyst of recombinant
tyrosine ammonia lyase with fusion protein and integrative chaperone in Escherichia
sustainability. coli for high-level p-Coumaric acid production. J. Taiwan Inst. Chem. Eng. 128,
64–72.
Elmore, J.R., Dexter, G.N., Salvachúa, D., Martinez-Baird, J., Hatmaker, E.A.,
CRediT authorship contribution statement Huenemann, J.D., Klingeman, D.M., Peabody, G.L., Peterson, D.J., Singer, C.,
Beckham, G.T., Guss, A.M., 2021. Production of itaconic acid from alkali pretreated
Priskila Adjani Diankristanti: Writing – original draft, Conceptu­ lignin by dynamic two stage bioconversion. Nat. Commun. 12, 1–12.
Feng, J., Li, C., He, H., Xu, S., Wang, X., Chen, K., 2022. Construction of cell factory
alization. I-Son Ng: Conceptualization, Resources, Supervision, Writing through combinatorial metabolic engineering for efficient production of itaconic
– original draft, Writing – review & editing. acid. Microb. Cell Factories. 21 (1), 1–10.
Fujie, N., Ito, M., Kishida, M., Hirata, Y., Kondo, A., Tanaka, T., 2022. Metabolic
engineering of Schizosaccharomyces pombe for itaconic acid production.
J. Biotechnol. 358, 111–117.
Declaration of Competing Interest Geiser, E., Przybilla, S.K., Engel, M., Kleineberg, W., Büttner, L., Sarikaya, E., Hartog, T.
D., Klankermayer, J., Leitner, W., Bölker, M., Blank, L.M., Wierckx, N., 2016a.
Genetic and biochemical insights into the itaconate pathway of Ustilago maydis
The authors declare the following financial interests/personal re­ enable enhanced production. Metab. Eng. 38, 427–435.
lationships which may be considered as potential competing interests: Geiser, E., Przybilla, S.K., Friedrich, A., Buckel, W., Wierckx, N., Blank, L.M., Bölker, M.,
The authors are grateful for the financial support received from the 2016b. Ustilago maydis produces itaconic acid via the unusual intermediate trans-
aconitate. Microb. Biotechnol. 9 (1), 116–126.
Ministry of Science and Technology (MOST 111-2221-E-006-012-MY3
Gnanasekaran, R., Dhandapani, B., Iyyappan, J., 2019. Improved itaconic acid
and MOST 110-2221-E-006-030-MY3) in Taiwan. production by Aspergillus niveus using blended algal biomass hydrolysate and
glycerol as substrates. Bioresour. Technol. 283, 297–302.
Gopaliya, D., Kumar, V., Khare, S.K., 2021. Recent advances in itaconic acid production
Data availability
from microbial cell factories. Biocatal. Agric. Biotechnol. 36, 102130.
Guo, Q., An, Y., Yun, J., Yang, M., Magocha, T.A., Zhu, J., Xue, Y., Qi, Y., Hossain, Z.,
Data will be made available on request. Sun, W., Qi, X., 2018. Enhanced D-tagatose production by spore surface-displayed L-
arabinose isomerase from isolated Lactobacillus brevis PC16 and biotransformation.
Bioresour. Technol. 247, 940–946.
Acknowledgments Hanko, E.K.R., Minton, N.P., Malys, N., 2018. A transcription factor-based biosensor for
detection of itaconic acid. ACS Synth. Biol. 7 (5), 1436–1446.
Harder, B.J., Bettenbrock, K., Klamt, S., 2016. Model-based metabolic engineering
The authors are grateful for the financial support received from the enables high yield itaconic acid production by Escherichia coli. Metab. Eng. 38,
Ministry of Science and Technology (MOST 111-2221-E-006-012-MY3 29–37.
and MOST 110-2221-E-006-030-MY3) in Taiwan. Harder, B.-J., Bettenbrock, K., Klamt, S., 2018. Temperature-dependent dynamic control
of the TCA cycle increases volumetric productivity of itaconic acid production by
Escherichia coli. Biotechnol. Bioeng. 115 (1), 156–164.
References Hevekerl, A., Kuenz, A., Vorlop, K.-D., 2014. Influence of the pH on the itaconic acid
production with Aspergillus terreus. Appl. Microbiol. Biotechnol. 98 (24),
10005–10012.
Ashokkumar, V., Venkatkarthick, R., Jayashree, S., Chuetor, S., Dharmaraj, S.,
Honda, K., Kimura, K., Ninh, P.H., Taniguchi, H., Okano, K., Ohtake, H., 2017. In vitro
Kumar, G., Chen, W.-H., Ngamcharussrivichai, C., 2022. Recent advances in
bioconversion of chitin to pyruvate with thermophilic enzymes. J. Biosci. Bioeng.
lignocellulosic biomass for biofuels and value-added bioproducts-A critical review.
124 (3), 296–301.
Bioresour. Technol. 344, 126195.
Hossain, A.H., Van Gerven, R., Overkamp, K.M., Lübeck, P.S., Taşpınar, H., Türker, M.,
Bafana, R., Sivanesan, S., Pandey, R.A., 2019. Optimization and scale up of itaconic acid
Punt, P.J., 2019. Metabolic engineering with ATP-citrate lyase and nitrogen source
production from potato starch waste in stirred tank bioreactor. Biotechnol. Prog. 35
supplementation improves itaconic acid production in Aspergillus niger. Biotechnol.
(3), e2774.
Biofuels. 12, 1–14.
Bansal, N., Dasgupta, D., Hazra, S., Bhaskar, T., Ray, A., Ghosh, D., 2020. Effect of
Hosseinpour Tehrani, H., Becker, J., Bator, I., Saur, K., Meyer, S., Rodrigues Lóia, A.C.,
utilization of crude glycerol as substrate on fatty acid composition of an oleaginous
Blank, L.M., Wierckx, N., 2019a. Integrated strain-and process design enable
yeast Rhodotorula mucilagenosa IIPL32: Assessment of nutritional indices. Bioresour.
production of 220 g L− 1 itaconic acid with Ustilago maydis. Biotechnol. Biofuels 12,
Technol. 309, 123330.
1–11.
Becker, J., Tehrani, H., Ernst, P., Blank, L.M., Wierckx, N., 2021. An optimized Ustilago
maydis for itaconic acid production at maximal theoretical yield. J. Fungi. 7, 20–35.

9
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Hosseinpour Tehrani, H., Saur, K., Tharmasothirajan, A., Blank, L.M., Wierckx, N., Narisetty, V., Renuka, G., Amulya, K., Brar, K.K., Magdouli, S., Binod, P., Kumar, V.,
2019b. Process engineering of pH tolerant Ustilago cynodontis for efficient itaconic Mohan, S.V., Pandey, A., Sindhu, R., 2023. Biological production of organic acids by
acid production. Microb. Cell Factories 18, 1–12. filamentous fungi. Curr. Dev. Biotechnol. Bioeng. 455–475.
Hsiang, C.-C., Diankristanti, P.A., Tan, S.-I., Ke, Y.-C., Chen, Y.-C., Effendi, S.S.W., Ng, I.- Nemestóthy, N., Bakonyi, P., Komáromy, P., Bélafi-Bakó, K., 2019. Evaluating aeration
S., 2022. Tailoring key enzymes for renewable and high-level itaconic acid and stirring effects to improve itaconic acid production from glucose using
production using genetic Escherichia coli via whole-cell bioconversion. Enzyme Aspergillus terreus. Biotechnol. Lett. 41 (12), 1383–1389.
Microb. Technol. 160, 110087. Nieder-Heitmann, M., Haigh, K.F., Görgens, J.F., 2018. Process design and economic
Huang, X., Chen, M., Lu, X., Li, Y., Li, X., Li, J.J., 2014. Direct production of itaconic acid analysis of a biorefinery co-producing itaconic acid and electricity from sugarcane
from liquefied corn starch by genetically engineered Aspergillus terreus. Microb. Cell bagasse and trash lignocelluloses. Bioresour. Technol. 262, 159–168.
Factories 13, 1–10. Noh, M.H., Lim, H.G., Woo, S.H., Song, J., Jung, G.Y., 2018. Production of itaconic acid
Jeon, H.-G., Cheong, D.-E., Han, Y., Song, J.J., Choi, J.H., 2016. Itaconic acid production from acetate by engineering acid-tolerant Escherichia coli W. Biotechnol. Bioeng. 115
from glycerol using Escherichia coli harboring a random synonymous codon- (3), 729–738.
substituted 5′ -coding region variant of the cadA gene. Biotechnol. Bioeng. 113 (7), Noh, M., Yoo, S.M., Kim, W.J., Lee, S.Y., 2017. Gene expression knockdown by
1504–1510. modulating synthetic small RNA expression in Escherichia coli. Cell Syst. 5 (4),
Kabeyi, M.J.B., Olanrewaju, O.A., Messineo, A., 2022. Biogas production and 418–426.
applications in the sustainable energy transition. J. Energy 2022, 1–43. Okabe, M., Lies, D., Kanamasa, S., Park, E.Y., 2009. Biotechnological production of
Kiefer, D., Merkel, M., Lilge, L., Henkel, M., Hausmann, R., 2021. From acetate to bio- itaconic acid and its biosynthesis in Aspergillus terreus. Appl. Microbiol. Biotechnol.
based products: underexploited potential for industrial biotechnology. Trends 84 (4), 597–606.
Biotechnol. 39 (4), 397–411. Olabi, A.G., Shehata, N., Sayed, E.T., Rodriguez, C., Anyanwu, R.C., Russell, C.,
Kim, D., Noh, M.H., Park, M., Kim, I., Ahn, H., Ye, D.Y., Jung, G.Y., Kim, S., 2022. Abdelkareem, M.A., 2023. Role of microalgae in achieving sustainable development
Enzyme activity engineering based on sequence co-evolution analysis. Metab. Eng. goals and circular economy. Sci. Total Environ. 854, 158689.
74, 49–60. Osire, T., Yang, T., Xu, M., Zhang, X., Long, M., Ngon, N.K.a., Rao, Z., 2021. Integrated
Kim, J., Seo, H.M., Bhatia, S.K., Song, H.S., Kim, J.H., Jeon, J.M., Choi, K.Y., Kim, W., gene engineering synergistically improved substrate-product transport, cofactor
Yoon, J.J., Kim, Y.G., Yang, Y.H., 2017. Production of itaconate by whole-cell generation and gene translation for cadaverine biosynthesis in E. coli. Int. J. Biol.
bioconversion of citrate mediated by expression of multiple cis-aconitate Macromol. 169, 8–17.
decarboxylase (cadA) genes in Escherichia coli. Sci. Rep. 7, 1–9. Pinhal, S., Ropers, D., Geiselmann, J., de Jong, H., Metcalf, W.W., 2019. Acetate
Klement, T., Büchs, J., 2013. Itaconic acid–a biotechnological process in change. metabolism and the inhibition of bacterial growth by acetate. J. Bacteriol. 201 (13).
Bioresour. Technol. 135, 422–431. Pomraning, K.R., Dai, Z., Munoz, N., Kim, Y.-M., Gao, Y., Deng, S., Lemmon, T., Swita, M.
Kreyenschulte, D., Heyman, B., Eggert, A., Maßmann, T., Kalvelage, C., Kossack, R., S., Zucker, J.D., Kim, J., Mondo, S.J., Panisko, E., Burnet, M.C., Webb-Robertson, B.-
Regestein, L., Jupke, A., Büchs, J., 2018. In situ reactive extraction of itaconic acid J., Hofstad, B., Baker, S.E., Burnum-Johnson, K.E., Magnuson, J.K., 2022. Itaconic
during fermentation of Aspergillus terreus. Biochem. Eng. J. 135, 133–141. acid production is regulated by LaeA in Aspergillus pseudoterreus. Metab. Eng. Comm.
Krull, S., Eidt, L., Hevekerl, A., Kuenz, A., Prüße, U., 2017. Itaconic acid production from 15, e00203.
wheat chaff by Aspergillus terreus. Process Biochem. 63, 169–176. Rong, L., Miao, L., Wang, S., Wang, Y., Liu, S., Lu, Z., Zhao, B., Zhang, C., Xiao, D.,
Krull, S., Lünsmann, M., Prüße, U., Kuenz, A., 2020. Ustilago rabenhorstiana—an Pushpanathan, K., Wong, A., Yu, A., 2022. Engineering Yarrowia lipolytica to produce
alternative natural itaconic acid producer. Fermentation. 6, 4. itaconic acid from waste cooking oil. Front. Bioeng. Biotechnol. 10, 888869.
Kuenz, A., Krull, S., 2018. Biotechnological production of itaconic acid: things you have Rothman, D.L., Stearns, S.C., Shulman, R.G., 2021. Gene expression regulates metabolite
to know. Appl. Microbiol. Biotechnol. 102 (9), 3901–3914. homeostasis during the Crabtree effect: Implications for the adaptation and
Li, Q., Lu, J., Zhang, G., Liu, S., Zhou, J., Du, G., Chen, J., 2022. Recent advances in the evolution of Metabolism. Proc. Nat. Acad. Sci. 118 (2) e2014013118.
development of Aspergillus for protein production. Bioresour. Technol. 126768. Saha, B.C., Kennedy, G.J., 2019. Phosphate limitation alleviates the inhibitory effect of
Li, A., Van Luijk, N., Ter Beek, M., Caspers, M., Punt, P., Van der Werf, M., 2011. A clone- manganese on itaconic acid production by Aspergillus terreus. Biocatal. Agri.
based transcriptomics approach for the identification of genes relevant for itaconic Biotechnol. 18, 101016.
acid production in Aspergillus. Fungal Genet. Biol. 48, 602–611. Sakkos, J.K., Wackett, L.P., Aksan, A., 2019. Enhancement of biocatalyst activity and
Li, Y., Zhao, M., Wei, D., Zhang, J., Ren, Y., 2022b. Photocontrol of itaconic acid protection against stressors using a microbial exoskeleton. Sci. Rep. 9, 1–12.
synthesis in Escherichia coli. ACS Synth. Biol. 11 (6), 2080–2088. Sakuntala, M., Kim, J.R., 2022. Recent advances and challenges in the bioconversion of
Lim, C.K., Villada, J.C., Chalifour, A., Duran, M.F., Lu, H., Lee, P.K., 2019. Designing and acetate to value-added chemicals. Bioresour. Technol. 128064.
engineering Methylorubrum extorquens AM1 for itaconic acid production. Front. Schlembach, I., Hosseinpour Tehrani, H., Blank, L.M., Büchs, J., Wierckx, N.,
Microbiol. 10, 1027. Regestein, L., Rosenbaum, M.A., 2020. Consolidated bioprocessing of cellulose to
Lin, L.u., Gong, M., Liu, Y., Li, J., Lv, X., Du, G., Liu, L., 2022. Combinatorial metabolic itaconic acid by a co-culture of Trichoderma reesei and Ustilago maydis. Biotechnol.
engineering of Escherichia coli for de novo production of 2′ -fucosyllactose. Bioresour. Biofuels. 13, 1–18.
Technol. 351, 126949. Sun, W., Vila-Santa, A., Liu, N.a., Prozorov, T., Xie, D., Faria, N.T., Ferreira, F.C., Mira, N.
Lu, K.W., Wang, C.T., Chang, H., Wang, R.S., Shen, C.R., 2021. Overcoming glutamate P., Shao, Z., 2020. Metabolic engineering of an acid-tolerant yeast strain Pichia
auxotrophy in Escherichia coli itaconate overproducer by the Weimberg pathway. kudriavzevii for itaconic acid production. Metab. Eng. Comm. 10, e00124.
Metab. Eng. Comm. 13, e00190. Szenk, M., Dill, K.A., de Graff, A.M.R., 2017. Why do fast-growing bacteria enter
Luo, G., Fujino, M., Nakano, S., Hida, A., Tajima, T., Kato, J., 2020. Accelerating itaconic overflow metabolism? Testing the membrane real estate hypothesis. Cell Syst. 5 (2),
acid production by increasing membrane permeability of whole-cell biocatalyst 95–104.
based on a psychrophilic bacterium Shewanella livingstonensis Ac10. J. Biotechnol. Ting, W.-W., Huang, C.-Y., Wu, P.-Y., Huang, S.-F., Lin, H.-Y., Li, S.-F., Chang, J.-S.,
312, 56–62. Ng, I.-S., 2021. Whole-cell biocatalyst for cadaverine production using stable,
Lv, X., Gu, J., Wang, F., Xie, W., Liu, M., Ye, L., Yu, H., 2016. Combinatorial pathway constitutive and high expression of lysine decarboxylase in recombinant Escherichia
optimization in Escherichia coli by directed co-evolution of rate-limiting enzymes and coli W3110. Enzyme Microb. Technol. 148, 109811.
modular pathway engineering. Biotechnol. Bioeng. 113 (12), 2661–2669. Tran, K.N.T., Somasundaram, S., Eom, G.T., Hong, S.H., 2019. Efficient Itaconic acid
Ma, Y.R., Wang, K.F., Wang, W.J., Ding, Y., Shi, T.Q., Huang, H., Ji, X.J., 2019. Advances production via protein–protein scaffold introduction between GltA, AcnA, and CadA
in the metabolic engineering of Yarrowia lipolytica for the production of terpenoids. in recombinant Escherichia coli. Biotechnol. Prog. 35, e2799.
Bioresour. Technol. 281, 449–456. Veerabagu, U., Jaikumar, G., Fushen, L.u., 2019. A facile synthesis of itaconic acid based
Magalhães, A.I., de Carvalho, J.C., Thoms, J.F., Souza Silva, R., Soccol, C.R., 2020. biodegradable co-polyesters: an in-vitro anticancer evaluation and controlled drug
Second-generation itaconic acid: An alternative product for biorefineries? Bioresour. delivery system. J. Polym. Environ. 27 (12), 2756–2768.
Technol. 308, 123319. Vuoristo, K.S., Mars, A.E., Sangra, J.V., Springer, J., Eggink, G., Sanders, J.P.M.,
Malina, C., Yu, R., Björkeroth, J., Kerkhoven, E.J., Nielsen, J., 2021. Adaptations in Weusthuis, R.A., 2015. Metabolic engineering of itaconate production in Escherichia
metabolism and protein translation give rise to the Crabtree effect in yeast. Proc. coli. Appl. Microbiol. Biotechnol. 99 (1), 221–228.
Natl. Acad. Sci. USA 118 e2112836118. Wang, Y., Guo, Y., Cao, W., Liu, H., 2022. Synergistic effects on itaconic acid production
Merkel, M., Kiefer, D., Schmollack, M., Blombach, B., Lilge, L., Henkel, M., in engineered Aspergillus niger expressing the two distinct biosynthesis clusters from
Hausmann, R., 2022. Acetate-based production of itaconic acid with Corynebacterium Aspergillus terreus and Ustilago maydis. Microb. Cell Factories 21, 1–13.
glutamicum using an integrated pH-coupled feeding control. Bioresour. Tech. 351, Wang, J., Ledesma-Amaro, R., Wei, Y., Ji, B., Ji, X.-J., 2020. Metabolic engineering for
126994. increased lipid accumulation in Yarrowia lipolytica–a review. Bioresour. Technol.
Mojarrad, M., Tajima, T., Hida, A., Kato, J., 2021. Psychrophile-based simple biocatalysts 313, 123707.
for effective coproduction of 3-hydroxypropionic acid and 1, 3-propanediol. Biosci. Wang, J., Ma, W., Wang, X., 2021. Insights into the structure of Escherichia coli outer
Biotech. Biochem. 85 (3), 728–738. membrane as the target for engineering microbial cell factories. Microb. Cell
Moon, Y.-M., Gurav, R., Kim, J., Hong, Y.-G., Bhatia, S.K., Jung, H.-R., Hong, J.-W., Factories 20 (1), 1–17.
Choi, T.R., Yang, S.Y., Park, H.Y., Joo, H.-S., Yang, Y.-H., 2018. Whole-cell Werpy, T., Petersen, G., 2004. Top value added chemicals from biomass: volume I–results
immobilization of engineered Escherichia coli JY001 with barium-alginate for of screening for potential candidates from sugars and synthesis gas.
itaconic acid production. Biotechnol. Bioprocess. Eng. 23 (4), 442–447. Wierckx, N., Agrimi, G., Lübeck, P.S., Steiger, M.G., Mira, N.P., Punt, P.J., 2020.
Narisetty, V., Prabhu, A.A., Al-Jaradah, K., Gopaliya, D., Hossain, A.H., Kumar Khare, S., Metabolic specialization in itaconic acid production: a tale of two fungi. Curr. Opin.
Punt, P.J., Kumar, V., 2021. Microbial itaconic acid production from starchy food Biotechnol. 62, 153–159.
waste by newly isolated thermotolerant Aspergillus terreus strain. Bioresour. Technol. Wu, X., Liu, Q., Deng, Y., Li, J., Chen, X., Gu, Y., Lv, X., Zheng, Z., Jiang, S., Li, X., 2017.
337, 125426. Production of itaconic acid by biotransformation of wheat bran hydrolysate with
Aspergillus terreus CICC40205 mutant. Bioresour. Technol. 241, 25–34.

10
P.A. Diankristanti and I.-S. Ng Bioresource Technology 384 (2023) 129280

Xu, Y., Li, Z., 2021. Utilization of ethanol for itaconic acid biosynthesis by engineered Zambanini, T., Hosseinpour Tehrani, H., Geiser, E., Merker, D., Schleese, S., Krabbe, J.,
Saccharomyces cerevisiae. FEMS Yeast Res. 21, foab043. Buescher, J.M., Meurer, G., Wierckx, N., Blank, L.M., 2017. Efficient itaconic acid
Xu, Y., Li, Z., 2023. Alleviating glucose repression and enhancing respiratory capacity to production from glycerol with Ustilago vetiveriae TZ1. Biotechnol. Biofuels. 10 (1),
increase itaconic acid production. Synth. Syst. Biotechnol. 8 (1), 129–140. 1–15.
Xu, Y., Wu, Y., Lv, X., Sun, G., Zhang, H., Chen, T., Du, G., Li, J., Liu, L., 2021. Design and Zeng, J., Liao, S., Qiu, M., Chen, M., Ye, J., Zeng, J., Wang, A., 2020. Effects of carbon
construction of novel biocatalyst for bioprocessing: recent advances and future sources on the removal of ammonium, nitrite and nitrate nitrogen by the red yeast
outlook. Bioresour. Technol. 332, 125071. Sporidiobolus pararoseus Y1. Bioresour. Technol. 312, 123593.
Xue, C., Yi, Y.C., Ng, I.S., 2021. Migration of glutamate decarboxylase by cold treatment Zhang, J., Jin, B., Hong, K., Lv, Y., Wang, Z., Chen, T., 2021. Cell catalysis of citrate to
on whole-cell biocatalyst triggered activity for 4-aminobutyric acid production in itaconate by engineered Halomonas bluephagenesis. ACS Synth. Biol. 10 (11),
engineering Escherichia coli. Int. J. Biol. Macromol. 190, 113–119. 3017–3027.
Yang, Z., Gao, X., Xie, H., Wang, F., Ren, Y., Wei, D., 2017. Enhanced itaconic acid Zhao, C., Cui, Z., Zhao, X., Zhang, J., Zhang, L., Tian, Y., Qi, Q., Liu, J., 2019. Enhanced
production by self-assembly of two biosynthetic enzymes in Escherichia coli. itaconic acid production in Yarrowia lipolytica via heterologous expression of a
Biotechnol. Bioeng. 114 (2), 457–462. mitochondrial transporter MTT. Appl. Microbiol. Biotechnol. 103 (5), 2181–2192.
Yang, S.-C., Ting, W.-W., Ng, I.-S., 2022. Effective whole cell biotransformation of Zhao, M., Li, Y., Wang, F., Ren, Y., Wei, D., 2022. A CRISPRi mediated self-inducible
arginine to a four-carbon diamine putrescine using engineered Escherichia coli. system for dynamic regulation of TCA cycle and improvement of itaconic acid
Biochem. Eng. J. 185, 108502. production in Escherichia coli. Synth. Syst. Biotechnol. 7 (3), 982–988.
Yang, J., Xu, H., Jiang, J., Zhang, N., Xie, J., Zhao, J., Bu, Q., Wei, M., 2020. Itaconic acid Zhu, X., Guo, Y., Liu, Z., Yang, J., Tang, H., Wang, Y., 2021. Itaconic acid exerts anti-
production from undetoxified enzymatic hydrolysate of bamboo residues using inflammatory and antibacterial effects via promoting pentose phosphate pathway to
Aspergillus terreus. Bioresour. Technol. 307, 123208. produce ROS. Sci. Rep. 11 (1), 1–9.
Ye, D.Y., Noh, M.H., Moon, J.H., Milito, A., Kim, M., Lee, J.W., Yang, J.S., Jung, G.Y., Zhu, X., Zhang, Y., Zhao, Y., Tao, L., Liu, H., Dong, W., Yang, G., Li, L., 2022. Effects of
2022. Kinetic compartmentalization by unnatural reaction for itaconate production. dietary supplementation with itaconic acid on the growth performance, nutrient
Nat. Commun. 13 (1), 5353. digestibility, slaughter variables, blood biochemical parameters, and intestinal
morphology of broiler chickens. Poult. Sci. 101 (4), 101732.

11

You might also like