You are on page 1of 18

Applied Microbiology and Biotechnology (2019) 103:5143–5160

https://doi.org/10.1007/s00253-019-09881-1

MINI-REVIEW

Biosynthetic strategies to produce xylitol: an economical venture


Yirong Xu 1 & Ping Chi 1 & Muhammad Bilal 2 & Hairong Cheng 1

Received: 10 January 2019 / Revised: 26 April 2019 / Accepted: 29 April 2019 / Published online: 17 May 2019
# Springer-Verlag GmbH Germany, part of Springer Nature 2019

Abstract
Xylitol is a natural five-carbon sugar alcohol with potential for use in food and pharmaceutical industries owing to its insulin-
independent metabolic regulation, tooth rehardening, anti-carcinogenic, and anti-inflammatory, as well as osteoporosis and ear
infections preventing activities. Chemical and biosynthetic routes using D-xylose, glucose, or biomass hydrolysate as raw
materials can produce xylitol. Among these methods, microbial production of xylitol has received significant attention due to
its wide substrate availability, easy to operate, and eco-friendly nature, in contrast with high-energy consuming and
environmental-polluting chemical method. Though great advances have been made in recent years for the biosynthesis of xylitol
from xylose, glucose, and biomass hydrolysate, and the yield and productivity of xylitol are substantially improved by metabolic
engineering and optimizing key metabolic pathway parameters, it is still far away from industrial-scale biosynthesis of xylitol. In
contrary, the chemical synthesis of xylitol from xylose remains the dominant route. Economic and highly efficient xylitol
biosynthetic strategies from an abundantly available raw material (i.e., glucose) by engineered microorganisms are on the hard
way to forwarding. However, synthetic biology appears as a novel and promising approach to develop a super yeast strain for
industrial production of xylitol from glucose. After a brief overview of chemical-based xylitol production, we critically analyzed
and comprehensively summarized the major metabolic strategies used for the enhanced biosynthesis of xylitol in this review.
Towards the end, the study is wrapped up with current challenges, concluding remarks, and future prospects for designing an
industrial yeast strain for xylitol biosynthesis from glucose.

Keywords Xylitol . Yarrowia lipolytica . Biosynthetic routes . Metabolic engineering . Synthetic biology

Introduction abandoned plant fermentations (Werpy et al. 2004; Brar et al.


2016). It has been known to organic chemistry at least since
Xylitol is a naturally occurring five-carbon sugar alcohol hav- the 1890s. French and German researchers who succeeded in
ing a similar sweetness as regular sugar, but 40% less caloric. isolating the molecule in the 1930s originally discovered xy-
It is one of the BTop Value-Added^ chemicals from biomass litol. Scientists obviously did not realize the biologic proper-
produced by Department of Energy (DOE), which can utilize ties of xylitol until research started to exploit its insulin-
independent nature after World War II. In 1975, the Finnish
Electronic supplementary material The online version of this article company Finnish Sugar Co. Ltd. commenced producing xyli-
(https://doi.org/10.1007/s00253-019-09881-1) contains supplementary tol on an industrial scale. Several unique properties such as
material, which is available to authorized users. protection from cavities, tooth decay, and demineralization of
enamel render xylitol an interesting candidate for food and
* Muhammad Bilal pharmaceutical industries. Xylitol is perfect for diabetic pa-
bilaluaf@hotmail.com
tients as a good sucrose substitute because of its insulin-
* Hairong Cheng independent metabolic regulation (Mussatto and Roberto
chrqrq@sjtu.edu.cn
2003; Albuquerque et al. 2014). It is used for parenteral nu-
1
State Key Laboratory of Microbial Metabolism, School of Life
trition (PN) in infusion therapy due to its inertness to amino
Sciences and Biotechnology, Shanghai Jiao Tong University, acids. This alternative sweetener can limit obesity, when used
Shanghai 200240, China regularly in the diet (Parajó et al. 1998a, b), and can also
2
School of Life Science and Food Engineering, Huaiyin Institute of promote tooth rehardening and remineralization to prevent
Technology, Huaian 223003, China dental caries when used in chewing gums, toothpaste, and
5144 Appl Microbiol Biotechnol (2019) 103:5143–5160

mouthwashes as anticariogenic products (Franceschin et al. progress (Nigam and Singh 1995; Converti et al. 1999).
2011; Mussatto and Roberto 2003). Xylitol possesses mois- Current manufacture strategy mainly employs chemical
ture retention properties and hence is used in toothpaste. methods to produce xylitol from xylose purified from biomass
Further, it prevents osteoporosis, ear infections, and in- hydrolysate. In order to make biosynthesis method available
flammation in children (Albuquerque et al. 2014). In com- for industrial production, extensive studies are needed to im-
bination with sugar alcohols and amino acids, xylitol has prove the yield of xylitol from a cheaply available material
been used as a stabilizing agent for proteins during their such as glucose. Regulation of tricarboxylic acid (TCA) cycle
extraction from natural membranes, thus avoiding dena- and pentose phosphate pathway or modifying metabolic path-
turation. At 80% weight/volume, it can be used as a sta- ways, including knockout and expression of critical genes in
bilizer for fibrinogen pastes for human plasma (Parajó the artificial glucose-to-xylitol pathway, is highly important to
et al. 1998a). Xylitol has almost equal sweetening power industrialize this method. Identification of novel strains with
to sucrose, but with low caloric content (2.4 cal/g), thus is tolerance to high concentrations of carbon sources (such as
used as an alternative to sucrose (Mohamad et al. 2015). glucose) and the high glucose flux ratio into the pentose phos-
Since the 1960s, it has been increasingly used as an alter- phate pathway could improve the productivity and yield of
native low-energy sweetener and food additive in the food xylitol from glucose. In this review, we summarized the recent
industries (Nigam and Singh 1995; Mohamad et al. 2015). progress in xylitol biosynthesis strategies, ongoing challenges,
Importantly, xylitol does not undergo detrimental Millard and future prospects. After a brief overview of chemical-based
reaction, so efficiently preserving the nutritional signifi- xylitol production, we described the diversity of microorgan-
cance of food proteins during thermal processing isms employed for microbial biosynthesis of xylitol with the
(Soleimani et al. 2006; Mussatto and Roberto 2003). main emphasis on molecular approaches applied at the
Due to the high endothermic heat of solution (34.8 cal/ cellular/molecular level for improving its yield and productiv-
g), xylitol causes a pleasant cooling effect and fresh sen- ity. In addition, current challenges, concluding remarks, and
sation in oral and nasal cavities after consumption; there- future prospects for designing an industrial strain for xylitol
fore, it is widely used as a xylitol-coated pharmaceutical, biosynthesis from glucose are also discussed.
confectionery products, and dietary complement prepara-
tions. The products’ flexibility and texture improving Chemical synthesis of xylitol production
properties along with a flavor (Mussatto and Roberto
2003) enable xylitol application (used solely or in combi- In the 1970s, the commercial-scale chemical process for xyli-
nation with other sugar substitutes) in food industries in tol production was developed in Finland by acid hydrolysis
the manufacturing of confectionery products. reduction of xylan-containing lignocellulosics from
Despite a large number of xylitol advantages, it still has a hemicellulosic vegetal materials (such as birch-wood,
laxative effect, which could cause diarrhea and gas, if corncob, and other rich flora) followed by nickel catalyst hy-
consumed at high doses (Silveira and Jonas 2002; drogenation of D-xylose at 80–140 °C and 8–10 MPa hydro-
Akinterinwa et al. 2008). In the case of extremely high intra- gen pressure (Granström et al. 2007a, b; Wang et al. 2016, b)
venous doses, it leads to high uric acid levels in the blood and (Fig. 1, S1). The high temperature, high pressure, expensive
alters the liver function tests (LFTs) resulting in kidney stones metals, and the use of a toxic catalyst make chemical method
and liver stress. Therefore, the expected daily consumption uneconomical, environmentally unsafe, and energy-intensive
level is ≤ 15 g/day as recommended by the Food and Drug production process (Sohyun et al. 2010). Additionally, the
Administration (FDA) in the USA (US FDA 2008). Like other downstream process also relies on various expensive and un-
sugar alcohols, D-mannitol and D-sorbitol (D-glucitol) are competitive chromatographic or recrystallization approaches
popular, which also have wide applications in the food and to separate and purify D-xylose from hemicellulose-xylan hy-
pharmaceutical industries. drolysates to remove these by-products such as color, proteins,
Naturally, xylitol is found in small amounts in plant fruits, metal ions, and other sugars (Silva et al. 1998). In this context,
microorganisms, animal tissues, and the human body researchers have rekindled their interest in bio-based xylitol
(Washüttl et al. 1973; Pepper and Olinger 1988; Mohamad biosynthesis due to its obvious environmentally friendly and
et al. 2015). Large-scale xylitol is mainly produced chemically easy-to-operate advantages.
by reducing D-xylose via a catalytic chemical reaction at high
pressure and temperature conditions, which is expensive, Biosynthetic methods of xylitol production
complicated, time-laborious, and environmentally unfriendly
(Albuquerque et al. 2014). Considering the huge commercial In view of serious contamination and safety concerns by the
demand of xylitol bioproduction, researchers have devoted chemical reduction of xylose, researchers are encouraged to
widespread attention to microbiological methods that con- utilize alternative methods to produce xylitol by microorgan-
sume less energy and cost during the process and gain much isms such as yeast, bacteria, fungus, and molds. These
Appl Microbiol Biotechnol (2019) 103:5143–5160 5145

Fig. 1 Chemical and


biotechnological routes for the
conversion of xylan-containing
lignocellulosics to xylitol
production

bioconversion methods are energy-efficient and ecologically is oxidized to xylulose by XDH (XYL2, Xyl2p) and incorpo-
justified, thus demonstrating a great socioeconomic potential rated xylulose-5-phosphate by xylulokinase (XK, EC
(Rodrigues et al. 2011). Hallborn et al. (1991) reported the 2.7.1.17) (XylB in E. coli, Xyl3 in P. stipitis) into the pentose
conversion of xylose to xylitol by an engineered phosphate metabolic pathway to ethanol and glycerol
Saccharomyces cerevisiae with a gene encoding the xylose (Aranda-Barradas et al. 2000) (Fig. 2). In bacteria, xylose is
reductase (XR, EC 1.1.1.307) of Pichia stipitis CBS 6054 first isomerized to xylulose by xylose isomerase (XI, EC
(Hallborn et al. 1991). The recombinant S. cerevisiae strain 5.3.1.5) and then reduced to xylulose-5-phosphate by XK into
transformed over 95% of xylose into xylitol, yield much the pentose phosphate pathway (Nigam and Singh 1995).
higher than the natural xylose-utilizing yeasts. In addition, Xylose can also be metabolized to D-xylonate, a versatile
Saccharomyces, Candida, Debaryomyces, Pichia, platform chemical with numerous applications (Toivari et al.
Torulopsis, Kloeckera, Hansenula, Trichosporon, 2012), such as its use as a precursor for 1,2,4-butanetriol, co-
Cryptococcus, Rhodotorula, Monilia, Kluyveromyces, polyamides, polyesters, and hydrogel synthesis in the
E n t e ro b a c t e r, P a c h y s o l e n , l i q u e f a c i e n s , a n d manufacturing of biomass-derived plastics (Niu et al. 2003;
Corynebacterium spp. can also produce xylitol (Hong et al. Toivari et al. 2012). D-Xylose is metabolized to D-xylonate
2014a, b). Depending on a great variety of substrates, several using NAD(P)+ or pyrroloquinoline quinol (PQQ)-dependent
different strategies have been developed and attempted for the xylose dehydrogenases (EC 1.1.1.175) or glucose oxidase
xylitol biosynthesis. (EC 1.1.3.4) (Hardy et al. 1993; Galar and Boiardi 1995). In
some bacteria and archaea, xylose is initially oxidized to D-
Bioconversion of xylose to xylitol xylonolactone with cytoplasmic NAD(P)+-dependent D-
xylose dehydrogenases (Johnsen and Schönheit 2004;
Metabolic pathway of xylose Stephens et al. 2007; Johnsen et al. 2009) and then cleaved
to D-xylonate by lactonase in the cytoplasm. The resulting D-
Xylitol can be produced from D-xylose by fermentation with xylonate is dehydrated to produce 2-keto-3-deoxy-pentanoate,
yeast and fungus. XR (EC 1.1.1.9) and xylitol dehydrogenase which is subsequently cleaved by an aldolase or dehydrated
(XDH, EC 1.1.1.9) are the two key enzymes involved in this an d r edu ce d to α- k e t o g l u t a r a t e t o p y r u v a t e a n d
process. In yeast, filamentous fungi, and other eukaryotes, the glycolaldehyde. Yeast and other fungi also produce D-
reduction of D-xylose to xylitol by XR (XYL1, Xyl1p) is the xylonic acid by D-xylose dehydrogenase (Kiessling et al.
first step in D-xylose metabolism. After reduction, the xylitol 1962; Suzuki and Onishi 1973; Kanauchi and Bamforth
5146 Appl Microbiol Biotechnol (2019) 103:5143–5160

Fig. 2 Metabolic pathway for xylose utilization; microbial xylitol is produced from xylose via either of two pathways: xylitol is directly converted to
xylitol by NAD(P)H-XR, or it is first isomerized to xylulose by XI and then reduced to xylitol by NADH-XDH

2012) or D-glucose oxidase (Chun et al. 2006; Pezzotti and Certain Candida strains exhibit dual co-enzyme specificity to
Therisod 2006) (Fig. 2). These enzymes require pyridine nu- utilize phosphorylated as well as unphosphorylated NAD+ as
cleotide co-factors, i.e., NAD+/NADH or NADP+/NADPH. co-factors. This is particularly advantageous because NADH+
The nicotinamide co-factor of XR is a dual dependent pool is more prevalent in the cell with higher stability as com-
(NADPH and NADH) in some yeasts, and the activity of pared to its phosphorylated form (Banta et al. 2002). P. stipitis
NADPH-XR is the highest, whereas XDH is a predominantly is also reported for xylitol biosynthesis; however, it is more
NAD+-dependent enzyme (Rizzi et al. 1989). studied for ethanol fermentation due to the presence of active
genes in a pathway downstream to xylitol. Kluyveromyces
Bioconversion of xylose to xylitol by yeast marxianus is known to carry out high-temperature fermenta-
tion of both pentoses and hexoses due to its natural xylose
Yeasts are preferred for xylitol fermentation primarily due to assimilation ability (De Albuquerque et al. 2015).
their high pentose assimilation rates, xylitol productivity, and S. cerevisiae naturally uses glucose rather than xylose as a
stable expression levels of XR and XDH. Xylitol production substrate and utilizes xylose only after the depletion of glu-
by both the wild-type and engineered yeasts have been exten- cose. It takes up and assimilates xylose by the hexose trans-
sively studied and reported in the literature. Microbial xylitol porter system, which has much higher affinity to glucose than
is produced from xylose via two steps: xylose is directly con- xylose. In this scenario, genetic engineering is a promising
verted into xylitol by NAD(P)H-XR, and the resultant xylitol approach to develop xylose-fermenting S. cerevisiae to over-
is then dephosphorylated by NAD+ or NADP+-dependent come the abovementioned drawback (Bae et al. 2004).
XDH. In some strains such as C. tropicalis (Ko et al. 2006), Xylose reductase is the key enzyme involved in single-step
S. cerevisiae (Kim et al. 1999), and P. stipitis (Kötter et al. reduction of xylose to xylitol. However, the high-level accu-
1990), xylitol is accumulated in the xylose-assimilation path- mulation of xylitol in broth may be hampered due to low
way. Candida sp. is known to be a potential source of xylitol expression levels of the enzyme. Several engineering ap-
production from xylose as a carbon source. It is demonstrated proaches have been applied to improve native XR expression
that xylitol dehydrogenase-deficient mutant of C. tropicalis levels in the parent strain. XR genes from Candida and Pichia
produced the xylitol yields of 0.97 g/g with a productivity of were integrated into S. cerevisiae and expressed under the
more than 3 g/L/h in shaking flask cultures (Ko et al. 2006). control of constitutive promoters. The high xylitol yield of
The NADH-dependent XR and xylitol dehydrogenase of 0.9–1.0 g/g with improved xylose assimilation rates in the
Candida sp. are usually cloned for heterologous expression. recombinant yeasts was attributed to elevated levels of XR
Appl Microbiol Biotechnol (2019) 103:5143–5160 5147

activities (Bae et al. 2004). Similarly, S. cerevisiae overex- et al. 2001). Therefore, the creation of a suitable expression
pressing endogenous aldose reductase gene presented im- cassette comprising XR and G6PD gene under the control
proved xylitol titer (22.4 g/L) and volumetric productivity of GAL promoter could effectively improve the xylitol titer
(0.28 g/L/h) than the wild-type strain, but the values were by galactose induction.
lower in contrast to heterologous XRs introduced from The knocking out or silence xylitol dehydrogenase gene
Candida or Pichia (Kogje and Ghoshalkar 2016). (xyl2) to hinder the metabolism of xylitol and cloning of xy-
During the yeast metabolic process, ethanol and glycer- lose reductase (xyl1) in microbial strains is an important mo-
ol production regenerate NAD+ for xylitol oxidation by lecular strategy in improving the final yield of xylitol
XDH, whereas acetate production regenerates NADPH (Bruinenberg et al. 1983). Xyl2 genes of C. tropicalis were
for xylose reduction by XR (Mohamad et al. 2015). disrupted to xyl2-disrupted mutant and then xylitol fermenta-
Several factors including pH, temperature, agitation rate, tion was carried out. By optimization of the process parame-
inoculum percentage, inoculum age, oxygen level, oxygen ters and screening suitable co-substrates, the productivity and
transfer rate, and initial xylose concentration can affect the yield of xylitol fermentation by the xyl2-disrupted mutant
biosynthesis of xylitol (Pal et al. 2016). The regeneration were significantly enhanced (Ko et al. 2006). Hong et al.
of co-factors is highly dependent on oxygen level and ox- (2014a, b) knocked out xylitol dehydrogenase gene (xyl2) to
ygen transfer rates (Furlan et al. 1994). Under oxygen- overexpress xylose reductase gene (xyl1) or silenced transfor-
deficient conditions, the imbalanced supply of NAD+ in- mation of D-xylulokinase gene (XSK or xyl3) to reduce the
hibits the activity of XDH leading to high xylitol accumu- expression of the XSK gene at a transcriptional level in
lation. Under high aeration conditions, NADH is oxidized Trichoderma reesei strains, which improved xylitol produc-
to NAD + ; some of the xylitol may be metabolized to tion (Hong et al. 2014a, b). Nevertheless, the xylitol produc-
xylulose due to the high NAD+/NADH ratio, thus resulting tion might be limited due to insufficient co-factor (NADPH)
in a lesser accumulation of xylitol. Moreover, redox imbal- regeneration or transport of xylose (Pal et al. 2013). Likewise,
ance between NADPH-XR and NAD-XDH in the cell is chemical mutagenesis in Candida varieties induced XDH
one of the main reasons for xylitol accumulation (Kötter point mutation resulting in functional loss of the active en-
et al. 1990). An oxidative phase of pentose phosphate path- zyme with xylitol yield improvement by ∼ 1.2-fold (Kumar
way (PPP) has been found the most potent NADPH sup- et al. 2010). An XDH-disrupted mutant of K. marxianus ac-
plier by two key enzymes G6PD and 6-PGD. cumulated improved xylitol concentration compared to its
Overexpression of these enzymes accompanied by an ex- wild-type strain in the presence of glycerol as a co-substrate.
ternal secondary substrate addition was used towards However, xylitol titers of the mutants were significantly lower
in vivo co-factor regeneration (Uppada et al. 2014). High than the recombinant Candida spp. (Zhang et al. 2014, b).
expression of endogenous G6PD increased NADPH sup- Another factor affecting xylose assimilation is the presence
ply in recombinant Saccharomyces BJ3505, resulting in of other carbon sources (mainly glucose), which repressed the
xylitol productivity improvement from 1.6 to 2.0 g/L/h in uptake capacity of enzymes to pentoses (Meinander and
glucose-deficient fed-batch fermentations (Kwon et al. Hahnhägerdal 1997). Repeated batch or fed-batch fermenta-
2006). Downregulation of phosphoglucoisomerase (PGI) tion-based processes are easier and time-saving with cleaning,
gene further increased the glucose flux towards PPP for sterilization, and inoculation and had advantages of maintain-
NADPH regeneration in S. cerevisiae. The recombinant ing glucose at the desired level (Bae et al. 2004). However,
strain with simultaneous overexpression of G6PD and re- Tochampa et al. (2005) developed a xylitol production model
duction in PGI activity led to a 1.9-fold increase in specific by the yeast C. mogii using a genetic algorithm to predict the
xylitol productivity (Oh et al. 2007). S. cerevisiae L2612 product formation rate and yield. In xylose fermentation by
transformed with Pichia XR, cellodextrin transporter, and the yeast C. mogii ATCC18364, biomass yield and volumetric
β-glucosidase gene from N. crassa displayed 85.7% in- productivity were substantially enhanced by an initial glucose/
crease in xylitol production from cellobiose and xylose xylose ratio of 10% as compared to xylose as the sole carbon
mixtures in comparison with the glucose and xylose (Zha source. Wannawilai et al. (2017) reported that furfural and
et al. 2013a, b). Recently, glycerol has been reported to glucose could enhance the conversion of xylose into xylitol
enhance the NADPH supply. Disruption of glycerol kinase by C. magnoliae TISTR 5663. Fig. S2 represents the biosyn-
and co-expression of P. stipitis glycerol dehydrogenase thetic route of xylitol in Yarrowia lipolytica yeast.
genes in C. tropicalis BSXDH-3 under the control of
GAPDH promoter enabled the plentiful supply of Bioconversion of hemicellulose to xylitol by yeast
NADPH co-factor and enhanced xylitol productivity
(Ahmad et al. 2013). Galactose can also facilitate Lignocellulosic biomass is an abundant and readily available
NADPH generation via the Leloir pathway coupled to material in some plants, such as corn cobs (Rivas et al. 2010),
PPP without integrating any additional gene (Govinden barley bran (Cruz et al. 2000), rice straw (Roberto et al. 1994),
5148 Appl Microbiol Biotechnol (2019) 103:5143–5160

aspen wood hemicellulosic hydrolysate (Pandey et al. 2000), If the employed yeast strain has good tolerance to toxic
hybrid poplar wood chips (Dien et al. 1997), corn fiber, and substances, it is not necessary to remove from the hydrolysates
sugarcane bagasse (Rao et al. 2006). Hemicellulose is a major (Peng et al. 2012). Zhang et al. demonstrated that Issatchenkia
component of lignocellulose and is the second most common occidentalis (Lj-3, CCTCC M 2006097) and I. orienalis (S-7,
polysaccharide in nature (Nigam and Singh 1995). CCTCC M 2006098) strains display good activity in
Hemicelluloses are short-branched chain degrading toxic substances, which significantly improves the
heteropolysaccharides with mixed hexosans and pentosans, xylitol productivity and yield (Zhang et al. 2009b). It effec-
especially xylan and arabinan, and are easily hydrolyzed tively reduces the burden of physical and chemical purifica-
(Rao et al. 2006). In order to reduce the xylitol production tion or detoxification processes. Fonseca et al. used the
cost, crude hemicellulose can be used as feedstock for xylitol potential of the yeast I. occidentalis CCTCC M 206097 as a
biosynthesis as a potential alternative to pure xylose (Rao et al. detoxification agent of the hemicellulosic hydrolysate to
2006). remove inhibitor compounds (Fonseca et al. 2011). The yeast
After the hydrolysis of plant hemicellulose by dilute sulfu- can efficiently metabolize furans and phenolic compounds in
ric acid, the resulting hydrolysates contain the presence of hemicellulosic hydrolysates of biomass (Fonseca et al. 2011).
degradation products with inhibitor components, including After nutritional supplementation, the detoxified hydrolysates
acetic acid, inorganic salt, sugar, hydroxybenzaldehyde were used as the source of xylose for xylitol production.
(HBA), syringaldehyde (SGA), vanillin, and In China, approximately 50,000–80,000 tons of waste xy-
hydroxymethylfurfural (HMF), which have significant influ- lose mother liquor (WXML) is produced every year that has
ence on yeast growth and fermentation performance become a substantial industrial concern. Regarding composi-
(Winkelhausen and Kuzmanova 1998; Tsao et al. 2002; Wei tion, WXML contains about 60–75% total sugars and only
et al. 2010). Therefore, these hydrolysates should be xylose accounts for 50–70% (Cheng et al. 2011). However,
pretreated to adsorb inhibiting compounds before the micro- WXML is challenging to recycle due to various sugars and
bial cultivation. The bench-scale process includes the follow- high content of inhibitors. Zhang et al. (2012) produced 100 g/
ing pretreatment methods such as activated carbon treatment L of xylitol from detoxified horticultural waste hemicellulosic
(Carvalho et al. 2010), CaO treatment (Silva et al. 1998), ion- hydrolysates by a new isolate of Candida athensensis SB18.
exchange resins (Palmqvist and Hahnhägerdal 2000), The furan compounds furfural and 5-hydroxymethylfurfural
Ca(OH) 2 (Dien et al. 2000), intracellular acidification (HMF) released from dilute acid hydrolysis were toxic to mi-
(Lohmeier-Vogel et al. 1998), organic solvents (Morita and croorganisms for the subsequent biotransformation. There are
Silva 2000; Palmqvist and Hahnhägerdal 2000), yeast strain two biological detoxification approaches to eliminate such by-
variation (Mancilha and Karim 2010), laccase enzyme product sugars (Fig. 3). The first scheme implicates one
(Cassland and Jönsson 1999), recombinant strains (Dien Bperfect^ strain to transform xylose to xylitol and consume
et al. 1997), overliming (Morita and Silva 2000; Wei et al. all of the by-product sugars under the stress of inhibitors.
2010), and continuous-effect membrane distillation. Another is to employ Bcomplementary^ strains, one of which
Membrane distillation is a separation process based on evap- could either transform xylose to xylitol or consume by-
oration through the pores of the hydrophobic membrane. It is product sugars or detoxify the inhibitors. Among these routes,
widely to concentrate non-volatile sugars and to eliminate the first scheme seems very simple in terms of processing, but
volatile inhibitors from hydrolysates. For example, Chen it is sometimes quite problematic to construct a Bperfect^
et al. (2013) reported that vacuum membrane distillation re- strain. In an earlier study, we proposed a technical route in
moved 98% of furfural and 25% of acetic acid from a ligno- which biodetoxification, biotransformation, and purification
cellulosic hydrolysate resulting in ethanol yield improvement were integrated using Candida maltosa and recombinant
by 17.8%. The recovery rate of glucose from the sugar-acid Bacillus subtilis with a disrupted xylose isomerase gene
mixture was greater than 99.12% and the sulfuric acid was xylA. An elevated titer of 213 g/L xylitol was obtained in
completely removed from the hydrolysate using a medium containing 250 g/L of xylose from WXML in the
continuous-effect membrane distillation process (Li et al. lab-scale fermenter (Cheng et al. 2011). Nevertheless, this
2019). Similarly, an aqueous solution of sulfuric acid was also technology needs further improvements due to its complicated
concentrated by membrane distillation for recycling purposes operation and large equipment investment. Recently, our
(Liu et al. 2016). In conclusion, the continuous-effect mem- group developed a one-pot biotransformation procedure to
brane distillation treatment can be effectively applied for sul- produce xylitol from WXML with C. tropicalis X828 and
furic acid recovery and sugar concentration, so it might im- B. subtilis Bs12, including detoxification, biotransformation,
prove the economic advantage for the bioconversion of hemi- and purification. A high level of pure xylitol (95 g/L) was
cellulose to the xylitol production process. As shown in Fig. 1, achieved from the fermentation medium containing 400 g/L
the inhibitor contents can be minimized through detoxification of WXML at a yield of 0.75 g/g xylose with the simultaneous
by pH alteration (Wei et al. 2010). depletion of by-products including sugars glucose, L-
Appl Microbiol Biotechnol (2019) 103:5143–5160 5149

Fig. 3 Scheme of biotransformation by one perfect strain (a) and (adopted from Wang et al. 2016; an open-access article distributed under
complementary strains (b). S1—main substrate xylose; S2—by-product the terms of the Creative Commons Attribution 4.0 International License
sugars; S3—inhibitors; P—product; I and II—complementary strains (http://creativecommons.org/licenses/by/4.0/))

arabinose, and galactose (Fig. 4) (Wang et al. 2016, b). The bacterium for the industrial production of chemicals due to
newly developed one-pot biotransformation process not only its ease of manipulation, cheaper cultivation medium require-
enhances the application of WXML to produce value-added ments, and higher growth rates and yields on recombinant
chemicals at industrial scale but also provides useful evidence proteins. Because of the presence of endotoxins in E. coli, it
for the efficient biotransformation of other compounds on the is not a suitable host to produce food additives such as xylitol
combination of biotechnological processes (Wang et al. 2016, b). (Suzuki et al. 1999). However, E. coli can be used as the host
of NAD(P)H-dependent co-factor, where central metabolism
Bioconversion of xylose to xylitol by engineered serves as the co-factor regeneration system (Cirino et al.
E. coli 2010). It has been reported that the xylose reductases from
C. boidinii (CbXR) (Bolen and Mccracken 1990; Kang et al.
Although some bacteria possess natural capability to synthe- 2003), C. tenuis (CtXR) (Häcker et al. 1999; Nidetzky et al.
size xylitol, little research efforts have focused on using bac- 2015), P. stipitis (PsXR) (Hallborn et al. 1991), and
teria for xylitol production. E. coli is an ideal engineering S. cerivisiae (ScXR) (Ford and Ellis 2001; Jeong et al.

Fig. 4 Complementary biotransformation of WXML by C. tropicalis glucose fed-batch biotransformation. d HPLC profiles of xylitol produc-
X828 and B. subtilis Bs12 in shake flasks. a, b Batch biotransformation tion (adopted from Wang et al. 2016; an open-access article distributed
and glucose fed-batch biotransformation; the arrow represents the addi- under the terms of the Creative Commons Attribution 4.0 International
tion of 15 g/L glucose at 40 h. c Ratio of strain Bs12 and X828 cells in License (http://creativecommons.org/licenses/by/4.0/))
5150 Appl Microbiol Biotechnol (2019) 103:5143–5160

2010), and xylitol dehydrogenases from Gluconobacter xylulokinase gene (xylB) and replacement with xylulokinase
oxydans (GoXDH) (Sugiyama et al. 2003) and P. stipitis (xyl3) gene from P. stipitis results in drastically enhanced
(PsXDH) (Cirino et al. 2010) were all functional in xylitol titers by engineered E. coli strains during growth on
E. coli to varying extents. Suzuki et al. (1999) expressed xylose (Akinterinwa and Cirino 2009). It is also revealed that
xylose reductase gene of C. tropicalis in E. coli and the xylulokinase activity was similar and showed significantly
successfully converted D-xylose to xylitol when D-xylose more activity on xylitol by XylB compared to Xyl3 when
(50 g/L) and D-glucose (5 g/L) were added to IPTG- examined the effects of plasmid-based expression of Xyl3
induced cells. Notably, a higher concentration of 13.3 g/L versus XylB on growth and xylitol production by engineered
of xylitol accumulated after 20 h of cultivation in shake E. coli strains (Akinterinwa and Cirino 2009).
flask fermentation (Suzuki et al. 1999). Similarly, xylose
reductase gene (xyl1) was cloned from C. tropicalis Inactivating xylB gene by Red recombinase
SCTCC 300249, which has relatively high xylitol produc- technology
tion, and expressed heterologously in E. coli BL21(DE3)
and S. cerevisiae W303-1A (Zhang et al. 2009a). Due to The phage lambda-derived Red recombination system
low catalytic activity and the presence operon governing of (Brecombineering^) is a simple and versatile technique for
prokaryotic cells, some methods are needed to improve the making targeted genetic changes (insertion, deletion, and
expression of XR in E. coli to enhance the productivity and point mutations on chromosomal) in E. coli. In the first ho-
yield of xylitol. These methods are as follows: mologous recombination, the plasmid pKD46 homologous
recombination system was used to replace the xylB gene with
Cyclic AMP receptor protein mutation the chloramphenicol resistance gene. The second homologous
recombination occurred at the flippase recognition target
The cyclic AMP receptor protein (CRP; also known as the (FRT) site by recombinase to eliminate the resistance gene.
catabolite receptor protein) is an allosteric transcription factor XylB gene of E. coli strain was successfully knocked out in
that regulates over 150 genes in Escherichia coli. Upon bind- the xylose disruption pathway and the resulting constructed
ing to cyclic AMP (cAMP), the homodimeric CRP undergoes mutant strain improved the xylitol conversion rate (Datsenko
a conformational change whereby two alpha helices reorient and Wanner 2000; Cirino et al. 2010).
to open a DNA-binding domain, allowing CRP to bind to
DNA and affect transcription (Einav et al. 2018). Cirino Increase the extracellular supply of NADPH
et al. (2010) replaced the native gene encoding CRP with a
mutant CRP (CRP*) by E. coli W3110, which reduced sensi- NADPH is a coenzyme of xylose reductase. Improving the
tivity to cAMP, to overcome glucose repression of xylose activity of xylose reductase by increasing NADPH quantity
transporter expression. An alternative strategy was to express is a useful strategy to produce high levels of xylitol. Chin and
either of the native xylose transporters, D-xylose/proton Cirino (2011) utilized engineered E. coli to produce enhanced
symporter (XylE) or the D-xylose ABC transporter levels of xylitol from glucose- and xylose-mixed fermentation
(XylFGH), under the control of a (CRP-independent) tac pro- medium after expressing NADPH-dependent xylose reduc-
moter growing on glucose (Khankal et al. 2008). tase from C. boidinii (CbXR). In another study, Nidetzky
et al. (2003) constructed a plasmid-encoded gene of xylitol
Heterologous expression of xylulokinase dehydrogenase from the yeast G. mastotermitis and expressed
in E. coli. The ratio of NADPH/NADP in E. coli was deter-
Xylulokinase plays an important role in xylose assimilation by mined by the deuterium-labeling method. Notably, the amount
all natural and engineered microorganisms. In bacteria, D- of NADP was decreased by increasing the amount of NADPH
xylose is first isomerized to xylulose by XI (xylA), which is in E. coli to increase the yield of xylitol (Nidetzky et al. 2003).
then phosphorylated to xylulose-5-phosphate by XK (xylB) in Besides the above strategies, improving the conversion ef-
the pentose phosphate pathway (Nigam and Singh 1995). D- ficiency of xylose by codon optimization and changing differ-
Xylulokinase from Aerobacter aerogenes has revealed to ent promoters or inactivating the genes of the related enzymes
phosphorylate xylitol at a low rate (Simpson 1966). This is an efficient way to improve xylitol titer. For example, xy-
non-specific activity of xylulokinases led to the buildup of lose isomerase (XylA) converts xylose to xylulose, which is
potentially toxic phosphorylated compounds that are not fur- then phosphorylated to X-5-P by xylulokinase (XylB).
ther metabolized. During xylitol production by E. coli, xylitol Akinterinwa and Cirino (2009) reported that CRP and the
phosphate was synthesized by XylB, which is toxic and in- xylose-inducible regulator XylR arrange xylA and xylB genes
hibits the use of xylose as the sole carbon source (Akinterinwa in E. coli in a single operon governed by a promoter.
and Cirino 2009). This toxicity can be relieved by a Subsequent deletion of the xylB gene (encoding xylulokinase)
xylulokinase gene mutation. Inactivation of an E. coli and expression of xylose reductase from C. boidinii (CbXR)
Appl Microbiol Biotechnol (2019) 103:5143–5160 5151

to construct engineering E. coli resulted in efficient xylitol C. guilliermondii xylitol dehydrogenase (XDH) or by the pu-
production from glucose-xylose mixtures (Akinterinwa and rified enzyme in vitro reaction using XDH (Onishi and Suzuki
Cirino 2009). 1969; Mayer et al. 2002). However, compared to the D-xylose
to xylitol reduction, the low productivity and yield of this
Bioconversion of xylose to xylitol by other method restrict its application to an industrial scale
engineering bacteria (Granström et al. 2007c). In addition, the low activity of
XDH limits the ultimate production of xylitol. Sugiyama
Besides yeasts, a few bacteria are also capable of producing and coworkers cloned the NAD-dependent XDH gene from
xylitol, but with a lower yield as compared with the yeasts. In G. oxydans and expressed it in G. oxydans to increase XDH
bacteria, D-xylose is converted to D-xylulose by xylose isom- activity and improve xylitol yield by 100%. Moreover, this
erase in a single step; however, the complete transformation of engineered microorganism reduces the three-step process into
D-xylose to xylitol is challenging due to its inhibiting effect on two steps. Unfortunately, the separation process and multi-
xylose isomerase. M. smegmatis (Izumori and Tuzaki 1988) fermentation process improvement was still insufficient and
and G. oxydans (Suzuki et al. 2002) have also been document- unsatisfactory (Sugiyama et al. 2003; Povelainen and
ed to produce xylitol in small amounts. Izumorim and Tuzaki Miasnikov 2007).
reported that immobilized D-xylose isomerase enzyme and
immobilized M. smegmatis with D-xylose as a substrate led Glucose to xylitol bioconversion by the one-step
to 80% xylitol yield. Moreover, the media needed D-xylulose fermentation process
for xylitol production to occur (Izumori and Tuzaki 1988).
Rangaswamy and Agblevor (2002) screened 17 cultures of In order to reduce the three-step fermentation process, numer-
facultative bacteria and found that Corynebacterium sp. pro- ous research efforts have been made to construct engineered
duced the highest amount of xylitol (maximum yield 0.57 g/ microorganisms that directly convert glucose to xylitol. The
g), with an initial xylose concentration of 75 g/L within 24 h of osmotolerant yeast species Debaryomyces hansenii (Jovall
cultivation. Yoshitake et al. (2014) screened Enterobacter et al. 1990), S. mellis (Weimberg 1962), Zygosaccharomyces
strain and showed that Enterobacter strain no. 553 accumu- rouxii (Saha et al. 2007), and S. rouxii (Ingram and Wood
lated 0.35 g/L/h xylitol by an NADPH-dependent XR using 1965) possess potential ability to convert D-glucose to D-
D-xylose as a sole substrate (Yoshitake et al. 2014). It is worth ribulose-5-PO4 via the pentose phosphate pathway (PPP)
mentioning that the enzymatic conversion is not confined to followed by dephosphorylating to D-ribulose. D-Ribulose
fungi and yeasts. B. subtilis, a safe and aerobe soil bacterium, can be reduced to D-arabitol by an NADP-dependent pentitol
can metabolize various sugars, such as xylose, L-arabinose, dehydrogenase or by a second pathway that reduces D-
glucose, and galactose (Cheng et al. 2011). Table 1 summa- ribulose-5-P to D-arabitol-5-P and dephosphorylates to D-
rizes the production of xylitol from xylose by different arabitol (Wong et al. 1995). In addition, the marine fungus
engineered microorganisms. Dendryphiella salina (Lowe and Jennings 1985) and some
other strains of the yeast S. rouxii (strain P3a) (Blakley and
Bioconversion of glucose to xylitol Spencer 1962) can convert D-glucose to D-xylulose-5-PO4
via PPP and then dephosphorylate to D-xylulose. The
Glucose to xylitol bioconversion by a three-step process resulting D-xylulose is reduced to D-arabitol by a NAD-
dependent pentitol dehydrogenase or D-xylulose-5-P dephos-
The process for xylitol production from D-xylose is econom- phorylates to D-arabitol after D-xylulose-5-P reduction to D-
ically not viable because of the high cost of the raw material arabitol-1-P (Wong et al. 1995). D-Arabitol could be oxidized
xylose (approximately US $2500/t) (Cheng et al. 2014), to D-xylulose by xylulose-forming dehydrogenase, followed
which limits the range of current applications in food and by the reduction of D-xylulose to xylitol with xylitol dehydro-
chemical technology. In contrast, D-glucose is an efficient, genase (Povelainen and Miasnikov 2007; Cheng et al. 2014).
readily available, and much cheaper substrate for large-scale Arabitol-5-P could be transformed to D-xylulose-5-P by a
xylitol production. In the late 1960s, Onishi and Suzuki have bacterial D-arabitol-phosphate dehydrogenase and then de-
produced xylitol from D-glucose, by a three-step fermentation phosphorylate to xylitol-5-P by XPDH. Further, xylitol-5-P
process as follows. At first, D-glucose is converted into D- was dephosphorylated to xylitol (Povelainen and Miasnikov
arabitol by some osmophilia yeast strains, such as Pichia, 2007). Figure 5 illustrates the three possible xylitol biosyn-
Zygosaccharomyces, Hansenula, Saccharomyces, Candida, thetic pathways from glucose that have been described in
and Debaryomyces (Escalante et al. 1990). In the second step, engineered yeast cells.
D-arabitol was oxidized to D-xylulose by Acetobacter Since D-arabitol is the key intermediate product in the met-
suboxydans-derived D-arabitol dehydrogenase (ArDH), abolic process, increasing its titer by the optimization of the
which is subsequently reduced to xylitol by microbial transformation of D-glucose into D-arabitol is
5152 Appl Microbiol Biotechnol (2019) 103:5143–5160

Table 1 Production of xylitol from xylose by engineered microorganisms

Microorganism Strain descriptions Initial xylose Xylitol Volumetric Yield Reference


concentration (g/L) titer (g/L) productivity (g/L/h) (g/g)

Yeast
Candida intermedia – 20 45.7 0.38 0.57 Wu et al. (2018)
FL023
Candida tropicalis Metabolize glucose and 400 g/L (waste xylose 95 – 0.75 Wang et al.
X828 inhibitors furfural and mother liquor) (2016)
hydroxymethylfurfural
Depletion of by-product sugars
Kluyveromyces Overexpression of transporter 152 150 3.40 0.99 Zhang et al.
marxianus genes KmFPS1, CiGXF1, (2015)
and CiGXS1
Saccharomyces Cloned CDT-1, gh1-1, and 20 19.2 19.2 1.0 Zha et al. (2013a,
cerevisiae XYL1 genes b)
Kluyveromyces Overexpression 100 71.3 4.43 1.01 Zhang et al.
marxianus NcXR gene, Δxyl1 (2014)
Kluyveromyces Expression of XR gene 40 23.4 0.325 0.70 Zhang et al.
marxianus Substitution of Kmxyl1 (2013)
with XR
Candida tropicalis Expressed of xylose 50 50 1.14 1.0 Jeon et al. (2013)
transporter gene
Candida tropicalis Codon optimization of 50 48 1.44 0.59 Jeon et al. (2012)
NcXR gene
Fungi
Ashbya gossypii Overexpression of endogenous 80 22.6 0.086 0.28 Díaz-Fernández
xylose utilization pathway et al. (2017)
Mutant Coniochaeta Two third silencing of XR 3.0 0.002 – 0.59 Nichols and Saha
ligniaria C8100 and XDH (2016)
Trichoderma reesei Overexpressed xyl1 gene 25 3.72 0.026 0.148 Hong et al.
Silenced xyiH gene (2014a, b)
Bacteria
Escherichia coli Co-expression of xylose reductase 250 161.6 8.0 Jin et al. (2018)
and glucose dehydrogenase
Corynebacterium araE integration, ΔldhA, CtXR, 200 166 7.9 0.83 Sasaki et al.
glutamicum ΔptsF, and ΔxylB (2010)
Corynebacterium Overexpressed XYL1 35 30.1 0.77 0.96 Kim et al. (2010)
glutamicum
Lactococcus lactis Expression of xylose reductase 160 70 1.75 0.43 Nyyssölä et al.
and xylose transporter (2005)

considered an important strategy for enhanced xylitol biosyn- capabilities to transform D-arabitol into D-xylulose due to
thesis. For instance, Zhu et al. (2010) described a process for the lack of D-arabitol-4-dehydrogenase enzyme. Moreover,
81.2 g/L D-arabitol production by K. ohmeri NH-9 in 72 h the XDH of G. oxydans expressed in P. pastoris showed
with productivity and yield of 1.278 g/L h and 0.406 g strong xylitol dehydrogenase activity and high efficiency in
arabitol/g glucose, respectively. Nozaki et al. (2003) achieved reducing D-xylulose to xylitol (Cheng et al. 2014). Harkki
the highest production of 81.4 g/L with a corresponding yield group constructed an engineered Z. rouxii strain, which could
and productivity of 0.078 g xylitol/g glucose and 0.701 g D- tolerate high concentration of glucose (400 g/L), by express-
arabitol/L h, respectively, by Metschnikowia reukaufii after ing the D-arabitol dehydrogenase gene from Klebsiella
116 h of fermentation duration. However, a wide variety of terrigena and xylitol dehydrogenase gene from P. stipitis to
by-product (polyhydric alcohols) was generated and compet- produce a high level of xylitol from glucose. The resulting
ed with the production, when certain osmophilic yeasts are recombinant strain produced about 15 g/L xylitol from
grown in the presence of a high concentration of glucose. 400 g/L glucose in 24 h with yield and productivity of
This accumulation of by-products can be minimized under 0.038 g xylitol/g glucose and 0.063 g/L h, respectively
controlled temperature and D-glucose feeding environment (Harkki et al. 2004). Recently, our laboratory exploited an
(Meng et al. 2014). Some osmophilic yeasts have no engineering P. pastoris for the conversion of glucose to xylitol
Appl Microbiol Biotechnol (2019) 103:5143–5160 5153

Fig. 5 The three metabolic pathway for the conversion of glucose to arabitol 1-P dehydrogenase; E5 and E13, presumptive intracellular (or
xylitol in engineered yeasts. PTS phosphotransferase system, P the membrane-associated) sugar-phosphate phosphatase; E6, xylulokinase;
phosphate moiety, PPP pentose phosphate pathway. P1 and P2, the first E7, NAD/NADP-dependent ketose reductase or NADH/NADPH-
pathway of conversion of glucose to arabitol (Wong et al. 1995); P3 and dependent polyol dehydrogenase; E8, NADP-dependent xylulose 5-P
P4, the second pathway of converting glucose to arabitol (Povelainen and reductase; E9, a presumptive intracellular (or membrane-bound) sugar-
Miasnikov 2007); arabitol was oxidized to xylulose, followed by the phosphate phosphatase; E10, ribulokinase; E11, NAD/NADP-dependent
reduction of D-xylulose to xylitol; P5, the third pathway of conversion ketose reductase or NADH/NADPH-dependent polyol dehydrogenase;
of glucose to D-xylitol-5-phosphate suggested by Povelainen and E12, ribulose-5P-reductase; E14, NAD-dependent xylulose-forming
Miasnikov (2007). The enzymes are designated as follows: E1, NADP- arabitol dehydrogenase from K. pneumoniae; E15, NAD-dependent xy-
dependent glucose 6-P dehydrogenase; E2, NADP-dependent gluconate litol dehydrogenase from P. stipitis or G. oxydans or its native enzyme
6-P dehydrogenase; E3, ribulose-5-P-epimerase; E4, NAD-dependent

by co-expressing XDH from Gluconobacter oxydans and D- hand, overexpression of the glucose-6-phosphate and
arabitol dehydrogenase (DalD) gene from Klebsiella gluconate-6-phosphate dehydrogenase genes can also im-
pneumoniae in P. pastoris GS2270L in a 3-L fermenter. prove the PPP flux from glucose leading to an enhanced yield
Fermentation results showed that recombinant P. pastoris cells of ribulose-5-phosphate. The overexpression of the phosphor-
with both DalD and XDH converted glucose to xylitol with ylase gene and ribulose reductase gene would then dephos-
the highest yield of 0.078 g xylitol/g glucose and productivity phorylate ribulose-5-phosphate to ribulose and reduce
of 0.29 g xylitol/L h. The obtained yield was fourfolds higher ribulose to D-arabitol, respectively. Further studies are ongo-
than those of Harkki et al. (2004). This was the first report to ing in our laboratory to improve the pentose phosphate flux
convert xylitol from glucose by the pathway of glucose–D- from glucose by P. pastoris GS225.
arabitol–D-xylulose–xylitol in a single process (Cheng et al.
2014). The recombinant yeast could be used as a yeast cell Enzymatic biosynthesis of xylitol
factory and has the potential to produce xylitol from glucose.
However, in contrast to some other yeasts that achieved a D- Enzymatic bioconversion of xylose into xylitol offers a note-
arabitol yield of 20–30% from glucose (Ahmed 2001), less worthy alternative towards the large-scale production of xyli-
than 10% of glucose was converted to D-arabitol by tol. Enzyme-mediated catalytic reactions are generally consid-
P. pastoris GS225 even when pre-adapted in high glucose ered safe, highly reproducible, cost-efficient, and environmen-
concentration. One of the probable reasons is the low capacity tally friendly (Bilal et al. 2017, 2018a, b, c; Wang et al. 2018).
of the PPP in the GS225 strain because D-arabitol was syn- Though enzymatic biosynthesis of xylitol presents several ad-
thesized via PPP. The yield of D-arabitol from glucose might vantages over fermentative production, lower yields and pre-
be increased by driving glucose through the PPP by requisite of a high-precise control render this approach less
inactivating phosphoglucose isomerase-encoding gene (pgi) favorable (de Freitas Branco et al. 2012). As described above,
and simultaneously manipulating the cells NADPH demand xylose-assimilating yeasts transform D-xylose via two con-
by introducing a transhydrogenase reaction. On the other secutive enzymatic oxidoreductive reactions with the
5154 Appl Microbiol Biotechnol (2019) 103:5143–5160

activities of XR and XDH. D-Xylose is reduced to xylitol by high glucose flux ratio into pentose phosphate pathway, which
XR-driven catalytic conversion. In these bioprocesses, en- largely produces the xylitol intermediate D-xylulose that is
zymes can be incorporated as biocatalysts to improve or sub- also an extremely imperative strategy to improve the yield
stitute production steps (Bon et al. 2008). For the first time, and productivity of xylitol from glucose. In addition,
Kitpreechavanich et al. (1984) reported the xylitol production engineered microbial strains capable of simultaneously assim-
from xylose using XR from C. pelliculosa accompanied by an ilating multiple sugars found in plant biomasses would greatly
oxidoreductase system of Methanobacterium sp. for NADP increase the perspective of plant biomass as a sustainable and
reduction. About 90% xylose was converted to xylitol at pH renewable feedstock for the biosynthesis of xylitol and other
7.5 and 35 °C after a 24-h reaction period. The co-enzyme was value-added compounds.
successfully regenerated and retained in a membrane reactor Yarrowia lipolytica, an oleaginous yeast, plays a key role in
during this process. Later on, Nidetzky et al. (1996) evaluated biosynthesis, metabolic engineering, and biodegradation as
the conversion of xylose to xylitol using an NADH-dependent well as biotransformation processes, due to its tremendous
XR from C. tenuis. For the co-enzyme, they explored a potential to produce a variety of value-added metabolites such
charged membrane reactor and an enzymatic regeneration as short-chain fatty alcohols, alkanes as biofuels, polyunsatu-
system. In spite of the relatively good result, this technique rated fatty acids, polyhydroxyalkanoates and dicarboxylic
did not grasp required attention due to economic problems. acids, carotenoid-type pigments, and campesterol as a precur-
Kroutil et al. (2004) suggested that the system connected to sor for steroid drugs (Darvishi Farshad et al., 2018). It is gen-
the substrate using a single enzyme, while simultaneously erally regarded as safe by GRAS (generally recognized as
converting the substrate as well as co-substrate. However, safe) and a safe microorganism by the Food and Drug
the co-substrate must be incorporated in greater quantities Administration (FDA) in the USA (Zinjarde 2014).
than the substrate to favorably shift the equilibrium of the Therefore, it is widely applied in the hyperproduction of food
reactions. In such cases, a second biocatalyst has to be utilized ingredients and other industrially pertinent bioproducts, such
to restore the reduced co-enzyme using a co-substrate and as erythritol (Li et al. 2017; Cheng et al. 2018), lipase, man-
generating a co-product. Formate dehydrogenase (EC nitol, fatty acids and lipids (Tai and Stephanopoulos 2013;
1.2.1.2) from a recombinant strain of Pseudomonas sp. has Gonçalves et al. 2014), and citric acid (Ledesma-Amaro
shown great potential for the regeneration of NAD(P)H in et al. 2016).
the reaction medium. Similarly, the enzymatic biosynthesis In the case of Y. lipolytica yeast, glucose-glycerol mixture,
of xylitol by applying the glucose dehydrogenase system to carbohydrates, n-alkanes, and different classes of lipids except
regenerate NADPH was also found to be interesting because for sucrose act as a carbon source in the metabolic process
of cost-effective and environmentally responsiveness. In fu- (Ludwika et al. 2012; Zhu and Jackson 2015). With the recent
ture, the discovery or engineering of a more chemically stable improvements in synthetic biology, Y. lipolytica is the most
XR to increase its half-life B) in-house production of co- popular non-conventional organism with a relatively high tol-
enzymes using microbial biomass and the development of erance capacity to elevated concentrations of salts, hydrocar-
continuous bioconversion processes by the immobilized XR bons, and waste carbon (Fickers et al. 2005; Papanikolaou and
or using membrane reactors are important to increase the eco- Aggelis 2010), and can produce sugar alcohols in response to
nomic viability of the enzymatic xylitol process (de Freitas increased external high osmotic pressure (Ludwika et al.
Branco et al. 2012). 2012). Y. lipolytica has achieved a series of success in the
production of lipase (Brígida et al. 2014) and lipids with sub-
Challenges and future prospects stantial engineering efforts. More recently, Y. lipolytica was
used in our laboratory for the synthesis of erythritol using
Xylitol is an important alternative sweetener and widely ap- glucose as the main carbon source (Cheng et al. 2018).
plied in the food and pharmaceutical industries. Current man- Overexpression of the newly isolated two erythrose reductase
ufacture strategy mainly employs chemical methods to pro- genes (ER10 or ER25) and engineering of NADPH co-factor
duce xylitol from xylose purified from biomass hydrolysate. metabolism by overexpression of 6-phosphogluconate dehy-
In order to make biosynthesis method available for industrial drogenase genes (GND1) and glucose-6-phosphate dehydro-
production, extensive studies are needed to improve the yield genase genes (ZWF1) in Y. lipolytica strain CGMCC7326 led
of xylitol from a cheaply available material such as glucose. to a considerable increase in erythritol titer compared to the
Regulation of TCA cycle and pentose phosphate pathway or wild-type strain (Cheng et al. 2018).
modifying metabolic pathways, including knockout and ex- The pentose phosphate pathway is the same metabolic pro-
pression of critical genes in the artificial glucose-to-xylitol cess between xylitol and erythritol, and the common interme-
pathway, is of worth significance to scaling up this technolo- diate is D-xylulose. Xylitol can be obtained by reducing
gy. Discovery of a novel yeast with high tolerance to elevated xylulose with xylitol dehydrogenase. Rodriguez et al. (2016)
concentrations of carbon sources (such as glucose) and the reported that Y. lipolytica is naturally unable to grow on xylose
Appl Microbiol Biotechnol (2019) 103:5143–5160 5155

as its sole carbon source; however, a small amount of weak In order to restrict EMP pathway and block the synthesis of
growth was observed on xylitol by a partially active xylose erythritol, we deleted transketolase genes (TKL2 and TKL1)
pathway (Rodriguez et al. 2016). They sought whether there and mannitol dehydrogenase (MDH) gene, which can de-
were xylose reductase (XYR1), xylitol dehydrogenase crease the number of by-products, such as erythritol and man-
(XDH), and xylulose kinase (XKS) genes in the oxidoreduc- nitol. According to Fig. 3, D-ribulose and D-xylulose can be
tase pathway of xylose metabolism in Y. lipolytica. Measuring reduced to D-arabitol and D-arabitol, respectively, using
native mRNA expression and extent of inducibility (ΔΔCT) of arabinitol dehydrogenase (Cheng et al. 2009). Disruption of
these three candidate genes by quantitative PCR, they found arabinitol dehydrogenase gene and overexpression of
that the weakened expression of XKS is one of the major xylulose-5-P phosphatase gene (convert xylulose-5-P to
bottlenecks for xylose and xylitol, and metabolism could be xylulose), and XDH gene (reduce xylulose to xylitol) can lead
the limited expression of XKS (Rodriguez et al. 2016). to the accumulation of xylitol (Fig. 6). Therefore, the
In Y. lipolytica yeast systems, especially for erythritol- xylulokinase gene (XSK) was inactivated to hinder the con-
producing strains, the pentose phosphate pathway is the major version of xylulose to 5-P-xylulose. As 5-P-ribulose, 5-P-
metabolic pathway for glucose utilization; however, about xylulose, and 5-P-ribose are isomers of each other, deletion
20% of glucose is metabolized by Embden–Meyerhof– of 5-P-ribulose isomerase (RPI) can improve the ratio of 5-P-
Parnas glycolytic pathway (EMP) to produce valuable ribulose to 5-P-xylulose by hindering the conversion of 5-P-
chemicals, including oleochemicals, nutraceuticals, organic ribulose to 5-P-ribose. Moreover, the xylulose conversion to
acids, and many other specialty chemicals (Markham and xylitol process is reversible, and substrate inhibition exists,
Alper 2018). Our group observed that the pentose phosphate which limits the accumulation of xylitol. Therefore, the im-
pathway is the same metabolic process between xylitol and portant step is to transport xylitol to extracellular by xylitol
erythritol, and D-xylulose is one of the common intermediates transporter protein.
converted from glucose. We attempted to utilize glucose as a Though several microorganisms such as bacteria, fungi,
carbon source to improve the yield and productivity of xylitol and yeasts have been reported in the literature for xylitol pro-
by knocking out genes belonging to EMP, which indirectly duction, yeasts are considered the best xylitol producers.
affect PPP (Mirończuk et al. 2017) and blocking the synthesis Among the yeasts, Y. lipolytica has distinct metabolic advan-
of erythritol in yeast to obtain xylitol by pathway engineering tages, such as great tolerance to chemicals, high protein pro-
and metabolic pathway. duction, and the lack of tight metabolic regulation, which

Fig. 6 Schematic representation of two metabolic pathways for the kinase; E6, NAD/NADP-dependent ketose reductase or NADH/
conversion of glucose to xylitol in engineered Y. lipolytica. PTS NADPH-dependent arabitol dehydrogenase; E7, NADH/NADPH-
phosphotransferase system. The enzymes are designated as follows: E1, dependent arabitol dehydrogenase; E8, NADH/NADPH-dependent man-
epimerase; E2, kinase or sugar phosphate phosphatase; E3, NAD- nitol dehydrogenase
dependent xylitol dehydrogenase; E4, NADPH transhydrogenase; E5,
5156 Appl Microbiol Biotechnol (2019) 103:5143–5160

make it an attractive host in synthetic biology. However, the environmental responsive technology to tackle dye-based industrial
pollutants—a review. Sci Total Environ 576:646–659
development of competitive genetic tools, improving transfor-
Bilal M, Iqbal HM, Hu H, Wang W, Zhang X (2018a) Metabolic engi-
mation efficiency, and utilization of the CRISPR/Cas9 for neering pathways for rare sugars biosynthesis, physiological func-
gene modification are some major limitations (Markham and tionalities, and applications—a review. Crit Rev Food Sci Nutr
Alper 2018). Moreover, the construction of metabolically ro- 58(16):2768–2778
Bilal M, Iqbal HM, Hu H, Wang W, Zhang X (2018b) Metabolic engi-
bust and stable strain, identification of new strains with high
neering and enzyme-mediated processing: a biotechnological ven-
tolerance to lignocellulosic hydrolysate inhibitors, scaled-up ture towards biofuel production—a review. Renew Sust Energ Rev
bioprocessing, efficient downstream processing technologies 82:436–447
for high product recovery, and economic bioconversion of Bilal M, Nawaz MZ, Iqbal H, Hou J, Mahboob S, Al-Ghanim KA, Cheng
H (2018c) Engineering ligninolytic consortium for bioconversion of
lignocellulosic biomass to xylitol are also the challenges that
lignocelluloses to ethanol and chemicals. Protein Pept Lett 25(2):
need to be addressed to make economical production of bio- 108–119
xylitol a reality. We believe that engineered Y. lipolytica will Blakley ER, Spencer JF (1962) Studies on the formation of D-arabitol by
be applied in industrial-scale xylitol production by a one-step osmophilic yeasts. Can J Biochem Physiol 40(12):1737–1748
Bolen PL, Mccracken DA (1990) Properties of aldose reductase from the
fermentation process from glucose as a carbon source.
methanol yeast Candida boidinii. J Ferment Bioeng 69(4):211–214
Bon EPS, Ferrara MA, Corvo ML (2008) Enzimas em Biotecnologia -
Funding This research was supported by the National Natural Science Produção, Aplicação e Mercado (Enzymes in biotechnology: pro-
Foundation of China (No. 21877078). duction, application and market). Interciência, Rio de Janeiro
Brígida AIS, Amaral PFF, Coelho MAZ, Gonçalves LRB (2014) Lipase
Compliance with ethical standards from Yarrowia lipolytica: production, characterization and applica-
tion as an industrial biocatalyst. J Mol Catal B Enzym 101(2):148–
158
Conflict of interest The authors declare that they have no conflict of
Bruinenberg PM, Bot PHMD, Dijken JPV, Scheffers WA (1983) The role
interest.
of redox balances in the anaerobic fermentation of xylose by yeasts.
Eur J Appl Microbiol Biotechnol 18(5):287–292
Ethical approval This article does not contain any studies with human Carvalho GB, Mussatto SI, Cândido EJ, Silva JBAE (2010) Comparison
participants or animals performed by any of the authors. of different procedures for the detoxification of eucalyptus
hemicellulosic hydrolysate for use in fermentative processes. J
Chem Technol Biotechnol 81(2):152–157
Cassland P, Jönsson LJ (1999) Characterization of a gene encoding
References Trametes versicolor laccase A and improved heterologous expres-
sion in Saccharomyces cerevisiae by decreased cultivation temper-
Ahmad I, Shim WY, Kim JH (2013) Enhancement of xylitol production ature. Appl Microbiol Biotechnol 52(3):393–400
in glycerol kinase disrupted Candida tropicalis by co-expression of Chen J, Zhang Y, Wang Y, Ji X, Zhang L, Mi X, Huang H (2013)
three genes involved in glycerol metabolic pathway. Bioprocess Removal of inhibitors from lignocellulosic hydrolyzates by vacuum
Biosyst Eng 36:1279–1284 membrane distillation. Bioresour Technol 144:680–683
Ahmed Z (2001) The properties of Candida famata R28 for D-arabitol Cheng H, Li Z, Jiang N, Deng Z (2009) Cloning, purification and char-
production from D-glucose. Online J Biol Sci 1(11):1005–1008 acterization of an NAD-dependent D-Arabitol dehydrogenase from
Akinterinwa O, Cirino P (2009) Heterologous expression of D- acetic acid bacterium, Acetobacter suboxydans. Protein J 28(6):263–
xylulokinase from Pichia stipitis enables high levels of xylitol pro- 272
duction by engineered Escherichia coli growing on xylose. Metab Cheng H, Wang B, Lv J, Jiang M, Lin S, Deng Z (2011) Xylitol produc-
Eng 11(1):48–55 tion from xylose mother liquor: a novel strategy that combines the
use of recombinant Bacillus subtilis and Candida maltosa. Microb
Akinterinwa O, Khankal R, Cirino PC (2008) Metabolic engineering for
Cell Factories 10:5
bioproduction of sugar alcohols. Curr Opin Biotechnol 19(5):461–
Cheng H, Lv J, Wang H, Wang B, Li Z, Deng Z (2014) Genetically
467
engineered Pichia pastoris yeast for conversion of glucose to xylitol
Albuquerque TLD, Jr IJDS, Macedo GRD, Rocha MVP (2014) by a single-fermentation process. Appl Microbiol Biotechnol 98(8):
Biotechnological production of xylitol from lignocellulosic wastes: 3539–3552
a review. Process Biochem 49(11):1779–1789 Cheng H, Wang S, Bilal M, Ge X, Zhang C, Fickers P, Cheng H (2018)
Aranda-Barradas JS, Delia ML, Riba JP (2000) Kinetic study and model- Identification, characterization of two NADPH-dependent erythrose
ling of the xylitol production using Candida parapsilosis in oxygen- reductases in the yeast Yarrowia lipolytica and improvement of
limited culture conditions. Bioprocess Eng 22(3):219–225 erythritol productivity using metabolic engineering. Microb Cell
Bae SM, Park YC, Lee TH, Kweon DH, Choi JH, Kim SK, Ryu YW, Seo Factories 17:133
JH (2004) Production of xylitol by recombinant Saccharomyces Chin JW, Cirino PC (2011) Improved NADPH supply for xylitol produc-
cerevisiae containing xylose reductase gene in repeated fed-batch tion by engineered Escherichia coli with glycolytic mutations.
and cell-recycle fermentations. Enzym Microb Technol 35(6):545– Biotechnol Prog 27(2):333–341
549 Chun BW, Dair B, Macuch PJ, Wiebe D, Porteneuve C, Jeknavorian A
Banta S, Swanson BA, Wu S, Jarnagin A, Anderson S (2002) Alteration (2006) The development of cement and concrete additive: based on
of the specificity of the cofactor-binding pocket of Corynebacterium xylonic acid derived via bioconversion of xylose. Appl Microbiol
2,5-diketo-D-gluconic acid reductase A. Protein Des Eng Sel 15: Biotechnol 129-132(1–3):645
131–140 Cirino PC, Chin JW, Ingram LO (2010) Engineering Escherichia coli for
Bilal M, Asgher M, Parra-Saldivar R, Hu H, Wang W, Zhang X, Iqbal xylitol production from glucose-xylose mixtures. Biotechnol
HMN (2017) Immobilized ligninolytic enzymes: an innovative and Bioeng 95(6):1167–1176
Appl Microbiol Biotechnol (2019) 103:5143–5160 5157

Converti A, Parego P, Domínguez JM (1999) Xylitol production from Granström TB, Izumori K, Leisola M (2007a) A rare sugar xylitol. Part I:
hardwood hemicellulose hydrosylates. Appl Biochem Biotechnol the biochemistry and biosynthesis of xylitol. Appl Microbiol
82:141–151 Biotechnol 74(2):277–281
Cruz JM, Domínguez JM, Domínguez H, Parajó JC (2000) Xylitol pro- Granström TB, Izumori K, Leisola M (2007b) A rare sugar xylitol. Part II:
duction from barley bran hydrolysates by continuous fermentation biotechnological production and future applications of xylitol. Appl
with Debaryomyces hansenii. Biotechnol Lett 22(23):1895–1898 Microbiol Biotechnol 74(2):273–276
Darvishi Farshad AM, Marella ER, Borodina I (2018) Advances in syn- Häcker B, Habenicht A, Kiess M, Mattes R (1999) Xylose utilisation:
thetic biology of oleaginous yeast Yarrowia lipolytica for producing cloning and characterisation of the xylose reductase from Candida
non-native chemicals. Appl Microbiol Biotechnol 102(14):5925– tenuis. Biol Chem 380(12):1395–1403
5938 Hallborn J, Walfridsson M, Airaksinen U, Ojamo H, Hahn-Hägerdal B,
Datsenko KA, Wanner BL (2000) One-step inactivation of chromosomal Penttilä M, Keräsnen S (1991) Xylitol production by recombinant
genes in Escherichia coli K-12 using PCR products. PNAS 97(12): Saccharomyces cerevisiae. Nat Biotechnol 9(11):1090–1095
6640–6645 Hardy GP, Mj TDM, Neijssel OM (1993) Energy conservation by
De Albuquerque TL, Gomes SDL, Marques JE Jr, da Silva IJ Jr, Rocha pyrroloquinoline quinol-linked xylose oxidation in Pseudomonas
MVP (2015) Xylitol production from cashew apple bagasse by putida NCTC 10936 during carbon-limited growth in chemostat
Kluyveromyces marxianus CCA510. Catal Today 255:33–40 culture. FEMS Microbiol Lett 107(1):107–110
Díaz-Fernández D, Lozano-Martínez P, Buey RM, Revuelta JL, Jiménez Harkki AM, Myasnikov AN, Apajalahti JHA, Pastinen OA (2004)
A (2017) Utilization of xylose by engineered strains of Ashbya Manufacture of xylitol using recombinant microbial hosts. United
gossypii for the production of microbial oils. Biotechnol Biofuels States Patent 6723540
10(1):3 Hong Y, Dashtban M, Kepka G, Chen S, Qin W (2014a) Overexpression
Dien BS, Hespell RB, Ingram LO, Bothast RJ (1997) Conversion of corn of D-xylose reductase (xyl1) gene and antisense inhibition of D-
milling fibrous co-products into ethanol by recombinant Escherichia xylulokinase (xyiH) gene increase xylitol production in
coli strains K011 and SL40. World J Microbiol Biotechnol 13(6): Trichoderma reesei. Biomed Res Int 2014:169705–169705
619–625 Hong Y, Dashtban M, Kepka G, Chen S, Qin W (2014b) Overexpression
Dien BS, Nichols NN, O'Bryan PJ, Bothast RJ (2000) Development of of d-xylose reductase (xyl1) gene and antisense inhibition of
new ethanologenic Escherichia coli strains for fermentation of lig- dxylulokinase (xyiH) gene increase xylitol production in
nocellulosic biomass. Appl Biochem Biotechnol 84-86:181–196 Trichoderma reesei. Biomed Res Int 2014:8–8
Einav T, Duque J, Phillips R (2018) Theoretical analysis of inducer and Ingram JM, Wood WA (1965) Enzymatic basis for D-arabitol production
operator binding for cyclic-AMP receptor protein mutants. PLoS by Saccharomyces rouxii. J Bacteriol 89(5):1186
One 13(9):e0204275 Izumori K, Tuzaki K (1988) Production of xylitol from D-xylulose by
Escalante J, Caminal G, Figueredo M, Mas CD (1990) Production of Mycobacterium smegmatis. J Ferment Technol 66(1):33–36
arabitol from glucose by Hansenula polymorpha. J Ferment Jeon WY, Yoon BH, Ko BS, Shim WY, Kim JH (2012) Xylitol produc-
Bioeng 70(4):228–231 tion is increased by expression of codon-optimized Neurospora
Fickers P, Benetti PH, Waché Y, Marty A, Mauersberger S, Smit MS, crassa xylose reductase gene in Candida tropicalis. Bioprocess
Nicaud JM (2005) Hydrophobic substrate utilisation by the yeast, Biosyst Eng 35(1–2):191–198
and its potential applications. FEMS Yeast Res 5(6–7):527–543 Jeon WY, Shim WY, Lee SH, Choi JH, Kim JH (2013) Effect of heter-
Fonseca BG, Moutta RO, Ferraz FO, Vieira ER, Nogueira AS, Baratella ologous xylose transporter expression in Candida tropicalis on xy-
BF, Rodrigues LC, Hourui Z, Da SS (2011) Biological detoxifica- litol production rate. Bioprocess Biosyst Eng 36(6):809–817
tion of different hemicellulosic hydrolysates using Issatchenkia Jeong EY, Sopher C, Kim IS, Lee H (2010) Mutational study of the role of
occidentalis CCTCC M 206097 yeast. J Ind Microbiol Biotechnol tyrosine-49 in the Saccharomyces cerevisiae xylose reductase. Yeast
38(1):199–207 18(11):1081–1089
Ford G, Ellis EM (2001) Aldo-keto reductases of the yeast Jin LQ, Xu W, Yang B, Liu ZQ, Zheng YG (2018) Efficient biosynthesis
Saccharomyces cerevisiae. Chem-Biol Interac 130-132(1–3):685– of xylitol from xylose by coexpression of xylose reductase and glu-
698 cose dehydrogenase in Escherichia coli. Appl Biochem Biotechnol
Franceschin G, Sudiro M, Ingram T, Smirnova I, Brunner G, Bertucco A 187(4), 1143–1157
(2011) Conversion of rye straw into fuel and xylitol: a technical and Johnsen U, Schönheit P (2004) Novel xylose dehydrogenase in the hal-
economical assessment based on experimental data. Chem Eng Res ophilic archaeon Haloarcula marismortui. J Bacteriol 186(18):
Des 89(6):631–640 6198–6207
de Freitas Branco R, Chandel AK, da Silva SS (2012) Enzymatic produc- Johnsen U, Dambeck M, Zaiss H, Fuhrer T, Soppa J, Sauer U, Schönheit
tion of xylitol: current status and future perspectives. In: D-Xylitol. P (2009) D-xylose degradation pathway in the halophilic archaeon
Springer, Berlin, pp 193–204 Haloferax volcanii. J Biol Chem 284(40):27290–27303
Furlan SA, Bouilloud P, Castro HFD (1994) Influence of oxygen on Jovall PA, Tunblad-Johansson I, Adler L (1990) 13C NMR analysis of
ethanol and xylitol production by xylose fermenting yeasts. production and accumulation of osmoregulatory metabolites in the
Process Biochem 29(8):657–662 salt-tolerant yeast Debaryomyces hansenii. Arch Microbiol 154(3):
Galar ML, Boiardi JL (1995) Evidence for a membrane-bound 209–214
pyrroloquinoline quinone-linked glucose dehydrogenase in Kanauchi M, Bamforth CW (2012) Use of xylose dehydrogenase from
Acetobacter diazotrophicus. Appl Microbiol Biotechnol 43(4): Trichoderma viride in an enzymic method for the measurement of
713–716 pentosan in barley. J Brewing 109(3):203–207
Gonçalves FAG, Colen G, Takahashi JA (2014) Yarrowia lipolytica and Kang MH, Ni H, Jeffries TW (2003) Molecular characterization of a gene
its multiple applications in the biotechnological industry. Sci World for aldose reductase (CbXYL1) from Candida boidinii and its ex-
J 2014:476207 pression in Saccharomyces cerevisiae. Appl Biochem Biotechnol
Govinden R, Pillai B, van Zyl WH, Pillay D (2001) Xylitol production by 105(108):265–276
recombinant Saccharomyces cerevisae expressing the Pichia stipitis Khankal R, Chin JW, Cirino PC (2008) Role of xylose transporters in
and Candida shehatae XYL1 genes. Appl Microbiol Biotechnol 55: xylitol production from engineered Escherichia coli. J Biotechnol
76–80 134(3):246–252
5158 Appl Microbiol Biotechnol (2019) 103:5143–5160

Kiessling H, Lindberg B, Mckay J, Block-Bolten A, Toguri JM (1962) Mayer G, Kulbe KD, Nidetzky B (2002) Utilization of xylitol dehydro-
Some products of the metabolism of D-xylose by Pullularia genase in a combined microbial/enzymatic process for production of
pullulans. Acta Chem Scand 16:1858–1862 xylitol from D-glucose. Appl Biochem Biotechnol 98-100(1–9):
Kim YS, Kim SY, Kim JH, Kim SC (1999) Xylitol production using 577–590
recombinant Saccharomyces cerevisiae containing multiple xylose Meinander NQ, Hahnhägerdal B (1997) Influence of cosubstrate concen-
reductase genes at chromosomal delta-sequences. J Biotechnol tration on xylose conversion by recombinant, XYL1-expressing
67(2–3):159–171 Saccharomyces cerevisiae: a comparison of different sugars and
Kim S-H, Yun J-Y, Kim S-G, Seo J-H, Park J-B (2010) Production of ethanol as cosubstrates. Appl Environ Microbiol 63(5):1959–1964
xylitol from D-xylose and glucose with recombinant Meng J, Sun W, Wang F, Deng W, Luo Y, Zhu J, Lin J, Wang X, Qi X
Corynebacterium glutamicum. Enzym Microb Technol 46(5):366– (2014) Microbial and enzymatic process for xylitol production from
371 D-glucose. Curr Org Chem 18(24):3131–3135
Kitpreechavanich V, Hayashi M, Nishio N, Nagai S (1984) Conversion of Mirończuk AM, Biegalska A, Dobrowolski A (2017) Functional overex-
D-xylose into xylitol by xylose reductase from Candida pelliculosa pression of genes involved in erythritol synthesis in the yeast
coupled with the oxidoreductase system of methanogen strain HU. Yarrowia lipolytica. Biotechnol Biofuels 10(1):77
Biotechnol Lett 6(10):651–656 Mohamad NL, Kamal SMM, Mokhtar MN (2015) Xylitol biological
Ko BS, Kim J, Kim JH (2006) Production of xylitol from D-xylose by a production: a review of recent studies. Food Rev Int 31(1):74–89
xylitol dehydrogenase gene-disrupted mutant of Candida tropicalis. Morita TA, Silva SS (2000) Inhibition of microbial xylitol production by
Appl Environ Microbiol 72(6):4207–4213 acetic acid and its relation with fermentative parameters. Appl
Kogje A, Ghoshalkar A (2016) Xylitol production by Saccharomyces Biochem Biotechnol 84-86(1–9):801–808
cerevisiae overexpressing different xylose reductases using non- Mussatto SI, Roberto IC (2003) Xylitol production from high xylose
detoxified hemicellulosic hydrolysate of corncob. 3 Biotech 6: concentration: evaluation of the fermentation in bioreactor under
127–136 different stirring rates. J Appl Microbiol 95(2):331–337
Kötter P, Amore R, Hollenberg CP, Ciriacy M (1990) Isolation and char- Nichols NN, Saha BC (2016) Production of xylitol by a Coniochaeta
acterization of the Pichia stipitis xylitol dehydrogenase gene, XYL2, ligniaria strain tolerant of inhibitors and defective in growth on
and construction of a xylose-utilizing Saccharomyces cerevisiae xylose. Biotechnol Prog 32(3):606–612
transformant. Curr Genet 18(6):493–500 Nidetzky B, Neuhauser W, Haltrich D, Kulbe KD (1996) Continuous
Kroutil W, Mang H, Edegger K, Faber K (2004) Recent advances in the enzymatic production of xylitol with simultaneous coenzyme regen-
biocatalytic reduction of ketones and oxidation of sec-alcohols. Curr eration in a charged membrane reactor. Biotechnol Bioeng 52:387–
Opin Chem Biol 8:120–126 396
Kumar J, Reddy MS, Rao LV (2010) Strain improvement of Candida
Nidetzky B, Helmer H, Klimacek M, Lunzer R, Mayer G (2003)
tropicalis ovc5 for xylitol production by random mutagenesis.
Characterization of recombinant xylitol dehydrogenase from
IIOAB J 1:24–28
Galactocandida mastotermitis expressed in Escherichia coli.
Kwon DH, Kim MD, Lee TH, Oh YJ, Ryu YW, Seo JH (2006) Elevation
Chem Biol Interact 143(8):533–542
of glucose 6 phosphate dehydrogenase activity increases xylitol pro-
Nidetzky B, Neuhauser W, Haltrich D, Kulbe KD (2015) Continuous
duction in recombinant Saccharomyces cerevisae. J Mol Catal B-
enzymatic production of xylitol with simultaneous coenzyme regen-
Enzyme 43:86–89
eration in a charged membrane reactor. Biotechnol Bioeng 52(3):
Ledesma-Amaro R, Lazar Z, Rakicka M, Guo Z, Fouchard F, Coq CL,
387–396
Nicaud JM (2016) Metabolic engineering of Yarrowia lipolytica to
Nigam P, Singh D (1995) Processes of fermentative production of
produce chemicals and fuels from xylose. Metab Eng 38:115–124
xylitol—a sugar substitute. Process Biochem 30(2):117–124
Li L, Wang H, Cheng H, Deng Z (2017) Isomaltulose production by yeast
surface display of sucrose isomerase from Pantoea dispersa on Niu W, Molefe MN, Frost JW (2003) Microbial synthesis of the energetic
Yarrowia lipolytica. J Funct Foods 32:208–217 material precursor 1,2,4-butanetriol. J Am Chem Soc 125(43):
Li Q, Qin Y, Liu Y, Liu J, Liu Q, Li P, Liu L (2019) Detoxification and 12998–12999
concentration of corn stover hydrolysate and its fermentation for Nozaki H, Suzuki S, Tsuyoshi N, Yokozeki K (2003) Production of D-
ethanol production. Front Chem Sci Eng 13(1):140–151 arabitol by Metschnikowia reukaufii AJ14787. Biosci Biotechnol
Liu J, Qin Y, Li P, Zhang K, Liu Q, Liu L (2016) Separation of the acid Biochem 67(9):1923–1929
sugar mixtures by using acid retardation and further concentration of Nyyssölä A, Pihlajaniemi A, Palva A, Von Weymarn N, Leisola M (2005)
the eluents by using continuous-effect membrane distillation. J Production of xylitol from D-xylose by recombinant Lactococcus
Chem Technol Biotechnol (Oxford, Oxfordshire) 91(4):1105–1112 lactis. J Biotechnol 118(1):55–66
Lohmeier-Vogel EM, Sopher CR, Lee H (1998) Intracellular acidification Oh YJ, Lee TH, Lee SH, Oh EJ, Ryu YW, Kim MD, Seo JH (2007) Dual
as a mechanism for the inhibition by acid hydrolysis-derived inhib- modulation of glucose-6-phosphate metabolism to increase
itors of xylose fermentation by yeasts. J Ind Microbiol Biotechnol NADPH-dependent xylitol production in Saccharomyces cerevisae.
20(2):75–81 J Mol Catal B-Enzyme 47:37–42
Lowe DA, Jennings DH (1985) Carbohydrate metabolism in the fungus Onishi H, Suzuki T (1969) Microbial production of xylitol from glucose.
Dendryphiella salina. V. The pattern of label in arabitol and poly- Appl Microbiol 18(6):1031–1035
saccharide after growth in the presence of specifically labelled car- Pal S, Choudhary V, Kumar A, Biswas D, Mondal AK, Sahoo DK (2013)
bon sources. New Phytol 101(4):399–403 Studies on xylitol production by metabolic pathway engineered
Ludwika T, Anita R, Witold G (2012) Production of erythritol and man- Debaryomyces hansenii. Bioresour Technol 147(8):449–455
nitol by Yarrowia lipolyticayeast in media containing glycerol. J Ind Pal S, Mondal AK, Sahoo DK (2016) Molecular strategies for enhancing
Microbiol Biotechnol 39(9):1333–1343 microbial production of xylitol. Process Biochem 51(7):809–819
Mancilha IMD, Karim MN (2010) Evaluation of ion exchange resins for Palmqvist E, Hahnhägerdal B (2000) Fermentation of lignocellulosic
removal of inhibitory compounds from corn stover hydrolyzate for hydrolysates. I: inhibition and detoxification. Bioresour Technol
xylitol fermentation. Biotechnol Prog 19(6):1837–1841 74(1):17–24
Markham KA, Alper HS (2018) Synthetic biology expands the industrial Pandey A, Vt NPS, Soccol CR (2000) Biotechnological potential of agro-
potential of Yarrowia lipolytica. Trends Biotechnol 36(10):1085– industrial residues. I: sugarcane bagasse. Bioresour Technol 74(1):
1095 69–80
Appl Microbiol Biotechnol (2019) 103:5143–5160 5159

Papanikolaou S, Aggelis G (2010) Biotechnological valorization of bio- reductase of Candida tropicalis and production of xylitol in
diesel derived glycerol waste through production of single cell oil Escherichia coli. J Ferment Bioeng 87(3):280–284
and citric acid by Yarrowia lipolytica. Lipid Technol 21(4):83–87 Suzuki S, Sugiyama M, Mihara Y, Hashiguchi K, Yokozeki K (2002)
Parajó JC, Domínguez H, Domínguez J (1998a) Biotechnological pro- Novel enzymatic method for the production of xylitol from D-
duction of xylitol. Part 1: interest of xylitol and fundamentals of its arabitol by Gluconobacter oxydans. Biosci Biotechnol Biochem
biosynthesis. Bioresour Technol 65(3):191–201 66(12):2614–2620
Parajó JC, Domínguez H, Domínguez J (1998b) Biotechnological pro- Tai M, Stephanopoulos G (2013) Engineering the push and pull of lipid
duction of xylitol. Part 2: operation in culture media made with biosynthesis in oleaginous yeast Yarrowia lipolytica for biofuel pro-
commercial sugars. Bioresour Technol 65(3):203–212 duction. Metab Eng 15(1):1–9
Peng F, Peng P, Xu F, Sun RC (2012) Fractional purification and biocon- Tochampa W, Sirisansaneeyakul S, Vanichsriratana W, Srinophakun P,
version of hemicelluloses. Biotechnol Adv 30(4):879–903 Bakker HH, Chisti Y (2005) A model of xylitol production by the
Pepper T, Olinger PM (1988) Xylitol in sugar-free confections. Food yeast Candida mogii. Bioprocess Biosyst Eng 28(3):175–183
Technol 42:98–106 Toivari MH, Nygard Y, Penttila M, Ruohonen L, Wiebe MG (2012)
Pezzotti F, Therisod M (2006) Enzymatic synthesis of aldonic acids. Microbial D-xylonate production. Appl Microbiol Biotechnol
Carbohydr Res 341(13):2290–2292 96(1):1–8
Povelainen M, Miasnikov AN (2007) Production of xylitol by metabol- Uppada V, Bhaduri S, Noronha SB (2014) Cofactor generation—an im-
ically engineered strains of Bacillus subtilis. J Biotechnol 128(1): portant aspect of biocatalysis. Curr Sci 106:946–957
24–31 US Food and Drug Administration (2008) Guidance for industry: a food
Rao RS, Chp J, Prakasham RS, Sarma PN, Rao LV (2006) Xylitol pro- labeling guide. Food and Drug Administration, Washington, DC
duction from corn fiber and sugarcane bagasse hydrolysates by Wang H, Li L, Zhang L, An J, Cheng H, Deng Z (2016) Xylitol produc-
Candida tropicalis. Bioresour Technol 97(15):1974–1978 tion from waste xylose mother liquor containing miscellaneous
Rivas B, Torre P, Domínguez JM, Perego P, Converti A, Parajó JC (2010) sugars and inhibitors: one-pot biotransformation by Candida
Carbon material and bioenergetic balances of xylitol production tropicalis and recombinant Bacillus subtilis. Microb Cell Factories
from corncobs by Debaryomyces hansenii. Biotechnol Prog 19(3): 15(1):82
706–713 Wang S, Bilal M, Hu H, Wang W, Zhang X (2018) 4-Hydroxybenzoic
Rizzi M, Harwart K, Bui-Thanh NA, Dellweg H (1989) A kinetic study acid—a versatile platform intermediate for value-added compounds.
of the NAD+ -xylitol-dehydrogenase from the yeast Pichia stipitis. J Appl Microbiol Biotechnol 102(8):3561–3571
Ferment Bioeng 67(1):25–30 Wannawilai S, Lee WC, Chisti Y, Sirisansaneeyakul S (2017) Furfural
Roberto IC, Mancilha IM, Souza CAD, Felipe MGA, Sato S, Castro HFD and glucose can enhance conversion of xylose to xylitol by Candida
(1994) Evaluation of rice straw hemicellulose hydrolysate in the magnoliae TISTR 5663. J Biotechnol 241:147–157
production of xylitol by Candida guilliermondii. Biotechnol Lett Washüttl J, Riederer P, Bancher E (1973) A qualitative and quantitative
16(11):1211–1216 study of sugar-alcohols in several foods. J Food Sci 38:1262–1263
Rodrigues RCLB, Kenealy WR, Jeffries TW (2011) Xylitol production Wei J, Yuan Q, Wang T, Wang L (2010) Purification and crystallization of
from DEO hydrolysate of corn stover by Pichia stipitis YS-30. J Ind xylitol from fermentation broth of corncob hydrolysates. Front
Microbiol Biotechnol 38(10):1649–1655 Chem Eng China 4(1):57–64
Rodriguez GM, Hussain MS, Gambill L, Gao D, Yaguchi A, Blenner M Weimberg R (1962) Mode of formation of D-arabitol by Saccharomyces
(2016) Engineering xylose utilization in Yarrowia lipolytica by un- mellis. Biochem Biophys Res Commun 8(6):442–445
derstanding its cryptic xylose pathway. Biotechnol Biofuels 9:149 Werpy T, Petersen G, Aden A, Bozell J, Holladay J, White J, Manheim A,
Saha BC, Sakakibara Y, Cotta MA (2007) Production of D-arabitol by a Eliot D, Lasure L, Jones S (2004) Top value added chemicals from
newly isolated Zygosaccharomyces rouxii. J Ind Microbiol biomass. Volume 1—results of screening for potential candidates
Biotechnol 34(7):519–523 from sugars and synthesis gas. Department of Energy, Washington
Sasaki M, Jojima T, Inui M, Yukawa H (2010) Xylitol production by DC
recombinant Corynebacterium glutamicum under oxygen depriva- Winkelhausen E, Kuzmanova S (1998) Microbial conversion of D-xylose
tion. Appl Microbiol Biotechnol 86(4):1057–1066 to xylitol. J Ferment Bioeng 86:1–14
Silva SS, Felipe MGA, Silva J, Prata AMR (1998) Acid hydrolysis of Wong B, Leeson S, Grindle S, Magee B, Brooks E, Magee PT (1995) D-
Eucalyptus grandis chips for microbial production of xylitol. arabitol metabolism in Candida albicans: construction and analysis
Process Biochem 33(1):63–67 of mutants lacking D-arabitol dehydrogenase. J Bacteriol 177(11):
Silveira MM, Jonas R (2002) The biotechnological production of sorbi- 2971–2976
tol. Appl Microbiol Biotechnol 59(4–5):400–408 Wu J, Hu J, Zhao S, He M, Hu G, Ge X, Peng N (2018) Single-cell
Simpson FJ (1966) D-Xylulokinase. Methods Enzymol 9:454–458 protein and xylitol production by a novel yeast strain Candida
Sohyun K, Yun JY, Sunggun K, Jinho S, Jinbyung P (2010) Production of intermedia FL023 from lignocellulosic hydrolysates and xylose.
xylitol from D-xylose and glucose with recombinant Appl Biochem Biotechnol 185:1–16
Corynebacterium glutamicum. Enzym Microb Technol 46(5):366– Yong SK, Sang YK, Kim JH, Sun CK (1999) Xylitol production using
371 recombinant Saccharomyces cerevisiae containing multiple xylose
Stephens C, Christen B, Fuchs T, Sundaram V, Watanabe K, Jenal U reductase genes at chromosomal δ-sequences. J Biotechnol 67(2–3):
(2007) Genetic analysis of a novel pathway for D-xylose metabo- 159–171
lism in Caulobacter crescentus. J Bacteriol 189(5):2181–2185 Yoshitake J, Ishizaki H, Shimamura M, Imai T (2014) Xylitol production
Sugiyama M, Suzuki S, Tonouchi N, Yokozeki K (2003) Cloning of the by an Enterobacter species. Biosci Biotechnol Biochem 37(10):
xylitol dehydrogenase gene from Gluconobacter oxydans and im- 2261–2267
proved production of xylitol from D-arabitol. Biosci Biotechnol Zha J, Li B-Z, Shen M-H, Hu M-L, Song H, Yuan Y-J (2013a)
Biochem 67(3):584–591 Optimization of CDT-1 and XYL1 expression for balanced copro-
Suzuki T, Onishi H (1973) Oxidation and reduction of D-xylose by cell- duction of ethanol and xylitol from cellobiose and xylose by
free extract of Pichia quercuum. Appl Microbiol 25(5):850–852 engineered Saccharomyes cerevisiae. PLoS One 8(7):e68317
Suzuki T, Yokoyama S, Kinoshita Y, Yamada H, Hatsu M, Takamizawa Zha J, Li BZ, Shen MH, Hu ML, Song H, Yuan YJ (2013b) Optimization
K, Kawai K (1999) Expression of xyrA gene encoding for D-xylose of cdt-1and xyl1 expression for balanced co-production of ethanol
5160 Appl Microbiol Biotechnol (2019) 103:5143–5160

and xylitol from cellobiose and xylose by engineered Zhang J, Zhang B, Wang D, Gao X, Hong J (2015) Improving xylitol
Saccharomyces cerevisiae. PLoS One 8:e68317 production at elevated temperature with engineered Kluyveromyces
Zhang FW, Qiao DR, Xu H, Liao C, Li SL, Cao Y (2009a) Cloning, marxianus through over-expressing transporters. Bioresour Technol
expression, and characterization of xylose reductase with higher 175:642–645
activity from Candida tropicalis. J Microbiol 47(3):351–357 Zhu Q, Jackson EN (2015) Metabolic engineering of Yarrowia lipolytica
Zhang HR, Qin XX, Silva SS, Sarrouh BF, Cai AH, Zhou YH, Ke J, Qiu for industrial applications. Curr Opin Biotechnol 36:65–72
X (2009b) Novel isolates for biological detoxification of lignocellu- Zhu HY, Xu H, Dai XY, Zhang Y, Ying HJ, Ouyang PK (2010)
losic hydrolysate. Appl Biochem Biotechnol 152(2):199–212 Production of D-arabitol by a newly isolated Kodamaea ohmeri.
Zhang JM, Geng AL, Yao CY, Lu YH, Li QB (2012) Xylitol production Bioprocess Biosyst Eng 33(5):565–571
from d-xylose and horticultural waste hemicellulosic hydrolysate by Zinjarde SS (2014) Food-related applications of Yarrowia lipolytica.
a new isolate of Candida athensensis SB18. Bioresour Technol 105: Food Chem 152:1–10
134–141
Zhang B, Li L, Zhang J, Gao X, Wang D, Hong J (2013) Improving
ethanol and xylitol fermentation at elevated temperature through
substitution of xylose reductase in Kluyveromyces marxianus. J
Ind Microbiol Biotechnol 40(3–4):305–316
Zhang J, Zhang B, Wang D, Gao X, Hong J (2014) Xylitol production at Publisher’s note Springer Nature remains neutral with regard to
high temperature by engineered Kluyveromyces marxianus. jurisdictional claims in published maps and institutional affiliations.
Bioresour Technol 152:192–201

You might also like