Está en la página 1de 60

Alimentos genéticamente

modificados
Profesora: Bravo Ayala, Marta Margot

Estudiantes:
❏ Flores Velasquez, Gabriela
❏ Oré Maldonado, Kevin Anthony
❏ Valdez Chirinos, Irene
2022 - I
RESUMEN

Con el objetivo de conocer la caracterización, función y ventajas de un transgénico, se


tomó una serie de definiciones y ejemplos que clarifican lo necesario sobre estos
alimentos genéticamente modificados, ya que son encargados de proporcionar
características concretas a los vegetales o frutas que desean emplear. En el caso de las
plantas, por ejemplo, se han alterado sus genes para hacerlas resistentes a
determinadas plagas, para intensificar la producción en el cultivo.En la actualidad, la
soja, algún tipo de patatas y el maíz transgénicos se cultivan en gran parte del mundo y
se han incorporado al consumo general, tanto en su forma natural como en la
producción de alimentos envasados y así como estos se emplean otros más que poseen
un fin determinado dentro de su estudio.

2
INTRODUCCIÓN
La biotecnología moderna o ingeniería genética ha presentado avances
notables desde hace poco más de treinta años. En la actualidad, el mundo es
testigo de los resultados de estas innovaciones a través de la creación de
plantas o animales con características nuevas provenientes de la manipulación
de sus genes, esta cruza barreras naturales entre especies, que no serían
transgredidas siguiendo una evolución natural. De esta manera se crean, en
laboratorios, organismos con nuevas características, denominados
organismos vivos modificados (OVM), conocidos también como transgénicos
[1].

3
ALIMENTOS GENÉTICAMENTE MODIFICADOS
Los alimentos genéticamente modificados (GM) tienen un ADN peculiar usando genes de
otras plantas o animales. Los científicos toman el gen de un rasgo deseado de una planta o
animal e insertan ese gen dentro de una célula de otra planta o animal.
El proceso para crear alimentos GM (transgénicos) es diferente a la cría selectiva. Esta
involucra la selección de plantas o animales con los rasgos deseados y su crianza que con el
tiempo resulta en la descendencia con los rasgos deseados [2].

BENEFICIOS:

● Alimentos más nutritivos y apetitosos


● Plantas resistentes a la sequía y a las enfermedades, que requieren menos recursos
ambientales (como agua y fertilizante)
● Menos uso de pesticidas
● Aumento en el suministro de alimentos a un costo reducido y con una mayor vida útil
● Crecimiento más rápido en plantas y animales
● Alimentos medicinales que se podrían utilizar como vacunas u otros medicamentos

4
ALIMENTOS GENÉTICAMENTE MODIFICADOS
Por su parte, en los OGM de segunda generación se pretende
mejorar la composición del producto o su valor nutritivo, por
medio de la inclusión de vitaminas, atributos medicinales,
eliminación de alérgenos naturales, modificación del contenido de
proteínas, aceites, etc [3].

Algunos ejemplos y para qué fueron tratados químicamente.

● Soja. Modificación en la semilla, para ser más resistente a los


herbicidas.
● Maíz. Genes insertados en el genoma de la planta, para
hacerlo más resistente a insectos.
● Carnes. Aumentar el tamaño y el peso de los animales, y
acelerar la velocidad de su crecimiento.
● Trigo. Hacerlo más resistente antes sequías.

5
APLICACIONES

Las enzimas alimentarias naturales a menudo tienen limitaciones en condiciones de


procesamiento de alimentos sofisticadas y extremas. Las enzimas modificadas
genéticamente están diseñadas para mejorar o alterar las propiedades enzimológicas y/o
aumentar la pureza y el rendimiento de la expresión, cambios que pueden atribuirse a la
secuencia de aminoácidos alterada, resultante de la manipulación de la secuencia del gen.
Este artículo revisa las estrategias básicas para producir enzimas modificadas
genéticamente, como carbohidrasas, proteasas, lipasas, etc., y su uso o uso potencial en el
procesamiento de alimentos. Aunque existen desafíos y preocupaciones de seguridad para el
uso de proteínas recombinantes en el procesamiento de alimentos, las enzimas modificadas
genéticamente son prometedoras debido a los beneficios potenciales para la industria
alimentaria, los consumidores y el medio ambiente.

6
APLICACIONES

Para investigar la importancia de la selectividad del sustrato para la funcionalidad de la xilanasa en la


elaboración de pan, el sitio de unión secundario (SBS) de las xilanasas de Bacillus subtilis (XBS) y
Pseudoalteromonas haloplanktisfue modificado. Esto dio como resultado dos xilanasas con actividad
relativa aumentada hacia arabinoxilano de trigo no extraíble en agua (WU-AX) en comparación con
arabinoxilano de trigo extraíble en agua, es decir, una selectividad de sustrato aumentada, sin cambiar otras
propiedades bioquímicas. La adición de ambas xilanasas modificadas en la elaboración de pan resultó en un
aumento de los volúmenes de pan en comparación con los tipos silvestres cuando se usaba harina débil.
Además, el aumento de volumen máximo se alcanzó con una dosis más baja del mutante en comparación con
el XBS de tipo salvaje. Las xilanasas modificadas pudieron solubilizar más WU-AX y redujeron más el grado
medio de polimerización del arabinoxilano soluble en la masa durante la fermentación. Esto posiblemente
permitió una liberación adicional de agua, lo que podría ser responsable del aumento de los volúmenes de
pan. Funcionalidad SBS alterada y, como resultado,
7
APLICACIONES
Dentro de la literatura, se ha demostrado que la
fracción de lectina (TBLF) del Frijol Tepary
(Phaseolus acutifolius ) se une específicamente e
induce la muerte celular de diferentes tipos de
células cancerosas. Sin embargo, no se ha
podido desarrollar un fármaco debido a que la
producción es costosa, lenta y presenta bajos
rendimientos.

Con lo cual, el trabajo tiene como objetivo planificar una alternativa para producir una lectina bioactiva por el proceso de
rizosecreción a través de exudados de raíces en plantas genéticamente modificadas. Entonces, se procedió a amplificar
los transcritos de frijol Tepary mediante cebadores degenerados, cuyos productos fueron secuenciados. Entre las
secuencias resultantes, se logró la dilucidación de las lectinas presentes en TBLF.

Su secuencia codificante fue introducida en la planta P. acutifolius utilizando Agrobacterium tumefaciens, para, luego,
hacerlas crecer in vitro. Una vez que las raíces crecieron, las plantas fueron transferidas a condiciones hidropónicas y se
analizaron los exudados de las raíces.

En los resultados se identificó la presencia de una lectina cisgénica glicosilada con actividad biológica, confirmando la
eficacia de la alternativa propuesta para la producción sintética y purificación de esta lectina.

8
APLICACIONES
Los terpenos, que se encuentran en las uvas, le
otorgan el sabor y aroma característicos a los
vinos, y sirven para identificar la calidad de los
productos vitivinícolas. Los terpenos se derivan a
través de las vías del mevalonato (MVA) y del
fosfato de metileritritol (MEP). En las células
vegetales se pueden manifestar ambas vías; pero
la levadura y otros hongos solo albergan la vía
MVA, mientras que las bacterias producen
terpenos exclusivamente a través de la vía MEP.

En el proceso de fermentación, como la vinificación, muchas cepas de S. cerevisiae pueden hidrolizar enzimáticamente los
terpenos unidos en el mosto de uva y así liberar las contrapartes volátiles libres en el vino. En los últimos años, la
producción de terpenos a través de fábricas de células microbianas se ha convertido en una alternativa prometedora por
manifestar un rápido crecimiento, ser ahorrador de terreno y controlable. Los productos de terpenos de microorganismos
modificados han mostrado un gran potencial en aplicaciones alimentarias que incluyen agentes aromatizantes, agentes
edulcorantes , conservantes e ingredientes alimentarios funcionales.

El trabajo de revisión tiene por objetivo investigar las vías MVA y MEP de la biosíntesis de terpenos y la ingeniería
metabólica de S. cerevisiae para promover la producción de terpenos.

9
CONCLUSIONES
La ingeniería genética promete traer importantes y rápidos avances en la producción de alimentos,
con el objetivo de lograr resolver los diversos problemas en la alimentación, como la desnutrición, el
hambre y la pobreza de una sociedad mundial que se encuentra en continuo crecimiento, mediante el
aumento de la producción, la mejora de la calidad y seguridad de los alimentos. Por lo tanto, es
necesario considerar con mucho cuidado los posibles efectos adversos para garantizar que esta
herramienta tecnológica sea un beneficio para la humanidad. A lo largo de estos años, en la población
han aparecido controversias sobre los alimentos modificados genéticamente, generados por un
escaso conocimiento objetivo del tema; por lo tanto, también se debe tener la intención de disipar los
temores de los consumidores mediante la aplicación de técnicas fiables, proporcionando mayor
evidencia experimental y promoviendo debates y seminarios para aumentar la confianza.

APORTES
Mejoramiento en la calidad, sabor, velocidad de producción, etc de los alimentos. De poder tratar
adecuadamente los efectos secundarios, y mejorando el proceso de los alimentos modificados
genéticamente, se puede solucionar los problemas de plagas y escasez de varios alimentos.

10
REFERENCIAS BIBLIOGRÁFICAS
1. Regulación De Los Transgénicos En El Perú. [citado el 22 de julio de 2022]. Disponible en:
https://www.minam.gob.pe/wp-content/uploads/2015/08/transgenicos_FINALpdf.pdf
2. Reyes S. María Soledad, Rozowski N Jaime. ALIMENTOS TRANSGÉNICOS. Rev. chil. nutr. [Internet]. 2003
Abr [citado 2022 Jul 22] ; 30( 1 ): 21-26. Disponible en:
http://www.scielo.cl/scielo.php?script=sci_arttext&pid=S0717-75182003000100003&lng=es.
http://dx.doi.org/10.4067/S0717-75182003000100003.
3. Spendeler Liliane. Organismos modificados genéticamente: una nueva amenaza para la seguridad
alimentaria. Rev. Esp. Salud Publica [Internet]. 2005 Abr [citado 2022 Jul 22] ; 79( 2 ): 271-282. Disponible
en: http://scielo.isciii.es/scielo.php?script=sci_arttext&pid=S1135-57272005000200013&lng=es.
4. MARTÍNEZ-ALARCÓN, Dania, et al. Rhizosecretion of a cisgenic lectin by genetic manipulation of Tepary
bean plants (Phaseolus acutifolius). Journal of Biotechnology. [Internet]. 2019. [Consultado 22 julio 2022].
vol. 306, p. 100013. Disponible en: https://www.sciencedirect.com/science/article/pii/S2590155919300095
5. LIANG, Zijian, et al. Genetic engineering of yeast, filamentous fungi and bacteria for terpene production and
applications in food industry. Food Research International, 2021, vol. 147, p. 110487. Disponible en:
https://www.sciencedirect.com/science/article/pii/S0963996921003860
6. 1. Zhang Y, Geary T, Simpson BK. Genetically modified food enzymes: a review. Curr Opin Food Sci. 2019 Feb
1;25:14–8.
7. 1. Leys S, Pauly A, Delcour JA, Courtin CM. Modification of the secondary binding site of xylanases illustrates
the impact of substrate selectivity on bread making. J Agric Food Chem [Internet]. 2016 Jul 6 [cited 2022 Jul
22];64(26):5400–9. Available from: https://pubs.acs.org/doi/abs/10.1021/acs.jafc.6b01473

11
Accepted Manuscript

Title: Genetically modified food enzymes: A review

Authors: Yi Zhang, Timothy Geary, Benjamin K Simpson

PII: S2214-7993(18)30091-2
DOI: https://doi.org/10.1016/j.cofs.2019.01.002
Reference: COFS 426

To appear in:

Please cite this article as: Zhang Y, Geary T, Simpson BK, Genetically
modified food enzymes: A review, Current Opinion in Food Science (2019),
https://doi.org/10.1016/j.cofs.2019.01.002

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
Genetically modified food enzymes: A review

Yi Zhang a, Timothy Geary b, Benjamin K. Simpson a, *

a
Department of Food Science and Agricultural Chemistry, McGill University, Ste-Anne-de-Bellevue,
Quebec H9X 3V9, Canada

PT
b
Institute of Parasitology, McGill University, Ste-Anne-de-Bellevue, Quebec H9X 3V9, Canada

RI
Corresponding author:
Benjamin K. Simpson. E-mail: benjamin.simpson@mcgill.ca

SC
Other authors:
Yi Zhang. E-mail: yi.zhang10@mail.mcgill.ca

U
Timothy Geary. E-mail: timothy.g.geary@mcgill.ca

Highlights
N
A
 Food enzyme modification is developed by advanced genetic techniques
M

 Genetically modified food enzymes can exhibit improved catalytic efficiency, stability, substrate
specificity

D

Most genetically modified food enzymes are designed to be applied in food processing involving
carbohydrates and lipids
TE

Abstract
EP

Natural food enzymes often have limitations in sophisticated and extreme food processing conditions.
Genetically modified enzymes are designed to improve or alter the enzymological properties and/or
increase purity and yield of expression, changes which can be attributed to the altered amino acid sequence,
CC

resulting from gene sequence manipulation. This article reviews the basic strategies for producing
genetically modified enzymes, such as carbohydrases, proteases, lipases, etc., and their use or potential use
A

in food processing. Although there are challenges and safety concerns for using recombinant proteins in
food processing, genetically modified enzymes are promising because of potential benefits for the food
industry, consumers and the environment.

Keywords
Enzyme; food; mutagenesis; genetic modification

Page 1 of 12
1. Introduction
According to the definition of genetically modified organisms (GMOs) by World Health Organization
(WHO), the term “genetically modified enzymes” can be used to refer to situations in which the DNA
encoding the enzyme of interest has been artificially altered. Non-genetically modified enzymes remain the
major source for the food enzyme market, which includes endogenous enzymes produced from animals,
plants and microorganisms. Modern food processing tends to be sophisticated and precise, and sometimes

PT
requires extreme conditions such as high temperature, high pressure, extreme pH, salinity and the use of
solvents. However, native enzymes often have limitations that need to be improved to suit specific food

RI
processing environments. Genetic modification aims to rationally improve enzymes to enhance
characteristics such as purity, yield, specificity, catalytic efficiency, stability, surface property and

SC
multifunctionality, as well as enable cost-effective production and sustainable development for food
processing.
The strategy for producing genetically modified food enzymes is based on genetic engineering and

U
requires comprehensive understanding and integration of bioinformatics, enzymology, biophysical

N
chemistry and molecular biology, etc. Most genetically modified enzymes are expressed in well-established
standard microbial model systems such as E. coli and Pichia pastoris. There are a few strategies for
A
producing genetically modified enzymes depending on information known about the target enzyme and the
M

purpose of its use:


(i) Gene sequence optimization is commonly used for heterologous expression of recombinant
enzymes to optimize codon bias of the host cell, as well as to limit mRNA secondary structure
D

that could inhibit translation [1]. These changes usually increase yield, while the amino acid
TE

sequence remains the same.


(ii) Gene truncation or fusion requires knowledge of the relationship between amino acid sequence
EP

and enzyme function. Gene truncation involves eliminating DNA sequences that are not
functionally important, enabling the enzyme to be expressed in higher yield or with higher
activity; gene fusion constructs a gene with 2 or more enzyme DNA sequences so that the
CC

recombinant enzyme has multiple functions [2].


(iii) Site-directed mutagenesis is a rational design process based on in-depth knowledge of the target
enzyme in terms of structure, catalytic mechanism, active site, etc. DNA sequence(s) encoding
A

specific amino acid(s) that are related to activity or structural stability is (are) replaced, inserted
or deleted to produce enzymes with desired or improved properties [3].
(iv) Directed evolution, a non-rational design process that requires no prior knowledge of the target
enzyme, is an in vitro accelerated process that mimics natural evolution for generating enzymes.
It consists of two key processes: genetic diversity generation for constructing combinatorial

Page 2 of 12
libraries, and high-throughput screening or selection. The first process can be achieved by
random mutagenesis using error-prone PCR techniques, or gene recombination using
representative DNA-shuffling techniques [4]. The second process for identifying or isolating
improved variants may be quite difficult to perform, so that developing efficient screening
protocols has been a primary goal of innovation [5].
(v) Semi-rational design involves mutations based on sequence, structure or computational models,

PT
followed by small-scale mutagenesis and screening methods [3]. An example of semi-rational
design is site saturation mutagenesis, in which a mutant library is created containing all possible

RI
substitutions at one or more target positions in a gene sequence [6].
This review article summarizes novel food enzymes produced by genetic modification, especially

SC
using strategies (ii)-(v), and discusses their improved properties as well as applications and potential
applications in food processing.

U
2. Genetically modified carbohydrases/glycosidases

N
Carbohydrases are the most widely-used enzymes in the food industry, being employed in baking,
brewing, sweetener production, etc. Some of these enzymes break down polysaccharides into simple sugars.
A
Alpha-amylase (EC 3.2.1.1) is ubiquitous in the starch industry; the commercially available Bacillus
M

licheniformis α-amylase is thermostable but acid-sensitive. Novel Bacillus licheniformis α-amylases


obtained using directed evolution by replacing active site domain His residues with Arg and Asp residues
had higher activity at pH 4.5 than the wild type enzyme [7,8], which enhances starch liquefaction,
D

saccharification and fermentation processes in which the pH of starch slurry is usually < 6.0 [9]. A modified
TE

Rhizopus oryzae α-amylase obtained using site-saturation mutagenesis of His 286 had a higher optimum
temperature (60 oC) and lower optimum pH (4.0–4.5) than the wild-type enzyme [10], properties better
suited for use in high maltose syrup production. A mutant Aspergillus aculeatus β-glucosidase (EC 2.3.1.21)
EP

obtained using site-saturation mutagenesis had improved hydrolytic efficiency especially to cellobiose, and
was used to accelerate the saccharification of alkaline-pretreated bagasse [11]. The substrate specificity of
CC

Thermotoga maritima β-glucosidases after genetic modification was enhanced for quercetin glucosides [12],
suggesting its usage in producing aglycones that have higher pharmaceutical activity compared with the
substrate. Endo-β-1,4-xylanase (EC 3.2.1.8) cleaves the β-1,4 glycosidic linkages in xylans. A variant of
A

this enzyme generated using site-directed mutagenesis exhibited higher specific activity and was utilized in
the production of xylooligosaccharides from wheat straw under thermal and alkaline conditions (50–65 oC,
pH 7–10) [13]. Novel endo-β-1,4-xylanases with modification in secondary binding sites also showed
increased specificity toward water-insoluble, but not water-soluble, wheat arabinoxylan, and its application
in bread making resulted in increased loaf volumes [14]. An engineered β-1,3-1,4-glucanase (EC 3.2.1.73),

Page 3 of 12
hydrolyzing β-1,3-1,4-glucan usually found in avena, rice, oats and barley, had improved catalytic
efficiency, thermostability and halostability following replacement of hydrophobic Lys 48 with Ala and
Leu [15]. Bacillus spp. is the main source of β-1,3-1,4-glucanase used in malt extract production in the
brewing industry, and novel β-1,3-1,4-glucanases were produced by replacing Lys with Ser to form rigid
β-sheet structures. The modified glucanases functioned effectively under industrial operating conditions
with improved optimal temperature and thermostability [16].

PT
Some types of carbohydrases hydrolyze disaccharides into monosaccharides. For example, a novel
Saccharomyces cerevisiae invertase (EC 3.2.26) was engineered to have optimal pH and stability ranges

RI
for the breakdown of sucrose into glucose and fructose; the enzyme was modified by the substitution of
hydrophilic residues in the active site region or peripheral loops with hydrophobic amino acids [17].

SC
Another genetically modified invertase with high transfructosylating activity was used for simple and
efficient production of prebiotic fructooligosaccharides [18]. Commercial β-galactosidase or lactase (EC
3.2.1.23) is commonly applied in lactose-free milk product processing, in which the process temperature is

U
relatively low. A variant of β-galactosidase obtained using directed evolution had high activity in industrial-

N
type conditions for milk processing, i.e., substrate lactose, buffer pH 6.75, and 8 °C [19]. To improve the
transglycosylating activity of this enzyme, a thermoresistant Thermotoga maritima β-galactosidase
A
modified using rational design was obtained with better specificity for the biosynthesis of galacto-
M

oligosaccharides, a prebiotic compound [20].

3. Genetically modified proteases/peptidases


D

The major use of proteases in the food industry is hydrolysis of protein matrices to enhance flavor, texture
TE

or functional properties in diary, meat and fish products. Commonly used food proteases are derived from
animals (such as trypsin, chymotrypsin, chymosin/rennet, pepsin), plants (such as papain, bromelain, ficin),
EP

and microorganism (such as the microbial acid protease Flavourzyme® from Aspergillus oryzae and
proteinase A from S. cerevisiae; bacteria neutral proteases produced by Bacillus circulans BM15 and
Streptomyces nogalator AC 80; and bacteria alkaline proteases produced by Aspergillus niger and Bacillus
CC

subtilis).
Unlike food carbohydrases and lipases, animal/plant-derived food proteases are generally less
commonly found in genetically modified forms, probably because the versatile properties of the native
A

proteases are adequate to suit most required temperature and pH conditions in food processing, and their
catalysis mechanism has been well investigated. It could also be due to the very ample availability of
microbial proteases for industrial uses. Another reason could be the limitations posed for efficient
expression of proteases derived from animal or plant sources in microbial systems.
Recent research on modified food proteases from Aspergillus and Bacillus species indicates that

Page 4 of 12
engineered metalloproteases obtained through site-saturation mutagenesis of His224 had improved affinity
for substrate, making the synthesis of Z-aspartame more cost-effective [21]. An acid protease from a mutant
A. oryzae strain was produced using solid state fermentation with potato pulp powder, with enhanced
glycine releasing activity [22]. A truncated neutral protease from A. oryzae had optimum pH of 8.0 and
optimum temperature of 55 oC, and its enzymological characterization suggested that it was efficient in
antihypertensive peptide production, debittering, and food oil processing [23].

PT
Bacillus spp. are the main producers of alkaline proteases with high thermal and pH stabilities. Subtilisin
nattokinase, a serine protease from B. subtilis var. natto, was modified by site-directed mutagenesis to

RI
obtain higher specific activity and oxidative stability, and may be useful as a potential cardiovascular drug
due to its strong fibrinolytic activity [24]. The cold activity at 10 oC and alkali-resistant properties of a

SC
Bacillus alcalophilus alkaline protease were improved through directed evolution using error-prone PCR
[25] for use in cold-temperature food processing. Similarly, an alkaline serine protease from mesophilic
Bacillus pumilus was engineered to have increased hydrolytic efficiency at 15 oC without compromising

U
thermostability [26]. Modified protases are also applied in the detergent industry. An alkaline protease

N
isolated from an oil-polluted mud flat metagenome by random mutagenesis displayed robust compatibility
with laundry detergents at low temperature (30 °C) and alkaline pH range (pH 8.0–11.0) [27].
A
M

4. Genetically modified lipases


Lipase (EC 3.1.1.3) is the most common biocatalyst for lipid (fats and oils) modification. Bacillus lipases
display activities over wide pH and temperature ranges, and some possess fatty acid specificity. Lipase A
D

from B. subtilis was expressed as a fusion protein with cell wall mannoprotein Pir4 resulting in an
TE

immobilized lipase, and was used as a leavening agent to enhance rheological and aromatic properties of
bread [28]. The secretion of Rhizopus oryzae lipase was improved by rational design of the N-glycosylation
EP

sites, which could meet the industrial demand for Rhizopus lipase, especially for the edible oil and fat
industries [29]. Candida rugosa lipase isozymes modified by site-directed mutagenesis had high catalytic
efficiency for producing fatty acid methyl esters and diglycerides as food emulsifiers, as well as for the
CC

conversion of crude Jatropha curcas seed oil into biodiesel [30,31]. Malassezia globose lipase with specific
activity especially on mono- and diacylglycerol was modified with enhanced thermostability to meet
industrial-scale requirements, making it a potential biocatalyst for synthesis of diacylglycerols in edible oils
A

with health benefits [32]. Engineered thermostable T1 lipases had low activity toward long-chain
triacylglycerols; thus, they are potential biocatalysts to enhance flavor in dairy products by generation of
short-chain fatty acids from milk fats [33]. A Thermomyces lanuginosus lipase modified using semi-rational
design was targeted to enhance methanol tolerance for potential use in biofuel production from waste food
oils and grease [34].

Page 5 of 12
5. Other genetically modified food enzymes
A D-psicose 3-epimerase (EC 5.1.3.31) modified by site-directed mutagenesis displayed high substrate-
binding affinity, catalytic efficiency and thermostability in catalyzing the isomerization of D-fructose to D-
psicose, an ultra-low-calorie sweetener with desirable physiological properties [35]. Similarly, engineered
Caldicellulosiruptor saccharolyticus cellobiose 2-epimerases (EC 5.1.3.11) showed increased specific
activity to directly convert lactose into lactulose without co-production of epilactose, and its application

PT
will boost the commercial availability of lactulose, a non-digestible disaccharide used as a prebiotic food
additive and medicine [36]. A novel trehalose synthase (EC 2.4.1.245) from Picrophilus torridus was

RI
prepared by replacing Pro residues to increase thermostability and catalytic efficiency, and was used to
convert maltose from low-value agricultural products such as sweet potato starch to high-value trehalose

SC
[37]. Yersinia phytases (EC 3.1.3.8) were genetically manipulated using site-directed mutagenesis to
improve pepsin and trypsin resistance and thermostability, enabling potential use for efficient hydrolysis of
phytic acid to inorganic phosphate to improve nutrient uptake from foods [38]. A novel di-D-fructose

U
dianhydride I (DFA I)-forming inulin fructotransferase was engineered using rational design to improve

N
its catalytic behavior, including activity and thermostability, so that it can produce prebiotic DFA I in
industrial processes using inulin as substrate [39]. Nitrilase (EC 3.5.5.1), which converts nitriles into
A
corresponding carboxylic acids, was genetically modified to achieve higher activity for stereospecific
M

production of (R)-(−)-mandelic acid, useful in pharmaceutical production [40]. A mutant Aspergillus niger
α-L-rhamnosidase with improved thermostability and substrate affinity reduced bitterness caused by
naringin in orange juice [41]. 1,4-α-glucan branching enzyme (EC 2.4.1.18) was engineered by Met349
D

mutation to achieve higher activity for the formation of α-1,6-glucosidic linkages, and could form highly
TE

branched potato starch [42]. A novel Bacillus stearothermophilus NO2 cyclodextrin glycosyltransferase
was produced through iterative saturation mutagenesis around the catalytic residues, and was used to
EP

produce 2-O-α-D-glucopyranosyl-L-ascorbic acid, which has great advantages in food processing due to its
stability and facile degradation to L-ascorbic and glucose [43].
CC

6. Safety concerns
Genetically modified food enzymes are currently produced from GMOs. Safety concerns have been
raised regarding potential contamination of food with bacterial toxins or mycotoxins, allergens or
A

uncharacterized extraneous substances as impurities [44,45]. Genetic modification of enzymes may also
change their allergenic properties, posing new potential health risks. For instance, type I sensitisation was
found in a study of 813 exposed industrial workers using genetically modified enzymes [46]. Thus, prior to
marketing, genetically modified food enzymes need approval from various regulatory bodies such as the
US Food and Drug Administration, the Association of Manufacturers and Formulators of Enzyme Products

Page 6 of 12
and the European Food Safety Authority, through processes that vary in different countries. In addition,
ethical and religious concerns have been raised for genetically modified enzymes. For instance, it has been
suggested that raw materials or ingredients used in fermentation of microorganisms to produce enzymes
should be halal [47].

7. Conclusion

PT
Genetically modified food enzymes are typically better suited for specific industrial applications than
their native counterparts, and research on their enzymological properties in comparison to wild-type

RI
enzymes enable us to better understand how to optimize structure-function relationships. Currently, novel
genetically modified food enzymes are mostly used for applications in food processing involving

SC
carbohydrates, followed by lipids. Although there are challenges for safe use, genetic modification
strategies for the development of novel food enzymes are highly promising for the future, and we expect
that more genetically modified enzymes will be introduced in various aspects of food processing.

U
Conflict of Interest
The authors declared no conflict of interest.
N
A
M
D
TE
EP
CC
A

Page 7 of 12
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest

References

PT
1. Rosano GL, Ceccarelli EA: Recombinant protein expression in Escherichia coli: advances and
challenges. Front Microbiol 2014, 5:1-17.

RI
2. Dediu D: An introduction to genetics for language scientists. Cambridge, UK: Cambridge University
Press; 2015.

SC
3. Hua W, El Sheikha AF, Xu J: Molecular techniques for making recombinant enzymes used in food
processing. In Molecular Techniques in Food Biology: Safety, Biotechnology, Authenticity and
Traceability. Edited by El Sheikha A F, Levin R E, Xu J. John Wiley & Sons; 2018:95-112.

U
* This book chapter described in detail the molecular strategies to produce recombinant food enzymes.

N
The two core steps in directed evolution, gene diversification and screening/selection are well discussed
with examples. The procedures and techniques used in rational design are also reviewed.
A
4. Labrou NE: Random mutagenesis methods for in vitro directed enzyme evolution. Curr Protein Pept
M

Sci 2010, 11:91-100.


5. Packer MS, Liu DR: Methods for the directed evolution of proteins. Nat Rev Genet 2015, 16:379.
6. Özgün GP, Ordu EB, Tütüncü HE, Yelboğa E, Sessions RB, Gül Karagüler N: Site saturation
D

mutagenesis applications on Candida methylica formate dehydrogenase. Scientifica 2016: 1-7.


TE

7. Liu Y, Fan S, Liu X, Zhang Z, Wang J, Wang Z, Lu F: A highly active alpha amylase from Bacillus
licheniformis: directed evolution, enzyme characterization and structural analysis. J
EP

Microbiol Biotechnol 2014, 24:898-904.


8. Liu Y, Huang L, Jia L, Gui S, Fu Y, Zheng D, Guo W, Lu F: Improvement of the acid stability of
Bacillus licheniformis alpha amylase by site-directed mutagenesis. Process Biochem 2017,
CC

58:174-180.
9. Cates ES, Dinwiddie JA, Aux G, Batie C, Crabb G: Process for starch liquefaction and fermentation.
U.S. Patent 7,727,726, Washington, DC; 2010.
A

10. Li S, Yang Q, Tang B, Chen A: Improvement of enzymatic properties of Rhizopus oryzae α-amylase
by site-saturation mutagenesis of histidine 286. Enzyme Microb Technol 2018, 117:96-102.
11. Baba Y, Sumitani J-i, Tanaka K, Tani S, Kawaguchi T: Site-saturation mutagenesis for β-glucosidase
1 from Aspergillus aculeatus to accelerate the saccharification of alkaline-pretreated bagasse.
Appl Microbiol Biotechnol 2016, 100:10495-10507.

Page 8 of 12
12. Sun H, Xue Y, Lin Y: Enhanced catalytic efficiency in quercetin-4′-glucoside hydrolysis of
Thermotoga maritima β-glucosidase A by site-directed mutagenesis. J Agric Food Chem 2014,
62:6763-6770.
13. Faryar R, Linares-Pastén JA, Immerzeel P, Mamo G, Andersson M, Stålbrand H, Mattiasson B,
Karlsson EN: Production of prebiotic xylooligosaccharides from alkaline extracted wheat
straw using the K80R-variant of a thermostable alkali-tolerant xylanase. Food Bioprod

PT
Process 2015, 93:1-10.
14. Leys S, Pauly A, Delcour JA, Courtin CM: Modification of the secondary binding site of xylanases

RI
illustrates the impact of substrate selectivity on bread making. J Agric Food Chem 2016,
64:5400-5409.

SC
15. Lee JM, Moon SY, Kim Y-R, Kim KW, Lee B-J, Kong I-S: Improvement of thermostability and
halostability of β-1, 3-1, 4-glucanase by substituting hydrophobic residue for Lys48. Int J Biol
Macromol 2017, 94:594-602.

U
16. Niu C, Zhu L, Zhu P, Li Q: Lysine-based site-directed mutagenesis increased rigid β-sheet structure

N
and thermostability of mesophilic 1, 3–1, 4-β-glucanase. J Agric Food Chem 2015, 63:5249-
5256.
A
17. Mohandesi N, Haghbeen K, Ranaei O, Arab SS, Hassani S: Catalytic efficiency and thermostability
M

improvement of Suc2 invertase through rational site-directed mutagenesis. Enzyme Microb


Technol 2017, 96:14-22.
18. Marín-Navarro J, Talens-Perales D, Polaina J: One-pot production of fructooligosaccharides by a
D

Saccharomyces cerevisiae strain expressing an engineered invertase. Appl Microbiol Biotechnol


TE

2015, 99:2549-2555.
19. Rentschler E, Schwarz T, Stressler T, Fischer L: Development and validation of a screening system
for a β-galactosidase with increased specific activity produced by directed evolution. Eur Food
EP

Res Technol 2016, 242:2129-2138.


20. Talens-Perales D, Polaina J, Marín-Navarro J: Structural dissection of the active site of Thermotoga
CC

maritima β-Galactosidase identifies key residues for transglycosylating activity. J Agric Food
Chem 2016, 64:2917-2924.
21. Zhu F, Jiang T, Wu B, He B: Enhancement of Z-aspartame synthesis by rational engineering of
A

metalloprotease. Food Chem 2018, 253:30-36.


** The authors modified a metalloprotease to improve affinity for producing Z-aspartame, a common food
sweetness additive. A computational strategy combining molecular dynamics simulation of the enzyme-
substrate complex with binding free energy calculations was established for engineering the enzyme.
22. Murthy PS, Kusumoto K-I: Acid protease production by Aspergillus oryzae on potato pulp powder

Page 9 of 12
with emphasis on glycine releasing activity: A benefit to the food industry. Food Bioprod
Process 2015, 96:180-188.
23. Ke Y, Huang W-Q, Li J-z, Xie M-q, Luo X-c: Enzymatic characteristics of a recombinant neutral
protease I (rNpI) from Aspergillus oryzae expressed in Pichia pastoris. J Agric Food Chem 2012,
60:12164-12169.
24. Weng M, Deng X, Bao W, Zhu L, Wu J, Cai Y, Jia Y, Zheng Z, Zou G: Improving the activity of the

PT
subtilisin nattokinase by site-directed mutagenesis and molecular dynamics simulation.
Biochem Biophys Res Commun 2015, 465:580-586.

RI
25. Liu Y, Zhang T, Zhang Z, Sun T, Wang J, Lu F: Improvement of cold adaptation of Bacillus
alcalophilus alkaline protease by directed evolution. J Mol Catal B Enzym 2014, 106:117-123.

SC
26. Zhao H Y, Feng H: Engineering Bacillus pumilus alkaline serine protease to increase its low-
temperature proteolytic activity by directed evolution. BMC Biotechnol 2018, 18:34.
27. Gong B L, Mao R Q, Xiao Y, Jia M L, Zhong X L, Liu Y, Xu P L, Li G: Improvement of enzyme

U
activity and soluble expression of an alkaline protease isolated from oil-polluted mud flat

N
metagenome by random mutagenesis. Enzyme Microb Technol 2017, 106:97-105.
28. Paciello L, Landi C, Orilio P, Di Matteo M, Zueco J, Parascandola P: Bread making with
A
Saccharomyces cerevisiae CEN. PK113‐ 5D expressing lipase A from Bacillus subtilis:
M

leavening characterisation and aroma enhancement. Int J Food Sci Technol 2015, 50:2120-
2128.
29. Yu X W, Yang M, Jiang C, Zhang X, Xu Y: N-Glycosylation engineering to improve the constitutive
D

expression of Rhizopus oryzae lipase in Komagataella phaffii. J Agric Food Chem 2017, 65:6009-
TE

6015.
30. Chang S W, Huang M, Hsieh Y H, Luo Y T, Wu T T, Tsai C W, Chen C S, Shaw J F: Simultaneous
EP

production of fatty acid methyl esters and diglycerides by four recombinant Candida rugosa
lipase’s isozymes. Food Chem 2014, 155:140-145.
31. Kuo T C, Shaw J F, Lee G C: Conversion of crude Jatropha curcas seed oil into biodiesel using
CC

liquid recombinant Candida rugosa lipase isozymes. Bioresour Technol 2015, 192:54-59.
32. Gao C, Lan D, Liu L, Zhang H, Yang B, Wang Y: Site-directed mutagenesis studies of the aromatic
residues at the active site of a lipase from Malassezia globosa. Biochimie 2014, 102:29-36.
A

33. Tang Q, Lan D, Yang B, Khan FI, Wang Y: Site‐ directed mutagenesis studies of hydrophobic
residues in the lid region of T1 lipase. Eur J Lipid Sci Technol 2017, 119:1600107.
34. Tian K, Tai K, Chua BJW, Li Z: Directed evolution of Thermomyces lanuginosus lipase to enhance
methanol tolerance for efficient production of biodiesel from waste grease. Bioresour Technol
2017, 245:1491-1497.

Page 10 of 12
35. Zhang W, Jia M, Yu S, Zhang T, Zhou L, Jiang B, Mu W: Improving the thermostability and catalytic
efficiency of the D-psicose 3-epimerase from clostridium bolteae ATCC BAA-613 using site-
directed mutagenesis. J Agric Food Chem 2016, 64:3386-3393.
36. Shen Q, Zhang Y, Yang R, Pan S, Dong J, Fan Y, Han L: Enhancement of isomerization activity and
lactulose production of cellobiose 2-epimerase from Caldicellulosiruptor saccharolyticus. Food
Chem 2016, 207:60-67.

PT
* This research developed a novel cellobiose 2-epimerase using a random mutagenesis and screening
protocol to enhance lactulose production. The modified enzyme overcame the drawbacks of the wild type

RI
counterpart, such as low enzyme activity and formation of epilactose as by-product.
37. Chou H H, Chang S W, Lee G C, Chen Y S, Yeh T, Akoh CC, Shaw J F: Site-directed mutagenesis

SC
improves the thermostability of a recombinant Picrophilus torridus trehalose synthase and
efficiency for the production of trehalose from sweet potato starch. Food Chem 2010,
119:1017-1022.

U
38. Niu C, Yang P, Luo H, Huang H, Wang Y, Yao B: Engineering of Yersinia phytases to improve

N
pepsin and trypsin resistance and thermostability and application potential in the food and
feed industry. J Agric Food Chem 2017, 65:7337-7344.
A
39. Yu S, Zhang Y, Zhu Y, Zhang T, Jiang B, Mu W: Improving the catalytic behavior of DFA I-forming
M

inulin fructotransferase from Streptomyces davawensis with site-directed mutagenesis. J Agric


Food Chem 2017, 65:7579-7587.
40. Liu Z Q, Zhang X H, Xue Y P, Xu M, Zheng Y G: Improvement of Alcaligenes faecalis nitrilase by
D

gene site saturation mutagenesis and its application in stereospecific biosynthesis of (R)-(−)-
TE

mandelic acid. J Agric Food Chem 2014, 62:4685-4694.


41. Li L J, Wu Z Y, Yu Y, Zhang L J, Zhu Y B, Ni H, Chen F: Development and characterization of an
α-L-rhamnosidase mutant with improved thermostability and a higher efficiency for
EP

debittering orange juice. Food Chem 2018, 245:1070-1078.


** The authors engineered an α-L-rhamnosidase to decrease bitterness in orange juice. The modified
CC

enzyme has improved thermostability, substrate affinity for naringin and better resistance to inhibition by
glucose. This research has practical value in citrus juice processing.
42. Liu Y, Ban X, Li C, Gu Z, Cheng L, Hong Y, Li Z: Met349 mutations enhance the activity of 1, 4-α-
A

glucan branching enzyme from Geobacillus thermoglucosidans STB02. J J Agric Food Chem
2017, 65:5674-5680.
43. Tao X, Wang T, Su L, Wu J: Enhanced 2-O-α-d-glucopyranosyl-L-ascorbic acid synthesis through
iterative saturation mutagenesis of acceptor subsite residues in Bacillus stearothermophilus
NO2 cyclodextrin glycosyltransferase. J Agric Food Chem 2018, 66:9052-9060.

Page 11 of 12
44. De Santis B, Stockhofe N, Wal J-M, Weesendorp E, Lallès J-P, van Dijk J, Kok E, De Giacomo M,
Einspanier R, Onori R: Case studies on genetically modified organisms (GMOs): Potential risk
scenarios and associated health indicators. Food Chem Toxicol 2018, 117:36-65.
45. Srivastava N: Production of food-processing enzymes from recombinant microorganisms. In
Enzymes in Food Biotechnology. Edited by Kuddus M. Academic Press; 2019:739-767.
* This book chapter summarized the technical and scientific involvement of microorganisms in food

PT
enzyme production, and the methodology for identifying genes encoding important enzymes, their cloning
and expression, and purification of recombinant food enzymes.

RI
46. Budnik LT, Scheer E, Burge PS, Baur X: Sensitising effects of genetically modified enzymes used in
flavour, fragrance, detergence and pharmaceutical production: cross-sectional study. Occup

SC
Environ Med 2017, 74:39-45.
47. Ermis E: Halal status of enzymes used in food industry. Trends Food Sci Technol 2017, 64:69-73.

U
N
A
M
D
TE
EP
CC
A

Page 12 of 12
Article

pubs.acs.org/JAFC

Modification of the Secondary Binding Site of Xylanases Illustrates


the Impact of Substrate Selectivity on Bread Making
Sofie Leys, Anneleen Pauly, Jan A. Delcour, and Christophe M. Courtin*
Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), KU Leuven,
Kasteelpark Arenberg 20, B-3001 Leuven, Belgium

ABSTRACT: To investigate the importance of substrate selectivity for xylanase functionality in bread making, the secondary
binding site (SBS) of xylanases from Bacillus subtilis (XBS) and Pseudoalteromonas haloplanktis was modified. This resulted in two
xylanases with increased relative activity toward water-unextractable wheat arabinoxylan (WU-AX) compared to water-extractable
wheat arabinoxylan, i.e., an increased substrate selectivity, without changing other biochemical properties. Addition of both
modified xylanases in bread making resulted in increased loaf volumes compared to the wild types when using weak flour.
Moreover, maximal volume increase was reached at a lower dosage of the mutant compared to wild-type XBS. The modified
xylanases were able to solubilize more WU-AX and decreased the average degree of polymerization of soluble arabinoxylan in
dough more during fermentation. This possibly allowed for additional water release, which might be responsible for increased
loaf volumes. Altered SBS functionality and, as a result, enhanced substrate selectivity most probably caused these differences.
KEYWORDS: xylanase, arabinoxylan, bread making, substrate selectivity, secondary binding site

■ INTRODUCTION
Endo-β-1,4-xylanases (EC 3.2.1.8), also referred to as xylanases,
of xylanases determines their efficiency in bread making as
well.15
are commonly used in bread making processes to enhance Recently, the existence of a secondary binding site (SBS)
dough manageability and bread quality. Although it is evident situated on the surface of the structural unit was discovered in
that their functionality is the result of their capacity to several single domain enzymes belonging to glycoside hydrolase
hydrolyze β-(1,4)-linkages in the backbone of arabinoxylan families (GH) 8, 10, and 11.16−18 Several functions have been
(AX), their mode of action in bread making is not yet fully proposed for these SBSs, including targeting of the enzyme
understood, despite much progress over the past decades.1−5 toward its substrate, guiding substrate into the active site
It has been hypothesized that water-unextractable wheat AX groove, substrate disruption, enhancing processivity, allosteric
(WU-AX) negatively affects bread volume due to its strong regulation, passing on reaction products, and anchoring the
water-holding capacity, while water-extractable wheat AX (WE- enzyme to the cell wall of its parent microorganism. These
AX) and solubilized wheat AX (S-AX) with a high molecular functions correspond well to the functions which are ascribed
weight have a positive effect since they form highly viscous to carbohydrate-binding modules (CBMs) in modular
aqueous solutions, stabilizing the dough foam structure.6−8 enzymes.19 Possibly, SBSs compensate for the lack of CBMs
Consequently, xylanases with a high substrate selectivity, i.e., in single domain enzymes.20 Site-directed mutagenesis to
the ratio of the capacity to solubilize WU-AX over the capacity modify this SBS resulted in an increased activity on water-
to hydrolyze WE-AX, are desired in bread making. Such unextractable substrates compared to water-extractable
endoxylanases preferentially solubilize WU-AX but hydrolyze ones,21−23 hence effectively changing the substrate selectivity
WE-AX and S-AX only to a minimal extent.9 of the enzyme.
To confirm this hypothesis, extensive research with xylanases In particular, the GH8 xylanase of Pseudoalteromonas
that differed in substrate selectivity was conducted.3,10−12 haloplanktis (XPH) and GH11 xylanase XynA of Bacillus
Disadvantageous for these studies was the use of xylanases of
subtilis (XBS) are both single domain enzymes which contain
different microbial origins which differed in more biochemical
an SBS. XBS has a β jelly roll fold structure, which is often
properties than only their substrate selectivity. The use of such
compared to a partially closed right hand, with an SBS located
xylanases made it difficult to investigate the influence of
substrate selectivity in bread making unambiguously. on the “knuckles” of the enzyme.24 XPH is a psychrophilic
Xylanase functionality in bread making is also influenced by enzyme with a (α/α)6-barrel structure (Figure 1).25 Since both
inhibitors. Three different types of xylanase inhibitors have enzymes show good performance in bread making, they are
been identified in wheat flour: Triticum aestivum xylanase ideal tools to investigate the influence of substrate selectivity on
inhibitor (TAXI), xylanase inhibiting protein (XIP), and xylanase functionality in bread making.
thaumatin-like xylanase inhibitor (TL-XI).13 Due to the
formation of enzyme−inhibitor complexes, inhibition sensitive Received: March 30, 2016
xylanases are less efficient than inhibition insensitive ones. Revised: June 8, 2016
These enzymes need higher dosages to ensure optimal Accepted: June 10, 2016
performance.14 Finally, the temperature and pH activity profile

© XXXX American Chemical Society A DOI: 10.1021/acs.jafc.6b01473


J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

Figure 1. On the left, the overall structures of the xylanases from Bacillus subtilis (XBS) (PDB 2QZ3) (A) and Pseudoalteromonas haloplanktis (XPH)
(PDB 2B4F) (B) in complex with xylooligosaccharides indicate the presence of a secondary binding site (SBS) on the surface of the enzyme. On the
right, the amino acids responsible for binding of the oligosaccharides to the SBS are indicated for XBS (A) and XPH (B). All figures were drawn
using PyMOL (http://pymol.sourceforge.net/).

The aim of this study is to validate the above-mentioned acids which were able to interact with the substrate by hydrophobic
hypothesis that substrate selectivity determines, at least in part, stacking interactions or hydrogen bonds were replaced by alanine to
the intrinsic quality of xylanases for bread making. This is done reduce their role in binding of substrate at the SBS (Figure 1).
by selectively modifying the substrate selectivity of XBS and Xylazyme AX tablets, liquid Azo-wheat AX, and xylooligosaccharides
up to xylohexaose (X6) were obtained from Megazyme (Bray, Ireland).
XPH xylanases through modification of their SBS, and studying
Xylazyme AX tablets and liquid Azo-wheat AX both contained highly
the impact thereof on the bread making process. This way, purified AX from wheat, while the xylooligosaccharides (purity >90%)
nearly identical xylanases are used, which differ only in were prepared from birchwood xylan. Two European wheat flours, free
substrate selectivity and not in other biochemical properties. from additives, were used in bread making. Crousti flour was a
Therefore, XBS and XPH were modified in their SBS and commercial bread wheat flour obtained from Dossche Mills (Deinze,
added to dough and bread followed by evaluating their impact Belgium). Soft wheat cultivar Claire from Limagrain (Rilland, The
on the AX population and aqueous extract viscosity of dough Netherlands) was conditioned to 16.0% moisture and subsequently
and bread samples. These results were compared to those milled with a Bühler MLU-202 laboratory mill (Bühler AG, Uzwil,
obtained with the wild-type enzymes. We here report on the Switzerland), yielding three break and three reduction fractions, which
outcome of this work. were combined.26 Protein contents [% dry matter (dm), N × 5.7]
were 13.2% and 10.6% for Crousti and Claire flour, respectively,

■ MATERIALS AND METHODS


Materials. Escherichia coli cells, transformed with expression
determined using an adaptation of the AOAC Official Method27 to an
automated Dumas protein analysis system (Vario Max Cube,
Elementar, Hanau, Germany). Ash contents (% dm) were 0.55%
plasmid pEXP5-CT-xyna, were available for heterologous expression and 0.48%, measured according to Approved Method 08-01.01,28
of XBS wild type (XBS WT) (UniProtKB P18429). A stop codon was while the total AX (% dm) and WE-AX contents (% dm) for Crousti
introduced in the plasmid after the last nucleotide encoding for the C- and Claire flour were 1.85% and 0.28% and 1.76% and 0.48%,
terminal amino acid of the native protein (W185).23 For expression of respectively. Fresh compressed baker’s yeast was from AB Mauri
XPH, E. coli cells transformed with expression plasmid pEXP5-CT-xph (Merelbeke, Belgium). Sodium chloride and sucrose used in the bread
which contained a de novo synthesized XPH wild-type (XPH WT) making trials were food grade. All chemicals, solvents, and reagents
gene (GenBank AJ427921.1) were also available.22 A triple mutant were purchased from Sigma-Aldrich (Bornem, Belgium) and were
plasmid of XBS (G56A-T183A-W185A) and a double mutant plasmid analytical grade, unless specified otherwise.
of XPH (W249A-Y315A), hereafter referred to as XBS 3A and XPH Recombinant Expression and Purification. The recombinant
2A, respectively, were constructed using a QuickChange site-directed expression of XBS, XPH, and their mutant variants in E. coli
mutagenesis kit (Stratagene, La Jolla, CA, USA) as described by BL21(DE3)pLysS cells was according to Van Craeyveld et al.30
Cuyvers et al.23 and were available in transformed E. coli cells. The Subsequently, the xylanases were purified by cation exchange
selection of amino acids for modification was based on the crystal chromatography as described elsewhere.29,30 The enzyme yields of
structures of XBS and XPH soaked with oligosaccharides.16,17 Amino recombinant XBS and XPH after purification were typically 50−90

B DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

mg/L and 100−180 mg/L of culture, respectively. Xylanase purity was sodium acetate (100 mM, pH 3.0, 4.0, and 5.0), sodium phosphate (20
verified by SDS−PAGE and silver staining performed on a mM, pH 6.0, 7.0, 8.0, and 9.0), sodium carbonate (50 mM, pH 10.0
PhastSystem Unit (GE Healthcare, Uppsala, Sweden) according to and 11.0), and potassium chloride/sodium hydroxide (20 mM, pH
GE Healthcare separation technique file 110 and development 12.0) buffers, all containing 0.50 mg/mL BSA. The enzyme activities
technique file 210, respectively.31 Protein concentration of the purified of these enzyme solutions on Xylazyme AX were measured at 30 °C as
enzymes was estimated by measuring the extinction at 280 nm in described above.
triplicate with a Nanodrop-1000 spectrophotometer (Thermo Fisher Inhibition Sensitivity. Wheat flour was suspended in sodium acetate
Scientific, Waltham, MA, USA) using the molar extinction coefficients buffer (100 mM) at pH 5.5 (mass ratio 1:10 flour:buffer). This
calculated with the ProtParam tool (http://expasy.org/tools/ suspension was shaken for 30 min at 7 °C (Laboshake, VWR
protparam.html). These calculations were based on the amino acid International, Leuven, Belgium) and centrifuged (10 min, 4000g, 7
sequences of the wild-type and mutant xylanases. Errors in the °C), after which the supernatant, containing xylanase inhibitors, was
extinction at 280 nm due to modifications in the amino acid structure isolated. Xylanase inhibition sensitivity was then measured by
were negligible. incubating an appropriate xylanase dilution (measurements in linear
Biochemical Characterization of Xylanases. Specific Activities area of enzyme concentration vs extinction) (500 μL; 100 mM sodium
toward Different Substrates. The specific activities of the wild-type acetate buffer pH 5.5) for 30 min at 30 °C with the Crousti wheat
and mutant xylanases were determined toward different substrates in flour extract (500 μL). Subsequently, the xylanase activity on
order to assess the relative preference for these substrates. All enzyme Xylazyme AX was measured as described above against a control
activities were measured at dough pH, which is 5.5,32 in a 100 mM made from wheat flour extract. Inhibition sensitivity was expressed as a
sodium acetate buffer containing 0.50 mg/mL bovine serum albumin reduction (%) of xylanase activity.
(BSA). The preincubation and incubation temperature were 30 °C in Bread Making Trials. Dough pieces and bread loaves were
all assays, i.e., the dough fermentation temperature. produced in triplicate at 10 g scale, according to the straight-dough
Specific xylanase activities against WU-AX and WE-AX were method.33 Flour (10.0 g, 14% moisture base), 0.53 g of fresh yeast, 1.5
estimated using the polymeric chromophoric substrates Xylazyme g of sodium chloride, 6.0 g of sucrose, deionized water (4.86 mL for
AX and Azo-wheat AX, respectively, as described by Cuyvers et al.23 Crousti and 4.20 mL for Claire), and 1.0 mL of enzyme solution
The bases for these measurements were changes in solubility in water (dialyzed overnight against 100 mM sodium acetate buffer pH 5.5
or in a water−ethanol mixture, respectively, after hydrolysis of the (volume ratio 1:160 enzyme solution:buffer)) were mixed in a 10 g pin
chromophoric substrate by the xylanase. The color intensity of the mixer (National Manufacturing, Lincoln, NE, USA) during 4.0 and 2.5
soluble fragments was then proportional to the xylanase activity. For min for Crousti and Claire flour, respectively. The dough baking
the determination of the specific activity on Xylazyme AX, an absorption and mixing times were based on Farinograph (Brabender,
appropriately diluted enzyme solution (1000 μL) (measurements in Duisburg, Germany) and Mixograph (National Manufacturing,
the linear area of enzyme concentration vs extinction) was equilibrated Lincoln, NE, USA) analyses, respectively.34,35 The xylanase activities
for 10 min. Then a Xylazyme AX tablet was added. After 60 min of of the dialyzed enzyme solutions were measured against Xylazyme AX
incubation, the reaction was terminated by addition of 10.0 mL of (as described above). For XBS and its mutant, dosages between 100
1.0% (w/v) tris(hydroxymethyl)aminomethane, vigorous vortex and 700 Xyl-U/kg of flour were used, while for XPH and its mutant,
stirring, and immediate filtration through a MN 615 filter dosages between 5 and 35 Xyl-U/kg of flour were used. For control
(Macherey-Nagel, Düren, Germany). The extinction of the filtrate dough and bread, the enzyme solution was replaced by 1.0 mL of
was determined at 590 nm (E590) [Ultraspec III UV/vis spectropho- sodium acetate buffer (100 mM, pH 5.5). The dough was fermented
tometer (Pharmacia Biotech, Uppsala, Sweden)] against a control, for 90 min in a fermentation cabinet (National Manufacturing) at 30
prepared by incubating the enzyme solution without substrate. One °C and 90% relative humidity with intermediate punching at 52 and 77
Xylazyme unit (Xyl-U) corresponds to the enzyme concentration min. After final punching at 90 min, the dough pieces were molded
required to obtain an E590 of 1.0, under the conditions of the assay. and proofed for 36 min (30 °C and 90% relative humidity). Finally,
The specific activity on Azo-wheat AX was determined by first baking was performed in a rotary oven (National Manufacturing) for
preincubating an appropriately diluted enzyme solution and the liquid 13 min at 232 °C and the bread was immediately weighed after baking.
substrate separately for 10 min. After addition of 500 μL of the Loaf volume was measured 120 min after baking by rapeseed
substrate to 500 μL of the enzyme solution, incubation was extended displacement.36 Dough (immediately after mixing and proofing) and
for 60 min, after which the reaction was terminated by addition of 2.5 bread samples were frozen in liquid nitrogen. After lyophilization, they
mL of ice cold ethanol and vigorous vortex stirring. The samples were were ground with a laboratory mill (model A10, IKA-Werke GmbH
kept on ice for 10 min. After centrifugation for 10 min at 4000g and 4 and Co. KG, Staufen, Germany) and sieved (⌀ = 250 μm) before
°C in a Sigma 6-16K centrifuge (Sigma Zentrifugen, Osterode am further analysis.
Harz, Germany), the E590 of the supernatant was measured against a Preparation of Aqueous Extracts from Flour, Dough, and
control, prepared by incubating the enzyme solution without substrate. Bread Samples. Aqueous extracts of flour, (fermented) dough, and
One Azo-wheat unit (Azo-U) was defined as the enzyme bread samples were prepared by suspending the samples (1.0 g) in
concentration needed to increase E590 by 1.0 under the conditions 10.0 mL of potassium chloride/hydrogen chloride (20 mM) buffer at
of the assay. The substrate selectivity factor (SSF), defined as the pH 3.0. Under these conditions, acid hydrolysis did not occur and
relative preference of xylanases toward WU-AX or WE-AX, was enzymatic breakdown of AX was minimized (results not shown). The
calculated as the ratio of the specific activity toward Xylazyme AX to suspensions were shaken for 30 min at 7 °C (Laboshake, VWR
that toward Azo-wheat AX. International, Leuven, Belgium), and, after centrifugation (10 min,
Specific xylanase activities against X6 and reaction products of this 4000g, 7 °C), the supernatants were analyzed immediately or frozen
hydrolysis were determined by high-performance anion-exchange until further analysis.
chromatography followed by pulsed amperometric detection. Sample AX Levels and Compositions. The carbohydrate contents and
preparation and quantification of the formed hydrolysis products were compositions obtained after hydrolysis of flour or aqueous extracts of
done as described by Cuyvers et al.23 One enzyme unit on X6 (X6-U) flour, dough, or bread samples, prepared as described in the previous
was then defined as the concentration of enzyme needed for the section, were determined by gas chromatography. Hydrolysis of flour
formation of 1.0 μM xylotriose. (10.0−15.0 mg) was performed in 2.0 M trifluoroacetic acid (TFA)
Temperature and pH Dependency. Temperature dependency of (5.0 mL), while aqueous extracts (2.5 mL) were hydrolyzed in 4.0 M
xylanase activity was determined by measuring the activity on TFA (2.5 mL), both for 60 min at 110 °C. Reduction and acetylation
Xylazyme AX, under the conditions described above, with incubation were performed with sodium borohydride and acetic anhydride,
temperatures ranging from 10 to 80 °C with intervals of 10 °C. To respectively, according the procedure of Englyst and Cummings.37 To
determine pH dependency of xylanase activity, enzyme dilutions were measure reducing end xylose and arabinose contents, reduction was
made in potassium chloride/hydrogen chloride (20 mM, pH 2.0), performed preceding the hydrolysis.38 A combination of reduction and

C DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

Table 1. Biochemical Characteristics of Wild-Type and Mutant Bacillus subtilis Xylanases (XBS WT and XBS 3A) and
Pseudoalteromonas haloplanktis Xylanases (XPH WT and XPH 2A)a
XBS WT XBS 3A XPH WT XPH 2A
GenBank accession no. P18429 AJ427921.1
modified amino acids G56A-T183A-W185A W249A-Y315A
GH 11 11 8 8
MM (kDa) 20.4 20.2 46.1 45.9
act. on xylohexaose (%)b 100 ± 5 a 96 ± 2 a 100 ± 3 A 115 ± 4 B
act. on Xylazyme AX (%)b 100 ± 2 a 43 ± 1 b 100 ± 6 A 155 ± 4 B
act. on Azo-wheat AX (%)b 100 ± 3 a 34 ± 1 b 100 ± 9 A 128 ± 5 B
SSF 8.3 ± 0.5 a 10.5 ± 0.4 b 10.1 ± 1.1 A 12.2 ± 0.3 B
Topt (°C) 50 a 50 a 30 A 30 A
pHopt 6−7 a 7 a 9 A 9 A
reduction of act. by inhibitors (%) 88 ± 2 a 90 ± 3 a 0±1 A 1±1 A
a
Values with the same small or capital letter for XBS and XPH, respectively, are not significantly different from each other (α = 0.05). bExpressed
relative to the activity of the wild-type enzyme (100%). The activities on xylohexaose (X6), Xylazyme AX, and Azo-wheat AX were for the wild-type
XBS 1 X6-U = 0.17 nM, 1 Xyl-U = 1.03 nM, and 1 Azo-U = 8.58 nM, respectively. For the wild-type XPH, 1 X6-U = 4.73 nM, 1 Xyl-U = 2.71 nM,
and 1 Azo-U = 27.28 nM.

Figure 2. Specific loaf volumes of the breads in which XBS (A, B) and XPH (C, D) wild types (XBS WT and XPH WT) and mutants (XBS 3A and
XPH 2A) were incorporated as a function of enzyme dosage. The specific loaf volumes are expressed relative to a control bread without xylanase
(fixed at 100%). Enzyme dosages are expressed in Xylazyme units per kg flour. Two types of wheat flour were used: Crousti (A, C) and Claire (B,
D). All values are averages of triplicate experiments, and error bars show the standard deviations. Values with the same small letter at different
enzyme dosages of one xylanase are not significantly different from each other (α = 0.05). Wild-type xylanases which significantly differ from the
mutant xylanase at the same dosage are indicated with an asterisk.

acetylation, without prior hydrolysis, was performed to determine free bread extracts was 0.88 times the sum of the xylose and arabinose
xylose and arabinose. The formed alditol acetates (1.0 μL) were contents. The total arabinose content was corrected for free arabinose
separated on a Supelco SP-2380 polar column (30 m × 0.32 mm inner as well as arabinose originating from arabinogalactan-peptides
diameter; 0.2 μm film thickness) (Supelco, Bellefonte, PA, USA) in an (AGP).39 The total xylose content was corrected for the presence of
Agilent chromatograph (Agilent 6890 series, Wilmington, DE, USA)
equipped with autosampler, splitter injection port (split ratio 1:20), free xylose. All contents were expressed on a dm base. The average
and flame ionization detector. The carrier gas was helium. Separation degree of polymerization (avDP) of the soluble AX was calculated as
was at 225 °C while injection and detection were at 270 °C. The total the sum of the xylose and arabinose content divided by the reducing
AX and WE-AX content of flour and the AX content in dough and end xylose content. The arabinose content was corrected for free

D DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

Figure 3. Levels of solubilized WU-AX in dough and bread samples supplemented with xylanases of Bacillus subtilis [XBS wild type and XBS mutant
(XBS 3A)] (300 Xylazyme units/kg flour) (A, B) and Pseudoalteromonas haloplanktis [XPH wild type and XPH mutant (XPH 2A)] (25 Xylazyme
units/kg flour) (C, D) after mixing, fermentation, and baking. Two types of flour were used: Crousti (A, C) and Claire (B, D) flour. No xylanase was
incorporated in control dough or bread. According to a Tukey test (α < 0.05), values with the same small letter at different bread making stages of
control bread or with the same xylanase are not significantly different. Values that are not significantly different for control bread or different
xylanases at one particular baking stage are indicated with the same capital letter.

arabinose and arabinose present in AGP, while the xylose and reducing an ideal substrate to measure differences in inherent activity.23
xylose content was corrected for free xylose. For XBS, mutations in the SBS had no effect on the activity on
Viscosity. The viscosity of extracts of flour, dough, and bread
X6. In contrast, the activity on X6 increased by 15% by mutating
samples (500 μL), was determined with a Brookfield DV II+
viscometer (Brookfield Engineering Laboratories Inc., Stoughton, the SBS of XPH WT (Table 1). The hydrolysis pattern of X6
MA, USA) at 30 °C with a CP40 cone and a constant shear rate of 60 was identical for all mutant xylanases and their wild-type
s−1. The specific viscosity was defined as the relative viscosity, i.e., the counterparts (results not shown).
ratio of the viscosity of the extract and that of a potassium chloride/ On both Xylazyme AX and Azo-wheat AX, significant
hydrogen chloride (25 mM; pH 3.0) buffer, minus one.3 differences in activity were revealed after mutating the SBS.
Statistical Analysis. Specific activities toward Xylazyme AX and XBS 3A showed only 34% of the activity of XBS WT for Azo-
Azo-wheat AX were determined in 5-fold, while all other analyses were
done in triplicate. All data were analyzed using statistical software JMP wheat AX, while for Xylazyme AX, XBS 3A activity was 43% of
Pro 11 (SAS Institute, Cary, NC, USA) to verify whether mean values that of the wild-type enzyme. In contrast to XBS, the activities
were significantly different at a difference level (α) of 0.05 using the on both chromophoric substrates increased for XPH 2A
two-way ANOVA procedure. compared to its wild type. Especially the activity on Xylazyme


AX (155%) increased, while the activity increase on Azo-wheat
RESULTS AX (128%) was less pronounced. Since the activity toward
Biochemical Characterization of Xylanases. Biochem- water-unextractable Xylazyme AX was higher for both mutant
ical properties of XBS and XPH enzymes modified in their SBS xylanases than toward water-extractable Azo-wheat AX, the SSF
were compared to those of their corresponding wild-type increased by 26% and 21% for XBS and XPH, respectively
enzymes to investigate whether modifying the SBS of a xylanase (Table 1).
influences the SSF without affecting other biochemical Temperature and pH Dependency of Xylanase Activity.
properties. The wild-type and modified XBS showed maximal xylanase
Specific Activity toward Different Substrates. Since X6 is activity at 50 °C, while the optimal temperature was 30 °C for
too small to reach the active site and SBS simultaneously, it is the psychrophilic wild-type XPH and its mutant (Table 1).
E DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

Figure 4. Degree of polymerization (avDP) of the soluble AX fraction in dough and bread samples enriched with xylanases of Bacillus subtilis [XBS
wild type and XBS mutant (3A)] (300 Xylazyme units/kg flour) (A, B) and Pseudoalteromonas haloplanktis [XPH wild type and XPH mutant (2A)]
(25 Xylazyme units/kg flour) (C, D) after mixing, fermentation, and baking. Two types of flour were used: Crousti (A, C) and Claire (B, D) flour.
To the control dough or bread, no xylanase was added. According to a Tukey test (α < 0.05), values with the same small letter at different bread
making stages of control bread or with the same xylanase are not significantly different. Values that are not significantly different for control bread or
different xylanases at one particular baking stage are indicated with the same capital letter.

Maximal activity was at pH 6.0 to 7.0 for XBS and at pH 9.0 3A was significantly lower than with XBS WT. In bread making
for XPH, for both wild-type enzymes as well as their mutant with Claire flour, the maximal increase in specific loaf volume
counterparts (Table 1). was reached at a lower dosage for XBS 3A (300 Xyl-U/kg flour)
Inhibition Sensitivity. The addition of a wheat flour extract compared to XBS WT (500 Xyl-U/kg flour). Moreover, the
reduced the xylanase activity of the XBS wild type and mutant volume increase at optimal dosages was higher when XBS 3A
88% and 90% respectively, compared to the activity measure- was used. For Crousti flour, no differences in specific loaf
ment in the absence of wheat flour extract. Neither XPH wild volume were observed when using the wild-type or mutant
type nor mutant was affected by inhibitors. XPH, regardless of the dosage used (Figure 2C). Maximal
Functionality of XBS and XPH in Bread Making. volume increases of 20.3% and 19.1% were obtained with XPH
Changes in Dough and Bread Properties. The addition of WT and XPH 2A, respectively, at an enzyme dosage of 25 Xyl-
wild-type and mutant xylanases to the bread making process
U/kg flour. Specific volumes of the Claire loaves supplemented
influenced the manageability of the dough. After mixing, dough
with XPH 2A were higher than those supplemented with XPH
supplemented with XPH enzymes felt drier compared to dough
supplemented with XBS enzymes. After fermentation, dough WT, for all dosages tested (Figure 2D). Maximal volume
supplemented with XPH enzymes became more sticky, while increases of 23.3% and 31.9% were obtained with XPH WT and
the dough manageability of dough supplemented with XBS XPH 2A, respectively.
enzymes improved. Dough manageability after 90 min AX Properties during Bread Making. To evaluate changes in
fermentation limited the enzyme dosages that can be used to solubilized WU-AX and avDP, the AX population was
700 Xyl-U/kg flour for XBS enzymes and 35 Xyl-U/kg flour for monitored during different steps of the bread making process
XPH enzymes, but no differences were observed between the (Figures 3 and 4). For this, enzyme dosages of 300 Xyl-U/kg
wild-type and mutant xylanases. flour for XBS wild type and mutant and 25 Xyl-U/kg flour for
Addition of xylanases significantly increased specific loaf XPH wild type and mutant were used since these dosages
volumes, for both flour types used (Figure 2). Only at 700 Xyl- resulted in significant differences in volume of Claire bread
U/kg Crousti flour, the specific loaf volume obtained with XBS between wild-type and mutant xylanases (cf. previous section).
F DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

Table 2. Solubilization of Water-Unextractable Arabinoxylan (WU-AX) and Decrease of the Average Degree of Polymerization
(avDP) during the Fermentation Stage in Crousti and Claire Bread Making Processesa
solubilization of WU-AX during fermentation (% of total WU-AX) decrease in avDP during fermentation
Crousti flour Claire flour Crousti flour Claire flour
control 14 ± 1 c 3±0 c 6±0 b 6±1 c
XBS WT 19 ± 3 b 7±4 b 4±5 b 12 ± 2 b
XBS 3A 29 ± 3 a 17 ± 4 a 11 ± 2 a 24 ± 2 a
control 14 ± 1 c 3±0 c 6±0 b 6±1 c
XPH WT 16 ± 3 b 12 ± 3 b 15 ± 4 a 31 ± 1 b
XPH 2A 26 ± 7 a 29 ± 3 a 20 ± 8 a 46 ± 1 a
a
Wild types and mutants of the xylanases of Bacillus subtilis (XBS) and Pseudoalteromonas haloplanktis (XPH) were added to both flour types. A
control dough without xylanase addition was also investigated. The results of the statistical analysis are shown per enzyme. Values with the same
small letter are not significantly different.

Figure 5. Specific viscosity of aqueous extracts of dough and bread samples relative to a potassium chloride/hydrogen chloride buffer (20 mM; pH
3.0). A wild-type and mutant xylanase of Bacillus subtilis [(XBS wild type and XBS mutant (3A)] (300 Xylazyme units/kg flour) (A, B) and
Pseudoalteromonas haloplanktis [XPH wild type and XPH mutant (2A)] (25 Xylazyme units/kg flour) (C, D) were incorporated in dough and bread
samples prepared with Crousti (A, C) and Claire (B, D) flour. To the control samples, no xylanase was added. According to a Tukey test (α < 0.05),
values with the same small letter at different bread making stages of control bread or with the same xylanase are not significantly different. Values that
are not significantly different for control bread or different xylanases at one particular baking stage are indicated with the same capital letter.

Solubilization of WU-AX. The level of WU-AX solubilization the wild-type counterpart. Again, the mutant XPH was able to
in dough after mixing and fermentation was significantly higher solubilize more WU-AX during fermentation compared to the
when xylanases were used (Figure 3). For Crousti as well as wild type. After baking, the amount of solubilized WU-AX was
Claire bread making, no differences in solubilized WU-AX decreased for all samples tested.
content after mixing were observed between incorporation of avDP of the Soluble AX Fraction. During mixing and
the wild-type and mutant XBS. During the fermentation stage, fermentation, the avDP of the soluble AX fraction of xylanase
XBS 3A solubilized more WU-AX than its wild-type counter- supplemented doughs was significantly lower compared to
part (Table 2). Immediately after mixing, solubilization of WU- control dough samples (Figure 4). When XBS enzymes were
AX in Claire flour had progressed less for XPH 2A compared to incorporated in Crousti bread, no differences between the wild-
G DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

type and mutant xylanase were observed after mixing. During after addition of XBS and XPH. Dough supplemented with
fermentation, XBS WT was not able to further lower the avDP XPH was dry after mixing but became more sticky during
of the soluble AX fraction and this in contrast to XBS 3A fermentation, while the inverse was observed for XBS. This can
(Table 2). When XBS was added to bread making with Claire be explained by the inhibition sensitivity of the xylanases: XPH
flour, the avDP of the soluble AX fraction was already lower is not sensitive to inhibitors and keeps working during the
after the mixing stage when XBS WT was compared to XBS 3A. entire process, while the activity of XBS, which is inhibited by
During fermentation, the avDP of the soluble AX fraction TAXI,40 most probably decreases rapidly during bread making.
further decreased. Since this decrease was more outspoken for Differences in inhibition sensitivity also explain why XPH was
XBS 3A, there was no significant difference anymore between added in lower dosages than XBS. Modifying the SBS had no
the wild-type and mutant XBS after fermentation. Furthermore, impact on inhibition sensitivity.
the avDP of the soluble AX population increased after baking, While little if any significant difference in specific loaf volume
except when XBS WT was added to Crousti bread. For the was observed between wild-type and mutant xylanases in bread
control breads, in contrast, avDP decreased after baking. making with Crousti (Figure 2), an additional volume increase
The results obtained with XPH were similar to those of XBS of 7.1% and 8.6% was obtained after supplementation of the
(Figure 4). After mixing and fermentation in Crousti bread mutant XBS and mutant XPH, respectively, at their optimal
making, no differences were observed between the wild-type dosages in Claire bread making. The stronger gluten network of
and mutant XPH. However, the avDP of the soluble AX dough made with Crousti flour probably masked possible
decreased more pronouncedly during fermentation when using additional improvements of the mutant xylanases. In contrast,
the mutant XPH (Table 2). After baking, the avDP of the when using the weak Claire flour, the effects of incorporation of
soluble AX fraction was significantly lower with XPH 2A than xylanases with different SSF were more pronounced.
with XPH WT. For Claire bread making, the avDP of the Since equivalent units (Xyl-U) of wild-type and mutant
soluble AX fraction after mixing was lower for the wild-type xylanases were used in bread making, it was expected that also
XPH compared to the mutant. During fermentation, the equivalent amounts of WU-AX were solubilized during the
decrease in avDP was more outspoken for XPH 2A. The avDP different bread making stages. The amount of solubilized WU-
of the soluble AX population increased again after baking. AX after mixing was indeed similar for wild-type and mutant
Extract Viscosity at Different Phases of Bread Making. xylanases (Figure 3). The increase in solubilized WU-AX was
While XBS enzymes had no impact on extract viscosity after accompanied by a decrease in the avDP of the soluble AX
mixing, Crousti dough supplemented with XBS enzymes population (Figure 4). It is possible that the fragments derived
showed a significantly lower extract viscosity after fermentation from solubilizing WU-AX had a lower avDP than the native
than control dough (Figure 5). No differences were observed WE-AX present in flour. Additionally, native WE-AX and S-AX
between XBS WT and XBS 3A. When XBS enzymes were used might have been hydrolyzed into smaller fragments, which
in dough and bread samples made with Claire flour, no would also result in a lower avDP. These fragments would have
differences were observed compared to the control. a minimal chain length of five xylose molecules since XPH and
Supplementation of XPH had no impact on extract viscosity XBS are not able to hydrolyze substrates with a degree of
after mixing. Dough and bread supplemented with XPH polymerization lower than six.41 However, since XBS 3A and
enzymes showed a significantly lower extract viscosity after XPH 2A had a lower activity toward WE-AX and S-AX than the
fermentation and baking than control dough and bread. No wild-type enzymes (Table 1), we had expected that
differences in specific viscosity were noticed between addition incorporation of the mutant xylanases in bread making would
of the wild-type and mutant XPH. result in differences in avDP of the soluble AX fraction. This

■ DISCUSSION
Modifying the SBS of XBS and XPH by replacing aromatic
was not the case, and, based on this, it can be concluded that
the lower avDP of solubilized WU-AX was probably responsible
for the decrease in overall avDP.
amino acids responsible for binding of the substrate by an inert During fermentation, an additional amount of WU-AX was
alanine residue resulted in an increased SSF and, hence, an solubilized (Figure 3 and Table 2). Especially for Crousti bread
increased preference for WU-AX (Table 1). Other biochemical making, this was not least caused by the xylanase activity of the
properties were not influenced by these mutations. As shown flour itself, as shown for the control dough. For xylanase
for XPH 2A, modifying of the SBS did not automatically supplemented dough samples, the amount of additionally
decrease the activity on polymeric substrates as the activity of solubilized WU-AX was lower compared to what was
the mutant XPH on Xylazyme AX and Azo-wheat AX was solubilized earlier during the mixing stage. Both mutant
increased by 55% and 28%, respectively, compared to XPH xylanases solubilized WU-AX to a larger extent and decreased
WT. With the wild-type XPH, the substrate was probably the avDP of soluble AX more strongly compared to their
bound too tightly to the SBS for optimal activity. Modifying respective wild types. This is remarkable since the same activity
this position could therefore allow a better positioning or faster on water-unextractable substrate of wild-type and mutant
throughput of the substrate with respect to the active site. As xylanases was added. In the past, several functions have already
mentioned by Cuyvers et al.,23 the increased activity of XPH 2A been described for SBSs.20 Possibly, a number of these
toward X6 was probably the result of subtle positional change of functions were slightly enhanced after modification and ensured
residues located in the active site after modification of the SBS. the xylanase to become more active. This resulted in a more
Since modifying the SBS provided xylanases that only differed convenient hydrolysis for the mutant xylanases compared to the
in SSF, they were ideal tools to investigate the specific influence wild type.
of substrate selectivity in bread making. XPH degraded S-AX more severely than XBS as deduced
Equivalent units (Xyl-U) of wild-type and mutant xylanases from the avDP of soluble AX after fermentation (Figure 4). A
were added to bread making processes based on Crousti and reason for the lower hydrolyzing capacity of XBS is probably its
Claire flour. Differences in dough manageability were observed inhibition sensitivity toward TAXI, as this inhibitor was present
H DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

in excess in wheat flour. It has already been shown that sels, Belgium) for a position as postdoctoral researcher. This
inhibition-insensitive xylanases show a stronger degradation of research is also part of the Methusalem program “Food for the
S-AX and WE-AX than inhibition-sensitive ones.11,14 future” (2007−2014) at the KU Leuven.
WE-AX with a high molecular weight have a high viscosity Notes
forming capacity and are able to stabilize the liquid films The authors declare no competing financial interest.


surrounding the gas cells in bread dough.42 Despite differences
in solubilization degree between wild-type and mutant ACKNOWLEDGMENTS
xylanases, no differences in specific viscosity were observed Dr. Emmie Dornez (Mauri Research, The Netherlands) is
(Figure 5). Liquid films in bread dough are indeed more gratefully thanked for fruitful discussions.


concentrated than the extracts used for measuring specific
viscosity. Alternatively, it is possible that differences in bread ABBREVIATIONS USED
loaf volume are only determined by WU-AX solubilization
degree and accompanying water release and not by the bulk α, difference level; AGP, arabinogalactan-peptides; avDP,
viscosity of aqueous phase. Roels and co-workers43 showed that average degree of polymerization; AX, arabinoxylan; Azo-U,
increasing the baking absorption up to 10% above the Azo-wheat unit; BSA, bovine serum albumin; CBMs,
Farinograph baking absorption results in higher loaf volumes. carbohydrate-binding modules; dm, dry matter; E590, extinction
This could also explain the increased specific loaf volume when at 590 nm; GH, glycoside hydrolase family; S-AX, solubilized
mutant xylanases were added to bread making. Since Claire AX; SBS, secondary binding site; SSF, substrate selectivity
flour is a weak flour type, the impact of additional water release factor; TAXI, Triticum aestivum xylanase inhibitor; TFA,
due to degradation of WU-AX might be more pronounced. trifluoroacetic acid; TL-XI, thaumatin-like xylanase inhibitor;
During the early baking phase, it was expected that XPH WE-AX, water-extractable AX; WT, wild type; WU-AX, water-
would still hydrolyze AX, which would then result in an unextractable AX; X6, xylohexaose; X6-U, xylohexaose unit;
additional decrease of the avDP of the soluble AX fraction. A XBS, xylanase XynA of Bacillus subtilis; XBS 3A, G56A-T183A-
decrease in solubilized WU-AX level was, however, observed W185A mutant of xylanase XynA of Bacillus subtilis; XIP,
(Figure 3). This indicates that part of the previously solubilized xylanase inhibiting protein; XPH, xylanase of Pseudoalteromonas
AX turned unextractable again, probably due to cross-linking of haloplanktis; XPH 2A, W249A-Y315A mutant of xylanase of
Pseudoalteromonas haloplanktis; Xyl-U, Xylazyme unit


AX molecules with other AX molecules or flour components
and/or due to physical inclusion in the gelatinized starch matrix REFERENCES
since no amylase was added during the extraction.3,44,45 For
Claire bread making, it appears that AX molecules with a low (1) Rouau, X.; El-Hayek, M. L.; Moreau, D. Effect of an enzyme
preparation containing pentosanases on the bread-making quality of
molecular weight preferentially turned unextractable again since flours in relation to changes in pentosan properties. J. Cereal Sci. 1994,
the avDP was increased after baking (Figure 4). On the one 19, 259−272.
hand, smaller AX molecules are more mobile and less sensitive (2) Courtin, C. M.; Delcour, J. A. Arabinoxylans and endoxylanases
to steric hindrance, which makes them able to cross-link more in wheat flour bread-making. J. Cereal Sci. 2002, 35, 225−243.
easily with other constituents. On the other hand, the fact that (3) Courtin, C. M.; Gelders, G. G.; Delcour, J. A. Use of two
small molecules are more mobile renders them also easier to endoxylanases with different substrate selectivity for understanding
extract. arabinoxylan functionality in wheat flour breadmaking. Cereal Chem.
In conclusion, this study shows that increasing the preference 2001, 78, 564−571.
of a xylanase for water-unextractable substrates compared to (4) Butt, M. S.; Tahir-Nadeem, M.; Ahmad, Z.; Sultan, M. T.
water-extractable ones, i.e., increasing its substrate selectivity, Xylanases and their applications in baking industry. Food Technol.
Biotechnol. 2008, 46, 22−31.
can enhance its functionality in bread making. Due to increased (5) Jaekel, L. Z.; da Silva, C. B.; Steel, C. J.; Chang, Y. K. Influence of
WU-AX solubilization during fermentation, these xylanases xylanase addition on the characteristics of loaf bread prepared with
ensure that more of the WU-AX that is detrimental in bread white flour or whole grain wheat flour. Ciênc. Technol. Aliment. 2012,
making is converted into S-AX. This possibly allows for 32, 844−849.
additional water release, responsible for an increase in specific (6) Izydorczyk, M. S.; Biliaderis, C. G. Cereal arabinoxylans: advances
loaf volume and/or a lower optimal dosage when weak flour in structure and physicochemical properties. Carbohydr. Polym. 1995,
was used. With the experimental approach followed here, 28, 33−48.
viscosity differences could not be pinpointed as driving (7) Meuser, F.; Suckow, P. Non-starch polysaccharides. In Chemistry
mechanism for differences between wild-type and mutant and Physics of Baking; Blanchard, J. M. V., Frazier, P. J., Galliard, T.,
enzymes. These conclusions, of course, have to be seen in the Eds.; The Royal Society of Chemistry: London, U.K., 1986; pp 42−61.
(8) Courtin, C. M.; Roelants, A.; Delcour, J. A. Fractionation−
right perspective. The functionality of xylanases in bread
reconstitution experiments provide insight into the role of
making is determined by more biochemical properties than endoxylanases in bread-making. J. Agric. Food Chem. 1999, 47,
only their substrate selectivity.


1870−1877.
(9) Moers, K.; Celus, I.; Brijs, K.; Courtin, C. M.; Delcour, J. A.
AUTHOR INFORMATION Endoxylanase substrate selectivity determines degradation of wheat
Corresponding Author water-extractable and water-unextractable arabinoxylan. Carbohydr.
*Phone: +32 (0) 16 32 19 17. Fax: +32 (0) 16 32 19 97. E- Res. 2005, 340, 1319−1327.
mail: christophe.courtin@biw.kuleuven.be. (10) Moers, K.; Courtin, C. M.; Brijs, K.; Delcour, J. A. A screening
method for endo-β-1,4-xylanase substrate selectivity. Anal. Biochem.
Funding 2003, 319, 73−77.
S.L. acknowledges the Agency for Innovation through Science (11) Dornez, E.; Verjans, P.; Arnaut, F.; Delcour, J. A.; Courtin, C.
and Technology in Flanders (IWT-Vlaanderen, Brussels, M. Use of psychrophilic xylanases provides insight into the xylanase
Belgium) for financial support. A.P. wishes to acknowledge functionality in bread making. J. Agric. Food Chem. 2011, 59, 9553−
the Research FoundationFlanders (FWO-Vlaanderen, Brus- 9562.

I DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry Article

(12) Verjans, P.; Dornez, E.; Delcour, J. A.; Courtin, C. M. Selectivity (30) Van Craeyveld, V.; Dornez, E.; Holopainen, U.; Selinheimo, E.;
for water-unextractable arabinoxylan and inhibition sensitivity govern Poutanen, K.; Delcour, J. A.; Courtin, C. M. Wheat bran AX properties
the strong bread improving potential of an acidophilic GH11 and choice of xylanase affect enzymic production of wheat bran-
Aureobasidium pullulans xylanase. Food Chem. 2010, 123, 331−337. derived arabinoxylan-oligosaccharides. Cereal Chem. 2010, 87, 283−
(13) Fierens, E.; Gebruers, K.; Courtin, C. M.; Delcour, J. A. 291.
Xylanase inhibitors bind to nonstarch polysaccharides. J. Agric. Food (31) Laemmli, U. K. Cleavage of Structural Proteins during the
Chem. 2008, 56, 564−570. Assembly of the Head of Bacteriophage T4. Nature 1970, 227, 680−
(14) Trogh, I.; Sørensen, J. F.; Courtin, C. M.; Delcour, J. A. Impact 685.
of inhibition sensitivity on endoxylanase functionality in wheat flour (32) Jayaram, V. B.; Cuyvers, S.; Lagrain, B.; Verstrepen, K. J.;
breadmaking. J. Agric. Food Chem. 2004, 52, 4296−4302. Delcour, J. A.; Courtin, C. M. Mapping of Saccharomyces cerevisiae
(15) Verjans, P. Functionality of extremophilic xylanases in bread metabolites in fermenting wheat straight-dough reveals succinic acid as
making. Ph.D. dissertation; K.U. Leuven: Leuven, Belgium, 2010; 185 pH-determining factor. Food Chem. 2013, 136, 301−308.
pp. (33) Shogren, M. D.; Finney, K. F. Bread-making test for 10 grams of
(16) De Vos, D.; Collins, T.; Nerinckx, W.; Savvides, S. N.; flour. Cereal Chem. 1984, 61, 418−423.
Claeyssens, M.; Gerday, C.; Feller, G.; Van Beeumen, J. (34) AACC International. Approved Methods of Analysis, 11th ed.;
Oligosaccharide binding in family 8 glycosidases: Crystal structures Method 54-21.02. Rheological Behaviour of Flour by Farinograph:
of active-site mutants of the β-1,4-xylanase pXyl from Pseudoaltermonas Constant Flour Weight Procedure. Approved Nov 3, 1999; AACC
haloplanktis TAH3a in complex with substrate and product. International: St. Paul, MN, 2000.
Biochemistry 2006, 45, 4797−4807. (35) AACC International. Approved Methods of Analysis, 11th ed.;
(17) Vandermarliere, E.; Bourgois, T. M.; Rombouts, S.; Van Method 54-40.02. Mixograph Method. Approved Nov 3, 1999; AACC
campenhout, S.; Volckaert, G.; Strelkov, S. V.; Delcour, J. A.; Rabijns, International: St. Paul, MN, 2000.
A.; Courtin, C. M. Crystallographic analysis shows substrate binding at (36) Vanhamel, S.; Vandenende, L.; Darius, P. L.; Delcour, J. A. A
the − 3 to + 1 active-site subsites and at the surface of glycoside volumeter for breads prepared from 10-grams of flour. Cereal Chem.
hydrolase family 11 endo-1,4-β-xylanases. Biochem. J. 2008, 410, 71− 1991, 68, 170−172.
79. (37) Englyst, H. N.; Cummings, J. H. Simplified method for the
(18) Ludwiczek, M. L.; Heller, M.; Kantner, T.; McIntosh, L. P. A measurement of total non-starch polysaccharides by gas-liquid
secondary xylan-binding site enhances the catalytic activity of a single- chromatography of constituent sugars as alditol acetates. Analyst
domain family 11 glycoside hydrolase. J. Mol. Biol. 2007, 373, 337− 1984, 109, 937−942.
354. (38) Courtin, C. M.; Van den Broeck, H.; Delcour, J. A.
(19) Guillén, D.; Sánchez, S.; Rodríguez-Sanoja, R. Carbohydrate- Determination of reducing end sugar residues in oligo- and
binding domains: multiplicity of biological roles. Appl. Microbiol. polysaccharides by gas−liquid chromatography. J. Chromatogr. A
Biotechnol. 2010, 85, 1241−1249. 2000, 866, 97−104.
(20) Cuyvers, S.; Dornez, E.; Delcour, J. A.; Courtin, C. M. (39) Loosveld, A.-M. A.; Grobet, P. J.; Delcour, J. A. Contents and
Occurrence and functional significance of secondary carbohydrate Structural Features of Water-Extractable Arabinogalactan in Wheat
binding sites in glycoside hydrolases. Crit. Rev. Biotechnol. 2012, 32, Flour Fractions. J. Agric. Food Chem. 1997, 45, 1998−2002.
93−107. (40) Gebruers, K.; Brijs, K.; Courtin, C. M.; Fierens, K.; Goesaert,
(21) Moers, K.; Bourgois, T.; Rombouts, S.; Beliën, T.; Van H.; Rabijns, A.; Raedschelders, G.; Robben, J.; Sansen, S.; Sørensen, J.
Campenhout, S.; Volckaert, G.; Robben, J.; Brijs, K.; Delcour, J. A.; F.; Van Campenhout, S.; Delcour, J. A. Properties of TAXI-type
Courtin, C. M. Alteration of Bacillus subtilis XynA endoxylanase endoxylanase inhibitors. Biochim. Biophys. Acta, Proteins Proteomics
substrate selectivity by site-directed mutagenesis. Enzyme Microb. 2004, 1696, 213−221.
Technol. 2007, 41, 85−91. (41) Pollet, A. Functional and structural analysis of glycoside
(22) Cuyvers, S.; Dornez, E.; Delcour, J. A.; Courtin, C. M. The hydrolase family 8, 10 and 11 xylanases with focus on Bacillus subtilis
secondary substrate binding site of the Pseudoalteromonas haloplanktis xylanase A. Ph.D. thesis; KU Leuven: Belgium, 2010; 207 pp.
GH8 xylanase is relevant for activity on insoluble but not soluble (42) Gan, Z.; Ellis, P. R.; Schofield, J. D. Gas cell stabilisation and gas
substrates. Appl. Microbiol. Biotechnol. 2011, 92, 539−549. retention in wheat bread dough. J. Cereal Sci. 1995, 21, 215−230.
(23) Cuyvers, S.; Dornez, E.; Rezaei, M. N.; Pollet, A.; Delcour, J. A.; (43) Roels, S. P.; Cleemput, G.; Vandewalle, X.; Nys, M.; Delcour, J.
Courtin, C. M. Secondary substrate binding strongly affects activity A. Bread Volume Potential of Variable-Quality Flours with Constant
and binding affinity of Bacillus subtilis and Aspergillus niger GH11 Protein Level As Determined by Factors Governing Mixing Time and
xylanases. FEBS J. 2011, 278, 1098−1111. Baking Absorption Levels. Cereal Chem. 1993, 70, 318−323.
(24) Collins, T.; Gerday, C.; Feller, G. Xylanases, xylanase families (44) Wang, M.; Hamer, R. J.; van Vliet, T.; Oudgenoeg, G.
and extremophilic xylanases. FEMS Microbiol. Rev. 2005, 29, 3−23. Interaction of water extractable pentosans with gluten protein: effect
(25) Van Petegem, F.; Collins, T.; Meuwis, M.; Gerday, C.; Feller, on dough properties and gluten quality. J. Cereal Sci. 2002, 36, 25−37.
(45) Wang, M.; van Vliet, T.; Hamer, R. J. How gluten properties are
G.; Van Beeumen, J. The structure of a cold-adapted family 8 xylanase
affected by pentosans. J. Cereal Sci. 2004, 39, 395−402.
at 1.3 Å resolution: Structural adaptations to cold and investigation of
the active site. J. Biol. Chem. 2003, 278, 7531−7539.
(26) Delcour, J. A.; Vanhamel, S.; De Geest, C. Physico-Chemical
and Functional Properties of Rye Nonstarch Polysaccharides. I.
Colorimetric Analysis of Pentosans and Their Relative Monosacchar-
ide Compositions in Fractionated (Milled) Rye Products. Cereal Chem.
1989, 66, 107−111.
(27) AOAC. Official Methods of Analysis, 16th ed.; Association of
Official Analytical Chemists: Washington, DC, 1995.
(28) AACC International. Approved Methods of Analysis, 11th ed.;
Method 08-01.01. AshBasic Method. Approved Nov 3, 1999;
AACC International: St. Paul, MN, 2000.
(29) Pollet, A.; Vandermarliere, E.; Lammertyn, J.; Strelkov, S. V.;
Delcour, J. A.; Courtin, C. M. Crystallographic and activity-based
evidence for thumb flexibility and its relevance in glycoside hydrolase
family 11 xylanases. Proteins: Struct., Funct., Genet. 2009, 77, 395−403.

J DOI: 10.1021/acs.jafc.6b01473
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Biotechnology 306S (2019) 100013

Contents lists available at ScienceDirect

Journal of Biotechnology
journal homepage: www.elsevier.com/locate/jbiotec

Rhizosecretion of a cisgenic lectin by genetic manipulation of Tepary bean


$
plants (Phaseolus acutifolius)
Dania Martínez-Alarcón a,1, * , Alejandra Mora-Avilés b,1 , Arantxa Espinoza-Núñez b ,
Luz M Serrano Jamaica b , Andrés Cruz-Hernández a , Angelina Rodríguez-Torres a ,
José L Castro-Guillen c, Alejandro Blanco-Labra c , Teresa García-Gasca a, *
a
Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Santiago de Querétaro, 76230, Querétaro, Mexico
b
Departamento de Biotecnología de Plantas, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias (INIFAP) Campo Experimental Bajío, Mexico
c
Departamento de Bioquímica y Biotecnología de Plantas, CINVESTAV Unidad Irapuato, Irapuato 36821, Guanajuato, Mexico

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

Keywords: Tepary bean (Phaseolus acutifolius) lectin fraction (TBLF) has been shown to specifically bind and induce cell death of
Genetic modification different types of cancer cells and also has exhibited an effect on early colon tumorigenesis. However, the
Phaseolus acutifolius development of a pharmaceutical formula is not possible yet because the production process is expensive and slow
Cisgenic lectin
and provides low yields. Therefore, the purpose of the present work was to develop a strategy to produce one
Rhizosecretion
Tepary bean
bioactive lectin by rhizosecretion through root exudates on genetically modified plants. Amplification of Tepary
bean transcripts was performed using degenerate primers, and the products obtained were sequenced. Multiple
alignments of sequences led to elucidating one of the lectins present in TBLF. Its coding sequence was flanked by an
N-terminal secretion signal peptide and a 6xHis-tail. This construction was introduced into P. acutifolius plants using
Agrobacterium tumefaciens to subsequently carry out the in vitro growth of the plants. When roots grew, plants were
transferred to hydroponic conditions and root exudates were analyzed. Results showed the presence of a
glycosylated cisgenic lectin with biological activity, confirming that the strategy followed provides an alternative for
the synthetic production and purification of this lectin.

1. Introduction (TBLF), which has a cytotoxic effect on colon cancer cells (García-Gasca
et al., 2012) and, under oral administration, displays low toxicity and
Lectins are proteins that interact with carbohydrates, either free or inhibits early colon tumorigenesis in rats by apoptosis (Ferriz-Martínez
bounded to cell membranes (Sharon and Lis, 2002). In plants, these et al., 2015; Moreno-Celis et al., 2017). The partial sequences of the two
proteins are found mainly in seeds and storage tissues, where they reach main lectins of TBLF (Lectin_A and Lectin_B) were identified by Torres-
up to 10% of the total proteins, whereas in leaves, roots, and stems, their Arteaga et al. (2016) through mass spectrometry sequencing. Also
concentration is lower (Liener, 1976; Etzler, 1985; Chrispeels and identified in these lectins was the presence of N-glycans, which were
Raikhel, 1991). Lectins from some species, mainly legumes, have shown shown to be necessary for their function.
an ability to selectively adhere to tumor-cell oligosaccharides and inhibit Extraction and purification of the Tepary bean lectins has been carried
their growth (Díaz et al., 1999; De Mejía and Prisecaru, 2005; Ferriz out through the use of a wide range of strategies. Unfortunately, these
Martínez et al., 2010). strategies commonly provide low yields (García-Gasca et al., 2012;
Tepary bean (Phaseolus acutifolius), which grows in the north of Torres-Arteaga et al., 2016). Therefore, it is necessary to develop a system
Mexico and in the south of the United States and its seeds contain high to obtain higher quantities of these lectins at low cost, without
concentration of lectins. By different chromatographic and electropho- compromising its functionality. Transformation facilities and modern
retic methods it was possible to obtain the Tepary bean lectin fraction plant tissue culture techniques allow genetically modified plants to

Abbreviations: IRM, induction and regeneration medium; PGIP, Phaseolus vulgaris polygalacturonase; PBS, phosphate buffer solution; cisTBL1, cisgenic Tepary bean
lectin; TBL, Tepary bean lectin; TBLF, Tepary bean lectin fraction.
$
This article was originally published in Journal of Biotechnology: X. Journal of Biotechnology: X is now discontinued and the article is republished here for the reader’s
convenience. For citation purposes, please use the publication details of this article; Journal of Biotechnology, 306S.
* Corresponding authors.
E-mail addresses: dania_667@hotmail.com (D. Martínez-Alarcón), mora_alejandra@yahoo.com (A. Mora-Avilés), alejandroblancolabra@gmail.com
(A. Blanco-Labra), tggasca@uaq.edu.mx (T. García-Gasca).
1
These authors share as first authors.

http://doi.org/10.1016/j.btecx.2019.100013
Received 26 April 2019; Received in revised form 5 August 2019; Accepted 4 September 2019
Available online 25 September 2019
0168-1656/© 2019 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
D. Martínez-Alarcón et al. Journal of Biotechnology 306S (2019) 100013

function as suitable bioreactors for the production of recombinant Escherichia coli Top10 chemocompetent cells by heat shock (Sambrook
proteins and/or industrially relevant metabolites (Giddings et al., 2000). et al., 1989). Transformed colonies were inoculated in LB medium with
Plants provide a suitable environment for protein folding and disulfide 100 mg L1 of ampicillin as the selection agent and incubated at 37  C for
bond formation, multimeric protein assembly, and post-translational 24 h. Finally, plasmids were purified by miniprep method (Birnboim and
modifications (Borisjuk et al., 1999). However, the biggest disadvantage Doly, 1979) and sequenced at the National Laboratory of Agricultural,
of producing proteins in genetically modified plants is that their Medical and Environmental Biotechnology (LANBAMA, San Luis Potosi,
purification involves tissue maceration which destroys the production Mexico).
system and the release of proteins of interest at all stages of production
(including partially folded or partially processed proteins), decreasing the 2.5. Cloning
efficiency of the process (Gaume et al., 2003; Cabanes-Macheteau et al.,
1999). To overcome these drawbacks, in 1999 Borisjuk et al. (Borisjuk A construction coupling the P. vulgaris polygalacturonase (PGIP)
et al., 1999) proposed, for the first time, a root-secretion system. This signal peptide at the 5-end, and 6xHis-tail at the 3-end of the Tepary bean
technique couples signaling peptides for the flux of functional proteins to lectin gene was synthesized by GenScript USA Inc. Once the construction
a hydroponic medium through the roots of genetically modified plants, was obtained, it was released from the commercial vector pUC57 by
providing higher yields than conventional extraction methods. As the digestion carried out with the BamHI restriction enzyme (Fermentas,
root-secretion system allows the expression of proteins from the original Ontario, Canada), and incubated at 37  C from 3 to 5 h. The sequence was
species, the correct folding and post-translational processing of complex introduced in a previously digested vector pBI121, by a ligation carried
proteins is assured, providing an alternative manufacturing platform that out with the T4 ligase (Fermentas, Ontario, Canada), incubating at 16  C
increases performance and simplifies the purification procedure (Drake for 16 h. Subsequently, the result of the previous ligation was cloned into
et al., 2009). Therefore, this could also represent an excellent alternative chemocompetent cells of E. coli Top 10 by heat shock (Sambrook et al.,
method of preserving the native N-glycans of leguminous lectins. As we 1989). The transformed colonies were recovered for growth on plates of
use one of the lectin genes from the same plant, the resulting product will LB medium with 100 mg/mL of ampicillin as selection agent, incubated at
be a cisgenic protein (Telem et al., 2013). Therefore, the objective of this 37  C for 24 h, and finally the plasmids were extracted by the miniprep
study was to transform Tepary bean plants in order to obtain a cisgenic method (Birnboim and Doly, 1979).
lectin by rhizosecretion and to evaluate the functionality of the mature
protein by hemagglutination analysis. 2.6. Modeling

2. Materials and methods To visualize the lectin molecular modeling, the program RASTOP
(Molecular Graphics Visualization Software) was used (Sayle and
2.1. Tepary bean lectin fraction Bernstein, 2007). The comparative modeling of the mRNA-deduced
protein sequence of the Tepary bean lectin was made using the following
Tepary bean lectin fraction was obtained by purification from Tepary five homology-based comparative modeling websites:
bean seeds, using Tris-HCl pH 8.0 as extraction solvent at 4  C, followed by
sequentialprecipitationwithammoniumsulfateat40%and60%saturation. 1 FUGUE-Homstrad: Sequence-structure homology recognition (Wil-
Finally, the protein extract was separated by molecular weight exclusion liams et al., 2001).
chromatography using G-75 Sephadex (Torres-Arteaga et al., 2016). 2 3D-JIGSAW—Comparative modeling (Bates et al., 2001).
3 CPHmodels 3.2 Server (Nielsen et al., 2010).
2.2. Phaseolus acutifolius specimens 4 ESyPred3D Web Server 1.0 (Lambert et al., 2002).
5 Geno3D: Automatic comparative molecular modelling of protein
Plants were germinated from Tepary bean seeds (P. acutifolius) (Combet et al., 2002).
purchased in the local market of Hermosillo, Sonora, Mexico. One specimen 2.7. Agrobacterium tumefaciens transformation
was deposited at the “Dr. J. Rzedowski” herbarium of the Natural Sciences
Faculty of the Autonomous University of Querétaro for its identification. Competent cells of A. tumefaciens strain GV2260 were transformed
with pBI121 (Promega, Madison, WI, USA) by the heat shock method
2.3. Preparation of the sample (Sambrook et al., 1989). Transformed cells were cultivated on solid LB
medium with 100 mg L1 of kanamycin as selection agent and incubated
Tepary bean seeds were previously washed with water, cultivated for at 28  C for 24–48 h. A single Agrobacterium colony was selected and
3 weeks and watered every third day. Subsequently, stems and leaves grown in a liquid culture to exponential growth (OD600 = 0.8).
were pulverized in a mortar with liquid nitrogen. Total RNA was extracted
using Trizol1 and incubated in the presence of DNase. The cDNA was 2.8. Phaseolus acutifolius transformation
generated by using the RevertAidTM H Minus First Strand Kit (# K1631
Fermentas, Ontario, Canada), according to the manufacturer's instruc- Tepary bean seeds were disinfected by the gas-chlorine method
tions. Subsequently, a polymerase chain reaction (PCR) was performed composed by hydrochloric acid and commercial bleach in a ratio of 5:0.16
with the degenerate primers F1 (50 - ATGGCTTCCTCCAACTTCTCCA-30 ), (v/v) for 8–10 h. Subsequently, bean seeds were soaked in sterile distilled
R1 (50 -TAGAGGATTTGGTTGAGGACGA-30 ), F2 (5-CCTCTTCCTTCCGC water for 17 h under aseptic conditions. After this time, embryos were
TTCTCAC-3) y R2 (5-AAGAGAGGAGGTCGTT CGTTTC-3). Those primers removed from the seeds and cultivated on petri dishes with induction and
were designed based on conserved sequences of 36 leguminous lectins regeneration medium (IRM), which consisted of solid Gamborg B5
from multiple origins. Amplification conditions were: 5 min at 95  C for medium (Gamborg et al., 1968), amended with 2% sucrose, 100 mg L1 of
one cycle; 1 min at 94  C, 1 min at 52  C, 2 min at 72  C for 35 cycles; and myo-inositol, 1 mg L1 of pyridoxine, mgL1 of thiamine, and 10 mg L1
7 min at 72  C for final extension. of BAP for 5 days at 25  2  C and a photoperiod of 16 h light and 8 h
darkness. Hypocotyls were obtained by dissecting apical and radicular
2.4. Sequencing meristems from five-day-old embryos. Liquid IRM was combined with A.
tumefaciens (O.D.600 = 0.8) in a 5:1 (v/v) ratio. Hypocotyls were
The PCR product was cloned into a pCR2.1-TOPO vector (Invitrogen incubated at room temperature for 10 min in this solution with gentle
Life Technologies, Carlsbad, CA, USA). Then, it was inserted into movements. Liquid was discarded and explants were placed in co-

2
D. Martínez-Alarcón et al. Journal of Biotechnology 306S (2019) 100013

Fig. 1. Multiple alignment of codifying sequences for leguminous lectins and primers design. A) Alignment of the 50 end of 36 leguminous lectins; green boxes indicate F1/
F2 primers and the location where they hybridize the consensus sequence. B) Alignment of the 30 end of 36 leguminous lectins; orange boxes indicate the complementary
sequences of R1/R2 primers and the location where they hybridize the consensus sequence. Conserved residues are indicated in blue, and the accession number of each
sequence is on the left.

cultivation medium, which consisted of IRM amended with 200 mM of selection medium to establish the hydroponic conditions for rhizosecre-
acetosyringone remaining in this medium for 5 days in the same tion. After seven days of incubation in liquid selection medium, an aliquot
environmental conditions previously mentioned. Hypocotyls were was collected and lyophilized.
transferred to IRM solid medium amended with 300 mg L1 of timentin
for Agrobacterium elimination for 6 days under the same conditions. 2.9. Periodic acid Schiff staining
Subsequently, hypocotyls were transferred to a selection medium which
consisted of IRM medium with 300 mg L1 of timentin and 50 mg L1 of Protein concentration was determined by the Bradford method
kanamycin added to it (Sigma Chemical Co., St. Louis, MO, USA). (Bradford, 1976). To verify the presence of glycosylations of the cisgenic
Explants were incubated for bud formation and shoot differentiation, lectin, 20 mL and 10 mL of crude root-exudate were run by SDS-PAGE,
with successive transfers to fresh medium every 6 days. Independent adding 40 mg native lectin as a positive control. Subsequently, they were
shoots were transferred to tall petri dishes with selection medium, where transferred to a nitrocellulose membrane, applying a current of 0.2 A for
they remained until reaching 1 cm height. Later, they were transferred to 60 min, and the membrane was stained using periodic acid–Schiff reagent
Magenta1 boxes with selection medium without growth regulators to (Sigma Chemical Co., St. Louis, MO, USA) according to the manufacturer's
promote elongation and root formation. Once the kanamycin-resistant instructions.
plants fully formed their rooting system, they were transferred to liquid

3
D. Martínez-Alarcón et al. Journal of Biotechnology 306S (2019) 100013

Fig. 2. Identification of lectins in transcripts of Phaseolus acutifolius. A) Amplification of sequences with different combinations of degenerate primers; molecular size
marker (1). F1/R1 (2), F2/R2 (3), F1/R2 (4), F2/R2 (5), amplicon purified from band excision of agarose electrophoresis (6). B) Polymerase chain reaction (PCR) of
transformed colonies, molecular size marker (1 and 16), PCR screening of E. coli colonies after transformation (2–15 and 17–21). C) Multiple alignment of the amplified
sequence from P. acutifolius (cisTBL1), the deduced sequence for P. acutifolius lectin (accession number: AAA82181.1) and the peptides obtained by mass spectrometry for
Lectin-A (LA_SequencedPeptides) and Lectin-B (LB_SequencedPeptides). Matches are indicated in red and mismatches are indicated in blue and black.

2.10. Immunoblotting USA). Subsequently, the columns were washed with an ascending
imidazole gradient until the desired product was recovered.
To verify the presence of cisgenic lectin in root exudates of the
genetically modified plants, 25 mL of each sample was placed in 2.12. Hemagglutination assay
nitrocellulose membranes previously activated with 1X TBS. The
membranes were left to dry for 15 min and then blocked for 15 min Glutaraldehyde fixed type A + erythrocytes (0.5% in PBS) (Turner
with anti-histidine primary monoclonal antibodies coupled to alkaline and Liener, 1975) were used for testing the hemagglutination activity of
phosphatase (Novus Biological Europe, Science Park, CAM, UK), in a rTBL1 (García-Gasca et al., 2012; Jaffé, 1980), protein concentration was
1:1000 dilution. Finally, the membranes were revealed using 3,3- determined by Bradford method (Bradford, 1976). Then, TBLF (8.2 mL,
diaminobenzidine (Sigma Chemical Co, St. Louis, MO, USA). 324.7 mg P/mL) was used as a positive control and PBS (100 mL) was used
as a negative control. The tested samples were 100 mL of non-transformed
2.11. Purification of the cisgenic protein from the culture media radicular exudate and 100 mL of genetically modified radicular exudate
(26.76 mg P/mL). Agglutination was determined using an inverted
The supernatant was diluted with phosphate buffer 4X pH: 7.4 in a 1:3 microscope after incubation at 37  C for 2 h.
ratio and flowed through 5 mL nickel columns (Invitrogen Carlsbad, CA,

4
D. Martínez-Alarcón et al. Journal of Biotechnology 306S (2019) 100013

Fig. 3. Modeling the structure of the deduced lectin. Models generated by EsyPred3D (A), CPH-models (B), Geno3D (C), 3D-JIGSAW (D). X: Front view. Y: Top view. The
order of the polypeptide sequence is indicated by a gradual change of color from blue (N-terminal end) to red (C-terminal end).

Fig. 4. Matched model of the predicted protein structure of lectins phylogenetically related to TBL1. A) Matched model without glycans. The level of conservation is
indicated in color scale from blue to red, blue being the most conserved regions and red the less conserved ones. B) Matched model with glycans: calcium ions (yellow),
manganese ion (green), and glycosylation (red). X. Front view. Y: Top view.

3. Results were sequenced independently and, based on multiple alignments, it was


determined a sequence of 831 nucleotides corresponded to a polypeptide
3.1. Analysis of lectin transcripts from P. acutifolius chain of 277 amino acids, later referred to as Tepary bean lectin 1 (TBL1).
This sequence displayed high identity with La and LB proteins obtained by
mRNA amplification of a Tepary bean lectin was performed using Torres-Arteaga et al. (2016), and with a P. acutifolius lectin previously
degenerated primers designed to bind conserved sequences of leguminous deduced by Mirkov et al. (1994) (accession number: AAA82181.1). The
lectins (Fig. 1). The amplification of lectin transcripts was specific, and the coverage percentage of identity was 97.83%, with 6 amino acid changes in
size ranged from 740 to 894 bp. The largest product was purified and position 17 P changed to A, in position 139 R changed to K, in position 180
introduced in a TOPO vector for subsequent transformation of E. coli GQ changed to VN, in position 228 R changed to S, and in position 239 T
(Fig. 2A). Nineteen colonies resulted positive from transformation, and changed to S (Fig. 2C). In contrast with the amplified sequence, none of the
plasmid purification was performed for three of them (Fig. 2B). Vectors sequences reported by Torres-Arteaga et al. (2016) displayed the N-

5
D. Martínez-Alarcón et al. Journal of Biotechnology 306S (2019) 100013

Fig. 5. Phaseolus acutifolius transformation. A) Synthesized nucleotide construction scheme. B–F) Development stages of P. acutifolius regeneration. A) Four-day-old
embryo germinated in induction and regeneration medium. B) Dissection of embryo to obtain the hypocotyl. C) Development of de novo organogenic buds. D)
Differentiation of kanamycin resistant shoots. E) Development of kanamycin resistant complete plants.

terminal peptide, indicating that this signal peptide is cleaved during carbohydrate-binding pocket in all models was found adjacent to calcium
protein processing. No additional lectin transcripts were identified in the and manganese divalent cations, which are also necessary for carbohy-
transcripts; however, the possibility that they are present is not ruled out. drate binding activity. Those structures also revealed that, in the
monomeric conformation, the C- and N- terminals are exposed,
3.2. TBL1 structure suggesting that their possible modification should not compromise its
quaternary structure.
Conformational modeling of the deduced polypeptide sequence
showed that TBL1 has a large proportion of beta sheets interconnected 3.3. Genetic construction design
by small regions of helices (Fig. 3). These data are consistent with
previous reports of legume lectins and provide a good explanation of the Based on the structural studies of these models, a genetic construction
high resistance that TBLF lectins display against degradation in the was designed for the synthetic expression of TBL1. The signal peptide of
gastrointestinal tract (Ferriz-Martínez et al., 2015). PGIP was coupled to the 50 end of the TBL nucleotide sequence and a
Multiple alignment of polypeptide lectin sequences from different 6xHis-tail was coupled to the 30 end followed by a stop codon. The
legumes revealed that the glycosylation sites are highly conserved construction was flanked by cutting sites for BamHI and codon
between species and are distant from the C-terminus and N-terminus, optimization was done in order to avoid non-specific internal cleavage
none of these sites coincide with differential residues between the TBL1 by restriction enzymes. Insert and vector were digested with BamHI, and
sequence and the peptides reported by Torres-Arteaga et al. (2016) for the construction was introduced into pBI121 under regulation of the
TBLF lectins (Fig. 4), suggesting that they are conserved. The CaMV 35 s promoter and NOS terminator (Fig. 5A).

6
D. Martínez-Alarcón et al. Journal of Biotechnology 306S (2019) 100013

Fig. 6. Characterization and purification of the cisgenic lectin in root exudates. A) SDS-PAGE stained by periodic acid–Schiff (PAS) reaction; (1) Positive control (10 mg of
TBL), (2 and 3) Root exudates from transformed Tepary bean plants (20 and 10 ml, respectively), (4) Negative control, radicular exudate of non-transformed Tepary bean
plants. B) Identification of cisgenic proteins by immunoblotting. On the left: sample (25 mL of root exudate from transgenic plants); on the right: negative control (25 mL of
root exudate from non-transformed plants). C) Purification of cisTBL1 from root exudates. (1) Elution with 200 mM phosphate buffer of Imidazole (10 mL, 1.2 mg) after
desalination, (2) Root exudate from a transformed plant prior to purification (20 mL), (3) Positive control (10 mg TBLF). D) Biological activity of root exudates. The first
row shows inverted microscopy images with a 10X magnification objective, whereas the second row shows these same regions with 20X magnifications. Column 1) PBS
negative control, Column 2) Root exudates from non-transformed plants, Column 3) Root exudates of genetically modified plants, Column 4) Positive control of native
lectin (TBLF).

3.4. Genetic transformation of Phaseolus acutifolius TBLF (Fig. 6C). These data also evidenced the absence of phase shifting
during transcription, as the His-tail introduced at the C-end of the protein
Tepary bean germination rate was between 90% and 95%. Five days was successfully translated. The cisTBL1 yield ranged between 12.21 and
after germination, embryos were dissected and cultivated in germination 20.97 mg of cisgenic protein per gram of dry root.
medium, and apical and radical meristems were cut off to obtain the
hypocotyl explant (Fig. 5B and C). After 10 days, the first direct 3.6. Agglutination activity of root exudates of genetically modified plants
organogenic buds were formed on the hypocotyl in selection medium
(Fig. 5D). Kanamycin-resistant differentiated shoots were separated from The agglutination test using human A + erythrocytes revealed that
the original explant and transferred to an elongation medium for further the root exudates of transgenic plants presented similar biological activity
growth and root formation (Fig. 5E and F). Once primary roots developed, with respect to TBLF (11,958.15 and 12,018.59 AU/mg protein), whereas
the plants were transferred to a liquid medium. no agglutination in root exudates from non-transformed plants was
observed (Fig. 6D).
3.5. Characterization and purification of cisTBL1
4. Discussion
Seven days after transferring the plants to hydroponic conditions, a
sample of crude culture medium was analyzed by periodic acid Schiff High identity between the amplified transcript with LA and LB
staining on SDS-PAGE revealing the presence of a unique glycoprotein in peptides indicates that TBL1 is one of the main components of TBLF. This
root exudates of genetically modified Tepary bean plants, whereas no information was later confirmed by the characterization and purification
evidence of a similar band in root exudates from wild-type plants was of the cisgenic product, where it was found that TBL1 corresponds to the
found (Fig. 6A). Subsequently, the presence of cisTBL1 in the root lowest molecular weight lectin observable in the electrophoretic profile of
exudates of genetically modified plants was confirmed by a dot-blot assay TBLF. Although it is true that cisTBL1 has a slightly larger size than its
with anti-histidine antibodies, but no presence of this protein was native counterpart, the alteration of the electrophoretic migration pattern
detected in root exudates of non-transformed plants (Fig. 6B). After could be attributed to the addition of 6xHis-tail at its C-terminal end.
purification by nickel affinity chromatography, a single protein was Structural models allow us to propose a three-dimensional structure of
obtained, which corresponded in size to the shorter lectin present in the TBL1 and its prosthetic groups, also allowing the identification of

7
D. Martínez-Alarcón et al. Journal of Biotechnology 306S (2019) 100013

common motifs between lectins, and to identify possible interferences Cabanes-Macheteau, M., Fitchette-Lainé, A.C., Loutelier-Bourhis, C., Lange, C., Vine, N.D.,
Ma, P., Lerouge, J.K.C., Faye, L., 1999. N-Glycosylation of a mouse IgG expressed in
where modifications are proposed. On the other hand, immunoblot transgenic tobacco plants. Glycobiol 9, 365.
analysis indicates that cisTBL1 was successfully secreted through the root Chrispeels, M.J., Raikhel, N.V., 1991. Lectins, lectin genes, and their role in plant defense.
exudates of genetically modified plants, and the Schiff staining assay Plant Cell 3, 1.
Combet, C., Jambon, M., Deléage, G., Geourjon, C., 2002. Geno3D: Automatic comparative
confirms that this protein was glycosylated, indicating that PGIP signal molecular modelling of protein. Bioinformatics 18, 213.
peptide successfully redirected the protein through the secretory De Mejía, E.G., Prisecaru, V.I., 2005. Lectins as bioactive plant proteins: a potential in
pathway. cancer treatment. Crit. Rev. Food Sci. Nutr. 45, 425.
Díaz, P.H., González, O.M., De Pablos Vélez, Y.R., Ganem Báez, C., 1999. Aplicaciones de
Finally, the agglutination analysis confirmed that the excreted las lectinas. Rev. Cuba. Hematol. Inmunol. Hemoter. 15, 91.
product maintained the carbohydrate-binding properties of the native Drake, P.M., Barbi, T., Sexton, A., McGowan, E., Stadlmann, J., Navarre, C., Paul, M.J.,
lectins of Tepary bean. Native lectins of TBLF have the ability to Ma, J.K., 2009. Development of rhizosecretion as a production system for recombinant
proteins from hydroponic cultivated tobacco. FASEB J. 10, 3581.
agglutinate A + erythrocytes. As lectins that are not properly folded lose
Etzler, M.E., 1985. Plant lectins: molecular and biological aspects. Ann. Rev. Plant Physiol.
their carbohydrate-binding activity, cisTBL1 agglutination suggest a 36, 209.
proper protein folding. Therefore, it is concluded that the proposed Ferriz Martínez, R.A., Torres-Arteaga, I.C., Blanco-Labra, A., García-Gasca, T., 2010. The
method allows the successful production and secretion of the bioactive role of plant lectins in cancer treatment. In: Mejia-Vazquez, C., Navarro-Fos, S. (Eds.),
New Approaches in the Treatment of Cancer, Vol. 1. Nova Science Publishers, Mexico,
lectin towards root exudates of genetically modified plants, providing a pp. 71–89 Ch. 4.
continuous production platform that avoids the inconveniences associat- Ferriz-Martínez, R.A., García-García, K., Torres-Arteaga, I., Rodríguez-Méndez, A.J.,
ed with the purification of lectins from seeds. The fact that the Guerrero-Carrillo, M.J., Moreno-Celis, U., Ángeles-Zaragoza, M.V., Blanco-Labra, A.,
Gallegos-Corona, M.A., Robles-Álvarez, J.P., Mendiola-Olaya, E., Andrade-
agglutination activity was conserved suggests that there is no change Montemayor, H.M., García, O.P., García-Gasca, T., 2015. Tolerability assessment of a
in the lectin molecular structure; however, this needs to be confirmed. lectin fraction from Tepary bean seeds (Phaseolus acutifolius) orally administered to rats.
Further studies will focus on optimizing this procedure, as well as on Toxicol. Reports. 2, 63.
Gamborg, O.L., Miller, R.A., Ojima, E., 1968. Nutrient requirements of suspension cultures
characterizing the cisgenic protein produced and its effects on cancer of soybean root cells. Cell Res. 50, 151.
cells. García-Gasca, T., Hernández-Rivera, E., López-Martínez, J., Castañeda, A.L., Yllescas-
Gasca, L., Rodríguez-Méndez, A.J., Mendiola-Olaya, E., Castro-Guillén, J.L., Blanco-
Labra, A., 2012. Effects of Tepary bean (Phaseolus acutifolius) protease inhibitor and
Author contributions semipure lectin fractions on cancer cells. Nutr. Cancer 64, 1269.
Gaume, A., Komarnytsky, S., Borisjuk, N., Raskin, I., 2003. Rhizosecretion of recombinant
Dania Martínez-Alarcón, Arantxa Espinoza-Núñez, Luz M Serrano proteins from plant hairy roots. Plant Cell Rep. 21, 1188.
Giddings, G., Allison, G., Brooks, D., Carter, A., 2000. Transgenic plants as factories for
Jamaica performed the experiments, Alejandra Mora-Avilés advised
biopharmaceuticals. Nat. Biotechnol. 18, 1151.
plant culture experiments, Andrés Cruz-Hernández and Angelina Jaffé, W.G., 1980. Hemagglutinins. Toxic Constituents of Plant Foodstuffs. Academic Press,
Rodríguez-Torres advised genetic transformation experiments, José L New York, NY, USA, pp. 73–102.
Castro-Guillen advised the molecular modeling, Alejandro Blanco-Labra Lambert, C., Leonard, N., De Bolle, X., Depiereux, E., 2002. ESyPred3D: Prediction of
proteins 3D structures. Bioinformatics 18, 1250.
and Teresa García-Gasca directed and advised the complete work and Liener, I., 1976. Phytohemagglutinins. Ann. Rev. Plant Physiol. 27, 291.
managed the resources. Mirkov, T.E., Wahlstrom, J.M., Hagiwara, K., Finardi-Filho, F., Kjemtrup, S., Chrispeels,
M.J., 1994. Evolutionary relationships among proteins in the phytohemagglutinin
Declaration of Competing Interest arcelin-alpha amylase inhibitor family of the common bean and its relatives. Plant Mol.
Biol. 26, 1103.
The authors declare no financial or commercial conflict of interest. Moreno-Celis, U., López-Martínez, J., Blanco-Labra, A., Cervantes-Jiménez, R., Estrada-
Martínez, L.E., García-Pascalin, A.E., Guerrero-Carrillo, M.J., Rodríguez-Méndez, A.J.,
Acknowledgments Mejía, C., Ferríz-Martínez, R.A., García-Gasca, T., 2017. Phaseolus acutifolius lectin
fractions exhibit apoptotic effects on colon cancer: preclinical studies using
dimethilhydrazine or azoxi-methane as cancer induction agents. Molecules 22, 1670.
We would like to thank F. Josué López Martínez and Elizabeth Nielsen, M., Lundegaard, C., Lund, O., Petersen, T.N., 2010. CPHmodels-3.0 - Remote
Mendiola-Olaya for their technical assistance, and Consejo Nacional de homology modeling using structure guided sequence profiles. Nucl. Acids Res. 576.
Sambrook, J., Fritsh, E., Maniatis, T., 1989. Molecular Cloning: A Laboratory Manual. Cold
Ciencia y tecnología (CONACYT) Programa de Fortalecimiento de la
Spring Harbor Laboratory Press, NY.
Calidad Educativa (PFCE) Ciencia Básica (CB-2014-01-241181) and Sayle, R., Bernstein, H.J., 2007. ValadonP. Molecular Graphics Visualisation Software.
PFCE program for their financial support. Version 2.1. .
Sharon, N., Lis, H., 2002. How proteins bind carbohydrates: lessons from legume lectins. J.
Agric. Food Chem. 50, 6586–6591.
References Telem, R.S., Wani, S.H., Singh, N.B., Nandini, R., Sadhukhan, R., Bhattacharya, S.,
Mandal, N., 2013. Cisgenics - a sustainable approach for crop improvement. Curr.
Bates, P.A., Kelley, L.A., MacCallum, R.M., Sternberg, M.J.E., 2001. Enhancement of Genomics 14 (7), 468–476.
protein modelling by human intervention in applying the automatic programs 3D- Torres-Arteaga, I., Mendiola-Olaya, E., García-Gasca, T., Ángeles-Zaragoza, M.V., García-
JIGSAW and 3D-PSSM. Proteins 5, 39. Santoyo, V., Castillo, J.A., Aguirre, C., Phinney, B., Blanco-Labra, A., 2016.
Birnboim, H.C., Doly, J., 1979. A rapid alkaline extraction procedure for screening Characterization of two non-fetuin-binding lectins from Tepary bean (Phaseolus
recombinant plasmid DNA. Nucl. Acids Res. 7, 1513. acutifolius) seeds with differential cytotoxicity on colon cancer cells. J. Glycobiol. 5, 1.
Borisjuk, N.V., Borisjuk, L.C., Logendra, S., Petersen, F., Gleba, Y., Raskin, I., 1999. Turner, R.H., Liener, I.E., 1975. The use of glutaraldehyde-treated erythrocytes for assaying
Production of recombinant proteins in plant root exudates. Nat. Biotechnol. 17, 466– the agglutinating activity of lectins. Anal. Biochem. 68 (2), 651–653.
469. Williams, M.G., Shirai, H., Shi, J., Nagendra, H.G., Mueller, J., Mizuguchi, K., Miguel, R.
Bradford, M.A., 1976. Rapid and sensitive method for the quantification of microgram N., Lovell, S.C., Innis, C.A., Deane, C.M., Chen, L., Campillo, N., Burke, D.F., Blundell,
quantities of protein utilizing the principle of protein-dye-binding. Anal. Biochem. 72, T.L., de Bakker, P.I., 2001. Sequence-structure homology recognition by iterative
248–252. alignment refinement and comparative modeling. Proteins 5, 92.

8
Food Research International 147 (2021) 110487

Contents lists available at ScienceDirect

Food Research International


journal homepage: www.elsevier.com/locate/foodres

Review

Genetic engineering of yeast, filamentous fungi and bacteria for terpene


production and applications in food industry
Zijian Liang, Hang Zhi, Zhongxiang Fang, Pangzhen Zhang *
School of Agriculture and Food, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 3010, Australia

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

Keywords: Terpenes are a major class of natural aromatic compounds in grapes and wines to offer the characteristic flavor
Terpenes and aroma, serving as important quality traits of wine products. Saccharomyces cerevisiae represents an excellent
Saccharomyces cerevisiae cell factory platform for large-scale bio-based terpene production. This review describes the biosynthetic path­
Escherichia coli
ways of terpenes in different organisms. The metabolic engineering of S. cerevisiae for promoting terpene
Fungi
biosynthesis and the alternative microbial engineering platforms including filamentous fungi and Escherichia coli
Metabolic engineering
Food applications are also elaborated. Additionally, the potential applications of the terpene products from engineered microor­
ganisms in food and beverage industries are also discussed. This review provides comprehensive information for
an innovative supply way of terpene via microbial cell factory, which could facilitate the development and
application of this technique at the industrial scale.

terpenes exclusively through the MEP pathway. In the fermentation


process such as winemaking, many S. cerevisiae strains can enzymati­
1. Introduction cally hydrolyze the bound terpenes in grape must and thus release the
free volatile counterparts in wine (Liang, Fang, et al., 2020). S. cerevisiae
Terpenes and their derivative terpenoids are a major class of natural strains can also utilize the MVA pathway to yield various monoterpenes
aromatic compounds present in both free and glycosidically bound but the amount is low due to the deficiency of monoterpene synthases
forms in many edible plants and their fruits, to offer a strong odor which (MTSs) (Pardo et al., 2015).
may protect the plants by deterring herbivores and by attracting pred­ Solvent extraction from plant materials is the conventional process
ators and parasites of herbivores (Liang, Fang, et al., 2020). They are for commercial terpene production, although with multiple demerits
structurally related to isoprene (C5H8) and classified as monoterpenes such as time-consuming and labor-intensive process, low yield and
(C10), norisoprenoids (C13), sesquiterpenes (C15), diterpenes (C20), environment unfriendliness (Liang, Zhang, & Fang, 2020). In recent
triterpenes (C30) and tetraterpenes (C40) based on the number of years, terpene production via microbial cell factories has emerged as a
isoprene units (Li, Howell, Fang, & Zhang, 2020). Terpenoids are promising alternative to the traditional method (Paramasivan & Mut­
modified from terpenes and usually contain oxygen atoms. This article turi, 2017). Microorganism serves as an efficient terpene-producing
uses the term terpenes to represent terpenes and terpenoids for easier platform for being fast-growing, land-saving and controllable. Yeast
discussion. Terpenes derived from grapes, especially monoterpenes, are especially S. cerevisiae with a clear genetic background, is the well-
responsible for various sensory characteristics of wine products, such as established host to express heterologous terpene synthase (TPS) genes.
citrus, fruity and floral flavors (Li et al., 2020). Additionally, terpenes Similar to plant cells, S. cerevisiae contains an inherent endomembrane
are engaged in the pharmaceutical industry due to their multiple bio­ system for microsomal enzymes to stand on (Sun, Zhao, & Li, 2019). A
activities, including antioxidative, antimicrobial and anti-inflammatory rational strain improvement by metabolic engineering can enhance
properties (Pardo, Rico, Gil, & Orejas, 2015). From the chemical aspect, terpene production in S. cerevisiae. Overexpression of HMG-CoA (3-hy­
terpenes are mainly derived from isopentenyl pyrophosphate (IPP) and droxy-3-methylglutaryl-CoA) reductase (HMGR) variant, ERG20 over­
its isomer dimethylallyl pyrophosphate (DMAPP) via mevalonate (MVA) expression, ERG9 downregulation, integration of extra IDI1 gene and
and methylerythritol-phosphate (MEP) pathways (Zhang et al., 2016). deletion of ROX1 constitute the main engineering strategies to modify
Both MVA and MEP pathways exist in plant cells. However, yeast and the MVA pathway in S. cerevisiae (Paramasivan & Mutturi, 2017). In
other fungi only harbor the MVA pathway, while bacteria produce

* Corresponding author.
E-mail address: pangzhen.zhang@unimelb.edu.au (P. Zhang).

https://doi.org/10.1016/j.foodres.2021.110487
Received 26 March 2021; Received in revised form 21 May 2021; Accepted 23 May 2021
Available online 31 May 2021
0963-9969/© 2021 Elsevier Ltd. All rights reserved.
Z. Liang et al. Food Research International 147 (2021) 110487

Nomenclature CyaA adenylate cyclase


cAMP cyclic adenosine monophosphate
IPP isopentenyl pyrophosphate CRP cAMP receptor protein
DMAPP dimethylallyl pyrophosphate CPPS ent-copalyl diphosphate synthase
MVA mevalonate KS ent-kaurene synthase
MEP methylerythritol-phosphate AA-CoA acetoacetyl-CoA
TPS terpene synthase MVAP mevalonate-5-phosphate
MTS monoterpene synthase MVAPP mevalonate-5-pyrophosphate
STS sesquiterpene synthase IP isopentenyl phosphate
DTS diterpene synthase DXP 1-deoxy-D-xylulose-5-phosphate
HMG-CoA 3-hydroxy-3-methylglutaryl-CoA CD-ME 4-(cytidine 5′ -diphospho)-2-C-methyl-D-erythritol
HMGR HMG-CoA reductase MEC 2C-methyl-D-erythritol-2,4-cyclodiphosphate
HMGS HMG-CoA synthase AACT acetoacetyl-CoA thiolase
GPP geranyl diphosphate MVK mevalonate kinase
GPPS GPP synthase PMK MVAP kinase
FPP farnesyl diphosphate MDD MVAPP decarboxylase
FPPS FPP synthase MPD MVAP decarboxylase
GGPP geranylgeranyl diphosphate IPK IP kinase
GGPPS GGPP synthase DXS DXP synthase
IDS isoprenyl diphosphate synthase DXR DXP reductoisomerase
G3P glyceraldehydes-3-phosphate MCT CD-ME cytidylyltransferase
IDI isopentenyl diphosphate isomerase CMK CD-ME kinase
MCT 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase MDS MEC synthase
CD-ME 4-(cytidine 5′ -diphospho)-2-C-methyl-D-erythritol HDS HMBPP synthase
HMBPP 1-hydroxy-2-methyl-2-(E)-butenyl 4-diphosphate HDR HMBPP reductase
NADH nicotinamide adenine dinucleotide GPPS GPP synthase
NADPH nicotinamide adenine dinucleotide phosphate FPPS FPP synthase
ADA acetaldehyde dehydrogenase (acylating) GGPPS GGPP synthase
xPK xylulose-5-phosphate specific phosphoketolase SS squalene synthase
PTA phosphotransacetylase KO-KAH kaurene oxidase-kaurenoic acid hydroxylase
UAS upstream activating cis-element CPR cytochrome P450 reductase
SUE sterol uptake enhancement UDP uridine diphosphate
NPP neryl diphosphate UGT UDP glucose-dependent glucosyltransferase
NPPS NPP synthase PKS polyketide synthase
ABC ATP-binding cassette PT prenyltransferase
MIC minimum inhibitory concentration FAD flavin adenine dinucleotide
GRAS generally recognized as safe FMO FAD-dependent monooxygenase
CRISPR clustered regularly interspaced short palindromic repeat AT acetyltransferase
sgRNA single guide RNA MT methyltransferase
AMA1 autonomous maintenance in Aspergillus SDR short-chain dehydrogenase/reductase
HA hemagglutinin TS taxadiene synthase
ATMT Agrobacterium tumefaciens-mediated transformation GMC glucose-methanol-choline
LB left border GRAS generally recognized as safe
RB right border

addition, construction of orthogonal pathway also exhibits great po­ highlights the future prospect of fungi as a reservoir of heterologous TPS
tential for efficient monoterpene production in S. cerevisiae (Ignea et al., genes, and filamentous fungi and E. coli as the alternative hosts to
2019). Nowadays, nearly 100 TPS genes have been identified in fungi S. cerevisiae in terpene synthesis, and further discusses the applications
(Zhang et al., 2020) and many of them are potential heterologous genes of the microbial engineering derived terpene products in food industry.
for the functional TPSs expression in S. cerevisiae, indicating fungi as a This review provides guidance for future researches and industrial ap­
reservoir of heterologous TPS genes. Filamentous fungi especially plications in terpene bioproduction from genetically engineered
Aspergillus oryzae can act as an alternative platform for terpene biosyn­ microorganisms.
thesis. Likewise, E. coli is also a common terpene-producing host, which
is highly efficient and more tolerant of monoterpene toxicity (Dugar & 2. Terpene biosynthesis
Stephanopoulos, 2011; Ingy & Wim, 2017).
So far, terpene products from engineered microorganisms are mainly Monoterpenes, sesquiterpenes and diterpenes as major terpene sub­
from the S. cerevisiae and E. coli platforms (Lyu, Lee, & Chen, 2019). classes are respectively produced from the corresponding intermediates
These products have displayed great potential in food applications geranyl diphosphate (GPP), farnesyl diphosphate (FPP) and ger­
including flavoring agents, sweetening agents, preservatives and func­ anylgeranyl diphosphate (GGPP) through MVA and MEP pathways
tional food ingredients, indicating the latent commercial value of the (Fig. 1A) (Zhuang, 2013). These intermediates are formed by a contin­
microbial technology. This review aims to investigate the MVA and MEP uous condensation of IPP and DMAPP from head to tail, then subjected
pathways of terpene biosynthesis and the metabolic engineering of to cyclization and post modifications (e.g., acetylation, oxidation) to
S. cerevisiae for promoting terpene production. The article also generate terpene products (Wang, Quan, & Xiao, 2019).

2
Z. Liang et al. Food Research International 147 (2021) 110487

Fig. 1. Terpene biosynthesis (A) via mevalonate (MVA) and/or methylerythritol-phosphate (MEP) pathways in plant (B), Saccharomyces cerevisiae (C) and Escherichia
coli (D). Dashed arrow lines represent multiple steps. TCA cycle, tricarboxylic acid cycle; AA-CoA, acetoacetyl-CoA; HMG-CoA, 3-hydroxy-3-methylglutaryl-CoA;
MVA, mevalonate; MVAP, mevalonate-5-phosphate; MVAPP, mevalonate-5-pyrophosphate; IP, isopentenyl phosphate; IPP, isopentenyl pyrophosphate; DMAPP,
dimethylallyl pyrophosphate; G3P, glyceraldehyde 3-phosphate; DXP, 1-deoxy-D-xylulose-5-phosphate; MEP, methylerythritol-phosphate; CD-ME, 4-(cytidine 5′ -
diphospho)-2-C-methyl-D-erythritol; MEC, 2C-methyl-D-erythritol-2,4-cyclodiphosphate; HMBPP, 4-hydroxy-3-methylbut-2-enyl diphosphate; GPP, geranyl pyro­
phosphate; FPP, farnesyl diphosphate;; GGPP, geranylgeranyl pyrophosphate; AACT, acetoacetyl-CoA thiolase; HMGS, HMG-CoA synthase; HMGR, HMG-CoA
reductase; MVK, mevalonate kinase; PMK, MVAP kinase; MDD, MVAPP decarboxylase; MPD, MVAP decarboxylase; IPK, IP kinase; IDI, isopentenyl diphosphate
isomerase; DXS, DXP synthase; DXR, DXP reductoisomerase; MCT, CD-ME cytidylyltransferase; CMK, CD-ME kinase; MDS, MEC synthase; HDS, HMBPP synthase;
HDR, HMBPP reductase; GPPS, GPP synthase; FPPS, FPP synthase; GGPPS, GGPP synthase.

The structural diversity of terpenes is determined by three enzymes condensed by squalene synthase (Erg9p) to yield squalene acting as a
isoprenyl diphosphate synthase (IDS), TPS and cytochrome P450. IDS is pivotal precursor of triterpene and sterol (Wang, Quan, et al., 2019).
responsible for controlling the branching points of terpene biosynthesis In MEP pathway, glyceraldehydes-3-phosphate (G3P) and pyruvate
by catalyzing the sequential condensation of IPP and DMAPP to form the are successively subjected to condensation and reduction to yield MEP,
intermediates with different chain lengths (Wang & Ohnuma, 1999). then catalyzed by 4-(cytidine 5′ -diphospho)-2-C-methyl-D-erythritol
TPSs can catalyze carbocation-driven cyclization, rearrangement, and (CD-ME) cytidylyltransferase (MCT) to form CD-ME. CD-ME is further
elimination reactions on acyclic prenyl diphosphate precursors, leading converted to 1-hydroxy-2-methyl-2-(E)-butenyl 4-diphosphate (HMBPP)
to the formation of a vast variety of acyclic and cyclic terpene scaffolds by phosphorylation, cyclization and ring opening. Then, one DMAPP
(Tholl, 2006). Cytochrome P450 is another modifying enzyme to add and five IPP molecules are simultaneously generated from HMBPP under
diversity to terpene structure through oxidation, hydroxylation, as well the catalysis of HMBPP reductase (HDR). DMAPP and IPP can be
as ring rearrangement and closure (Zhou & Pichersky, 2020). condensed by corresponding catalytic enzymes to form GPP, FPP and
GGPP, thus contributing to the production of different terpenes. The
biosynthesis of sesquiterpenes has been detailed in our previous review
2.1. Terpene biosynthesis in plant (Li et al., 2020). It is worth mentioning that GGPP also contributes to the
biosynthesis of norisoprenoids (Wang, Quan, et al., 2019).
In plant cells, MVA and MEP pathways are primarily employed to
meet the biosynthetic need for diverse terpenes (Fig. 1B). The MVA
pathway starts with the successive condensation of three molecules of 2.2. Terpene biosynthesis in yeast, other fungi and bacteria
acetyl-CoA into HMG-CoA via acetoacetyl-CoA thiolase encoded by
ERG10 and HMG-CoA synthase (HMGS) encoded by ERG13. Afterward, S. cerevisiae is the most widespread yeast species responsible for
HMG-CoA is catalyzed by HMGR to form mevalonate. Mevalonate is fermentations in food and beverage production. Similar to plant cells, S
further subjected to a two-step phosphorylation and decarboxylation to cerevisiae also serves as a production host for diverse terpenes but only
produce IPP that can be isomerized to DMAPP by isopentenyl diphos­ harbors the MVA pathway within the cytoplasm (Zhuang, 2013)
phate isomerase (IDI) encoded by IDI1. One DMAPP and two IPP mol­ (Fig. 1C). With acetyl-CoA as the primary precursor, GPP, FPP, GGPP
ecules are further acted upon by FPP synthase (FPPS) encoded by ERG20 and squalene are natively synthesized in the yeast as the precursors of
to form FPP, a precursor of sesquiterpene. Two molecules of FPP can be terpenes, norisoprenoids and sterols (Paramasivan & Mutturi, 2017).

3
Z. Liang et al. Food Research International 147 (2021) 110487

Likewise, other fungi only possess the MVA pathway for terpene pro­ 3.1. Overexpression of HMG-CoA reductase (HMGR) variant
duction (Zhang, Nielsen, & Liu, 2017). Fungi contain a large quantity of
TPS genes that can encode crucial enzymes for terpene biosynthesis HMGR was identified as a key rate-limiting enzyme in the MVA
(Zhang et al., 2020). Sesquiterpenes, diterpenes and triterpenes are the pathway in the 1980s, which is responsible for the conversion of HMG-
major classes of terpenes found in fungi (Quin, Flynn, & Schmidt- CoA into mevalonate in yeast (Bach, 1986). Hmg1p and Hmg2p are two
Dannert, 2014). Most bacteria especially E. coli produce terpenes reductase isozymes separately encoded by HMG1 and HMG2, thereinto,
exclusively through the MEP pathway (Fig. 1D), which can be sub­ Hmg1p plays a bigger role by expressing about 83% of HMGR activity
divided into two modules as that of plant cell: the feeding module where and presents in a more stable form (Wang, Quan, et al., 2019). HMGR is
G3P and pyruvate are generated from sugar substrates and the MEP composed of two terminal domains. The hydrophobic N-terminus an­
module which produces terpenes as the final product (Liu, Sun, et al., chors the protein to endoplasmic reticulum membrane, while the hy­
2013). drophilic C-terminus possesses all the enzyme activity and is oriented for
projection into the cytoplasm (Liscum et al., 1985). Overexpression of
3. Engineering of Saccharomyces cerevisiae HMGR is known to promote the MVA pathway, despite it is still nega­
tively affected by feedback regulation (DeBose-Boyd, 2008). The trans­
The metabolic engineering of S. cerevisiae is based on the modifica­ lation of Hmg1p may be regulated by the upstream intermediates such
tion in the MVA and orthogonal pathways employing genetic tools and as mevalonate, although the actual mechanism remains unknown. In
techniques, which is mainly ascribed to two actions: increasing pre­ contrast, the downstream intermediates such as sterol and nonsterol
cursor supply and downregulating competing pathways (Paramasivan & isoprenoids impose feedback inhibition on Hmg2p, especially at high
Mutturi, 2017). Vector is the most common genetic tool responsible for pathway flux. Nonsterol signal GGPP can change Hmg2p folding, lead­
delivering and manipulating foreign DNA inside a host cell. Plasmid is ing to Hmg2p degradation that is further stimulated by oxysterol (Burg
recommended as a viable vector due to its small size and easy manip­ & Espenshade, 2011). In addition, FPP-derived farnesol also contributes
ulation for controlled gene expression. The recombinant expression by to the alteration of the Hmg2p conformation into a less folded state,
plasmid is maintained at gene dosage levels similar to that of the thereby increasing the susceptibility of Hmg2p to degradation (Kuranda,
chromosome while avoiding the interruption of native genes (Jones, François, & Palamarczyk, 2009). To overcome the limitation, the N-
Kim, & Keasling, 2000). Additionally, the plasmid with multiple copy terminal truncated and soluble form of Hmg1p (tHmg1p) which is free
numbers can increase the expression of target proteins (Chen, Partow, from feedback regulation, is available to increase the accumulation of
Scalcinati, Siewers, & Nielsen, 2012). Therefore, the optimization of intermediates and thus terpene precursors, thereby boosting the terpene
plasmid is beneficial to the efficiency of terpene synthesis in engineered yield (Farhi et al., 2011; Wang, Quan, et al., 2019). For monoterpene,
microorganism. So far, overexpression of HMGR variant, ERG20 over­ limonene can be produced in S. cerevisiae by expressing tHMG1 and
expression, ERG9 downregulation, integration of extra IDI1 gene, dele­ Citrus limon limonene synthase at the yield level of 1.48 mg/L (Beh­
tion of ROX1 and construction of orthogonal pathway have been rendorff, Vickers, Chrysanthopoulos, & Nielsen, 2013). For sesquiter­
adopted as the effective engineering strategies for enhancing terpene pene, in the study of Farhi et al. (2011), the overexpression of tHMG1,
production in S. cerevisiae (Fig. 2). Here, we summarize terpene products mitochondrion-targeted heterologous FPPS (mtFDPS) and a
including their yield from different S. cerevisiae strains engineered by mitochondrion-targeted sesquiterpene synthase (mtCsTPS) can produce
these strategies (Table 1). 1.5 and 20 mg/L of valencene and amorphadiene respectively, which
accounted for 8- and 20-fold enhancements in the production. When the

Fig. 2. Engineering strategies for enhancing terpene production in Saccharomyces cerevisiae. Dashed arrow lines represent multiple steps. HMG-CoA, 3-hydroxy-3-
methylglutaryl-CoA; MVA, mevalonate; IPP, isopentenyl pyrophosphate; DMAPP, dimethylallyl pyrophosphate; GPP, geranyl pyrophosphate; FPP, farnesyl
diphosphate; GGPP, geranylgeranyl pyrophosphate; NPP, neryl diphosphate; AACT, acetoacetyl-CoA thiolase; HMGS, HMG-CoA synthase; HMGR, HMG-CoA
reductase; MVK, mevalonate kinase; PMK, mevalonate-5-phosphate (MVAP) kinase; MDD, mevalonate-5-pyrophosphate (MVAPP) decarboxylase; IDI, isopentenyl
diphosphate isomerase; SS, squalene synthase; FPPS, FPP synthase; GGPPS, GGPP synthase; NPPS, NPP synthase; MTS, monoterpene synthase; STS, sesquiterpene
synthase; DTS, diterpene synthase.

4
Z. Liang et al. Food Research International 147 (2021) 110487

Table 1
Selective studies of terpenes/terpenoids from engineered Saccharomyces cerevisiae strains.
Terpene/ Maximum Strain Engineering strategy Terpene synthase Reference
Terpenoid yield (mg/L)

Monoterpene
(+)-Limonene 1.48 EPY210C Overexpression of tHMG1 Limonene synthases from Citrus (Behrendorff
limon and Mentha spicata et al., 2013)
0.028 AE9 K197G Expression of an ERG20 mutant (K197G) (+)-Limonene synthase from (Jongedijk et al.,
Citrus limon 2015)
166 AM94 a. Construction of orthogonal pathway using substrate NPP for NPP-specific variant limonene (Ignea et al.,
monoterpene synthase synthase from Citrus limon 2019)
b. Introduction of ergosterol-repressed ERG1 promoter upstream of (ClLimS)
the ERG20 gene
917.7 yJGZ1 a. Construction of orthogonal pathway using substrate NPP for N-terminal truncated limonene (Cheng et al.,
monoterpene synthase of terpenes/terpenoids from engineered b. synthase from Citrus limon 2019)
Dynamic downregulation of ERG20 by replacing the ERG20 promoter
with the glucose-sensing HXT1 promoter
9.8 AY12α a. Construction of orthogonal pathway using substrate NPP for N-terminal truncated limonene (Hu et al., 2020)
monoterpene synthase of terpenes/terpenoids from engineered b. synthase from Citrus limon
Expression of N-terminal truncated NPPS encoded by tNDPS1
c. Expression of tLS–tNDPS1 fusion enzyme
(-)-Limonene 0.060 AE9 K197G Expression of an ERG20 mutant (K197G) (-)-Limonene synthase from (Jongedijk et al.,
Perilla frutescens 2015)
Sabinene 17.5 AM94 a. Expression of a degradation-stabilized variant of HMG2 (bearing a Sabinene synthase from Salvia (Ignea et al.,
K6 to R mutation) pomifera 2014)
b. Expression of ERG20 mutants (A99C, A99L, A99F, A99W, F96W,
N127W, F96W-N127W)
c. Monoallelic deletion of ERG9
113 AM94 a. Construction of orthogonal pathway using substrate NPP for NPP-specific variant sabinene (Ignea et al.,
monoterpene synthase synthase from Salvia pomifera 2019)
b. Introduction of ergosterol-repressed ERG1 promoter upstream of (SpSabS)
the ERG20 gene
Cineole 485 BY4741 a. Integration of a K6R variant of HMG2 into the genome Cineole synthase from Salvia (Ignea et al.,
b. Integration of a promoter-IDI1-ts cassette fruticosa (CinS1) 2011)

Monoterpenoid
Geraniol 5 AE9 K197K Expression of ERG20 mutants (K197G, E, S) Geraniol synthase from Ocimum (Fischer et al.,
basilicum 2011)
1690 YZG13-GE1 a. Dynamic control of ERG20 by replacing the ERG20 promoter with Geraniol synthase from (Zhao et al.,
the glucose-sensitive HXT1 promoter Valeriana officinalis 2017)
b. Optimization of carbon source feeding strategies (glucose and
ethanol ratio)
c. Deletion of OYE2 or ATF1
d. LEU2 auxotrophic complementation
6.77 CEN.PK2-1C a. Overexpression of tHMG1 N/A (Liu, Zhang,
b. Integration of IDI1 gene et al., 2013)
Linalool 0.3 AE9 K197K Expression of ERG20 mutants (K197G, E, S) N/A (Fischer et al.,
2011)
Citronellol 0.3 AE9 K197K Expression of ERG20 mutants (K197G, E, S) N/A (Fischer et al.,
2011)

Sesquiterpene
Farnesene 13.8 CEN.PK113-5D a. Expression of acetoacetyl-CoA synthase (nphT7) Farnesene synthase from Citrus (Tippmann
b. Overexpression of tHMG1 junos (FarnSyn) et al., 2017)
c. Overexpression of mutated acetyl-CoA carboxylase (ACC1**)
2240 CEN.PK2-1C, CEN.PK2- a. Substituting ALD6, ACS1 and ACS2 (‘PDH-bypass’) reactions with N/A (Meadows et al.,
1D or CEN.PK113-7D acetaldehyde dehydrogenase (acylating) (ADA) 2016)
b. Replacing the native NADPH-dependent HMGR with an NADH-
specific version (NADH-HMGR) from Silicibacter pomeroyi
c. Overexpression of xylulose-5-phosphate specific phosphoketolase
(xPK) and phosphotransacetylase (PTA)
Bisabolene 994 EPY300 a. Overexpression of UPC2-1, tHMG1, ERG20 Abies grandis α-bisabolene (Peralta-Yahya
b. Downregulation of ERG9 synthase (AgBIS) et al., 2011)
400 BY4741, CEN.PK2 a. Deletion of ROX1 Abies grandis α-bisabolene (Özaydın et al.,
synthase (AgBIS) 2013)
800 BY4741, CEN.PK2 a. Deletion of ROX1 Abies grandis α-bisabolene (Özaydın et al.,
b. Deletion of YJL064W and YPL062W synthase (AgBIS) 2013)
Caryophyllene 125 BY4741 a. Tandem heterologous deletion of ubiquitin ligase Ubc7p, Ssm4p Caryophyllene synthase from (Ignea et al.,
and the endoplasmic reticulum resident protein Pho86p Salvia fructicosa 2012)
b. Overexpression of a degradation stabilized variant of Hmg2p (K6R)
δ-Cadinene 40 BY4741 a. Overexpression of mutated HMG2 (K6R) and ERG20 Sesquiterpene synthase (P330) (Ignea et al.,
b. Downregulation of ERG9 from Salvia pomifera 2011)
trans- 10 BY4741 a. Integration of a K6R variant of HMG2 into the genome Sesquiterpene synthase (P330) (Ignea et al.,
β-Caryophyllene b. Upregulation of ERG20 from Salvia pomifera 2011)
c. Haploinsufficiency for ERG9
Valencene 3.0 CEN.PK113-5D Downregulation of ERG9 Valencene synthase (GFTpsD) (Asadollahi
from grapefruit et al., 2008)
1.5 BDXe
(continued on next page)

5
Z. Liang et al. Food Research International 147 (2021) 110487

Table 1 (continued )
Terpene/ Maximum Strain Engineering strategy Terpene synthase Reference
Terpenoid yield (mg/L)

a. Overexpression of tHMG1 Valencene synthase from Citrus (Farhi et al.,


b. Expression of mtFDPS sinensi (CsTPS1) 2011)
c. Expression of mtCsTPS
539.3 BJ5464 a. Deletion of ROX1 Valencene synthase from (Chen et al.,
b. Overexpression of ERG10, ERG13, tHMG1, ERG12, ERG8, ERG19, Callitropsis nootkatensis 2019)
ERG20 and IDI1
c. Downregulation of ERG9
d. Deletion of BTS1, DPP1, LPP1
Amorphadiene 20 BDXe a. Overexpression of tHMG1 Amorpha-4,11-diene synthase (Farhi et al.,
b. Expression of mtFDPS from Artemisia annua 2011)
c. Expression of mtCsTPS
α-Copaene 12 BY4741 a. Integration of a K6R variant of HMG2 into the genome Sesquiterpene synthase (P330) (Ignea et al.,
b. Upregulation of ERG20 from Salvia pomifera 2011)
c. Haploinsufficiency for ERG9
α-Cubebene 7 BY4741 a. Integration of a K6R variant of HMG2 into the genome Sesquiterpene synthase (P330) (Ignea et al.,
b. Upregulation of ERG20 from Salvia pomifera 2011)
c. Haploinsufficiency for ERG9

Sesquiterpenoid
trans-Nerolidol 64.4 BEJY12 a. Delection of ERG9 N/A (Jackson, 2005)
b. Overexpression of tHMG1
Farnesol 20.71 BEJY12 a. Delection of ERG9 N/A (Jackson, 2005)
b. Overexpression of tHMG1
Cubebol 1.6 CEN.PK113-5D Downregulation of ERG9 Cubebol synthase (GFTpsC) (Asadollahi
from grapefruit et al., 2008)
9.9 CEN.PK113-5D a. Overexpression of tHMG1 Cubebol synthase from Citrus (Asadollahi
b. Downregulation of ERG9 paradisi et al., 2010)
Patchoulol 16.9 CEN.PK113-5D Downregulation of ERG9 Patchoulol synthase (Asadollahi
(PatTps177) from patchouli et al., 2008)

Diterpene &
Diterpenoid
Casbene 108.5 NCYC 3608 a. Expression of a truncated GGPPS from Phomopsis amygdali Casbene synthase from Ricinus (Callari et al.,
b. Dynamic control of ERG20 and ERG9 by replacing their native communis 2018)
promoters with glucose-sensitive HXT1 promoter and ergosterol-
sensitive ERG1 promoter

Steviol glycosides N/A Haploid strain (MATα a. Optimization of the kaurene oxidase-kaurenoic acid hydroxylase Kaurene synthase (Gold et al.,
HOΔura3Δhis3-Δleu2Δ) (KO-KAH) activities 2018)
b. Expression of cytochrome P450 reductase (CPR)
c. Expression of UDP (uridine diphosphate) glucose-dependent
glucosyltransferases (UGTs)
Sclareol 750 BY4741 a. Deletion of ROX1 Sclareol synthase from Salvia (Trikka et al.,
b. Deletion of YGR259C, YNR063W, VBA5, YER134C and DOS2 sclarea 2015)

Triterpene &
Triterpenoid
β-Amyrin 36.5 INVSc1 a. Integration of IDI1 gene from E. coli β-Amyrin synthase from (Zhang et al.,
b. Overexpression of ERG20 and ERG9 from S. cerevisiae Glycyrrhiza glabra 2015)
Protopanaxadiol 1189 BY4742 a. Overexpression of tHMG1, ERG20, ERG9 Protopanaxadiol synthase from (Dai et al., 2013)
b. Overexpression of squalene epoxidase (ERG1) Panax ginseng
c. Increasing protopanaxadiol synthase activity through codon
optimization

Tetraterpene
β-Carotene 47.18 BY4741, BY4742 a. Introduction of the positive GGPP synthase mutant (crtE03M) N/A (Zhou et al.,
b. Overexpression of tHMG1, crtI and crtYB 2017)
c. Increasing β-carotene ketolase activity via directed evolution
d. Balancing the metabolic flux by adjusting copy numbers of the rate-
limiting enzymes

Norisoprenoid
β-Ionone 5 SCIGS22 a. Integration of the tHMG1 gene N/A (López et al.,
b. Overexpression of ERG20 2015)
c. Downregulation of ERG9
d. Deletion of LPP1 and DPP1 genes
e. Overexpression of BTS1, crtYB and crtI genes
f. Expression of CCD1 gene

N/A, not available; S. cerevisiae, Saccharomyces cerevisiae; E. coli, Escherichia coli; NPP, neryl diphosphate; NPPS, NPP synthase; ADA, acetaldehyde dehydrogenase
(acylating); NADPH, nicotinamide adenine dinucleotide phosphate; NADH, nicotinamide adenine dinucleotide; HMGR, HMG-CoA (3-hydroxy-3-methylglutaryl-CoA)
reductase; xPK, xylulose-5-phosphate specific phosphoketolase; PTA, phosphotransacetylase; GGPP, geranylgeranyl diphosphate; GGPPS, GGPP synthase; KO-KAH,
kaurene oxidase-kaurenoic acid hydroxylase; CPR, cytochrome P450 reductase; UDP, uridine diphosphate; UGTs, UDP glucose-dependent glucosyltransferases.

6
Z. Liang et al. Food Research International 147 (2021) 110487

tHMG1 engineering combined with the expression of acetoacetyl-CoA of limonene by expressing (+)-limonene synthase from C. limon, and
synthase (nphT7), mutated acetyl-CoA carboxylase (ACC1**) and far­ (-)-limonene synthase from Perilla frutescens (Jongedijk et al., 2015). In
nesene synthase from Citrus junos (FarnSyn), the final farnesene yield can addition, a double mutation of F96 to W and N127 to W was also
reach 13.8 ± 4.1 mg/L (Tippmann, Ferreira, Siewers, Nielsen, & Chen, introduced into the ERG20 gene to enhance the production of sabinene
2017). During the biosynthesis, excess nicotinamide adenine dinucleo­ by up to 10.4-fold to 0.53 mg/L in diploid S. cerevisiae strain. The fusion
tide (NADH) is readily oxidized by O2, leading to the inefficient capture of the mutated Erg20p (F96W-N127W) with sabinene synthase from
of free energy from glucose in farnesene (Dugar & Stephanopoulos, Salvia pomifera (SpSabS1) can boost sabinene yield by 3.5-fold to 1.87
2011). Therefore, another study replaced the native nicotinamide mg/L. A 7-fold increase in production can be achieved when the fusion
adenine dinucleotide phosphate (NADPH)-specific HMGR with an occurred in the erg20/ERG20 heterozygous deletion strain, while the
NADH-dependent one in the strain, together with the substitution of introduction of a second plasmid vector generating the same fusion can
acetaldehyde dehydrogenase (ALD6) and acetyl-CoA synthases (ACS1 further boost the production to 17.5 mg/L (Ignea, Pontini, Maffei,
and ACS2) (‘PDH-bypass’) reactions with acetaldehyde dehydrogenase Makris, & Kampranis, 2014). Ignea et al. (2011) integrated galactose
(acylating) (ADA) and overexpression of xylulose-5-phosphate specific promoter in one of the ERG20 alleles in diploid S. cerevisiae strain,
phosphoketolase (xPK) and phosphotransacetylase (PTA), which suc­ leading to 25%, 30% and 50% increases in δ-cadinene, cineole and trans-
cessfully boosted the farnesene production to 2240 mg/L (Meadows β-caryophyllene production respectively, whereas the contents of
et al., 2016). It is worth mentioning that cubebol production can be α-copaene and α-cubebene grew slightly. Another study reported the
increased by 30% when overexpressing tHMG1 from an episomal high dynamic control of ERG20 by replacing the ERG20 promoter with the
copy plasmid by the integration of strong inducible GAL10 promoter glucose-sensing HXT1 promoter, which can successfully redistribute
rather than genomic integration (Asadollahi, Maury, Schalk, Clark, & GPP flux and thus attain a 3.8-fold increase to 867.7 mg/L in geraniol
Nielsen, 2010). In addition, the overexpression of tHMG1 can also aid in production after optimizing carbon source feeding. The geraniol yield
tetraterpene biosynthesis. Zhou et al. (2017) overexpressed tHMG1, can be further elevated to 1.69 g/L by involving OYE2 (encoding an
phytoene desaturase (crtI), phytoene synthase (crtYB) and GGPP syn­ NADPH oxidoreductase) deletion and LEU2 (auxotrophic marker)
thase (GGPPS) mutant (crtE03M) in an engineered diploid strain of complementation in addition to ERG20 dynamic control (Zhao et al.,
S. cerevisiae with increased β-carotene ketolase activity and balanced 2017). It is well worth mentioning that an auxin-inducible degradation
metabolic flux, resulting in a 1.8-fold increase to 47.18 mg/L in astax­ of Erg20p can successfully redirect metabolic flux towards monoterpene
anthin production. Engineering of Hmg2p also aims for improved production, which is an advanced metabolic engineering tool
terpene production while inhibiting protein degradation. For instance, a demanding further study to determine its feasibility (Lu, Peng, Ebert,
single point mutation replacing lysine 6 with an arginine (K6R) in HMG2 Dumsday, & Vickers, 2021).
led to nearly three times higher production of cineole, α-copaene and
α-cubebene in S. cerevisiae while trans-β-caryophyllene and δ-cadinene 3.3. Downregulation of ERG9
also increased by two-fold (Ignea et al., 2011). The stability of Hmg1p
and Hmg2p are negatively affected by their positive interactors, Another possible strategy to increase the availability of terpene
including ubiquitin ligase Ubc7p, Ssm4p and the endoplasmic reticulum precursors is to lower their flux toward sterol synthesis (Paramasivan &
resident protein Pho86p. These proteins participate in the degradation Mutturi, 2017). Sterol is a cell membrane component derived from
of endoplasmic reticulum transmembrane proteins, and the deletion of squalene, being responsible for maintaining the permeability, fluidity,
them contributes to the improved stabilization of Hmg1p and Hmg2p as well as the structural integrity of cell membranes and presenting as
(Paramasivan & Mutturi, 2017). For instance, the caryophyllene yield signal compounds when cells communicate with one another (Zhuang,
using S. cerevisiae expressing caryophyllene synthase from Salvia fructi­ 2013). ERG9, which encodes squalene synthase (Erg9p), is targeted for
cosa can be boosted to 125 mg/L by deleting the target genes of all three further modification of the MVA pathway in addition to ERG20. Erg9p
proteins and overexpressing the stabilized variant of Hmg2p, which is functions in the transformation of FPP into squalene while acting
11-fold higher than that of the wild strain (Ignea et al., 2012). downstream of Erg20p (Ignea et al., 2011). The downregulation of ERG9
can be attained via the deletion of the gene. In the study of Chen et al.
3.2. Upregulation of ERG20 (2019), deleting 45-bp in the range of upstream activating cis-element
(UAS) in ERG9 promoter can promote the production of valencene in the
Upregulation of the ERG20 gene is a specific strategy to stimulate S. cerevisiae strain with valencene synthase from Callitropsis nootkatensis.
monoterpene biosynthesis in S. cerevisiae by increasing the intracellular Monoallelic deletion of ERG9 together with the expression of HMG2
GPP pool in the MVA pathway (Liu, Zhang, Du, Chen, & Zhou, 2013). (bearing a K6 to R mutation) can yield 0.05 mg/L sabinene (Ignea et al.,
GPP, formed by the condensation of IPP and DMAPP, is known as the 2014). Further, ERG9 downregulation can also be coupled with the
principal precursor for monoterpene biosynthesis (Wang, Quan, et al., overexpression of tHMG1, ERG20, global transcription regulator of ste­
2019). Unlike plants, yeast does not harbor a specific GPP synthase rol pathway UPC2-1, as well as Abies grandis α-bisabolene synthase
(GPPS), resulting in no or limited monoterpene production (Zhuang, (AgBIS), reaching 994 ± 241 mg/L in bisabolene production (Peralta-
2013). In S. cerevisiae, Erg20p, the dual-specificity enzyme encoded by Yahya et al., 2011). The combination of ERG9 deletion and tHMG1
ERG20, has both GPPS and FPPS activities, catalyzing the sequential overexpression was also reported to produce 20.71 mg/L farnesol and
formation of GPP and thus FPP. However, GPP only functions as an in­ 64.40 mg/L trans-nerolidol which were not identified in the wild type
termediate of FPP synthesis since it is tightly bound to the catalytic site yeast strains (Jackson, 2005). Nevertheless, deletion of ERG9 gene can
of Erg20p and oriented to the conversion to FPP (Zhao, Bao, Li, Shen, & compromise cellular activity in haploid cells, while no detrimental effect
Hou, 2016). To increase the GPP flux in S. cerevisiae, a series of ERG20 but decreased mRNA levels of the gene is reported when deleting one
mutants are constructed for the endogenous shift from FPP to GPP allele in diploid cells. In the study of Ignea et al. (2011), a loxP-his5-loxP
accumulation (Zhuang, 2013). In the study of Fischer, Meyer, Claudel, cassette was used to delete one of the two ERG9 alleles in the S. cerevisiae
Bergdoll, and Karst (2011), S. cerevisiae ERG20 gene was subjected to a strain, resulting in a significant decline in monoterpene production (e.g.
sequence of amino acid mutations at position K197, where K197 G, E cineole) but a drastic increase in sesquiterpene production (e.g. trans-
and S successfully boosted the production of geraniol, linalool and β-caryophyllene, α-copaene, δ-cadinene). This may be ascribed to FPP
citronellol by 10- to 20-fold. It is worth mentioning that the sterol accumulation, which might lead to feedback inhibition on Erg20p with
content, growth rate, and monoterpenol production abilities varied in GPPS activity and thus reduce the GPP availability (Ignea et al., 2015).
different mutated strains. Similarly, an ERG20 mutant strain (AE9 During aerobic growth conditions, yeast cells can not actively assimilate
K197G) was also deployed to simultaneously generate both enantiomers exogeneous ergosterol that is vital for yeast growth, so the complete

7
Z. Liang et al. Food Research International 147 (2021) 110487

deletion of ERG9 gene responsible for endogenous ergosterol cannot be mentioning that the expression of IDI1 may exert no enhancing effect on
attained (Asadollahi et al., 2010). To complement the gene deletion, sesquiterpene yield, but contribute to the expansion of squalene pool for
Zhang, Jennings, Robinson, and Poulter (1993) adopted a SUE (sterol triterpene overproduction. For instance, the co-expression of IDI1,
uptake enhancement) mutation which is a yeast line that enables the ERG20 and ERG9 in S. cerevisiae with β-amyrin synthase from Glycyr­
aerobic uptake and utilization of exogenous sterol, while the endoge­ rhiza glabra can enhance the accumulation of squalene and thus
nous ergosterol biosynthetic pathway becoming dispensable. In β-amyrin, resulting in 30% and 46% increases in β-amyrin production in
response, a large quantity of pathway intermediates (DMAPP, IPP and plasmid-based and genome-integrated yeast strains respectively (Zhang
FPP) are diverted to the biosynthesis of other non-essential terpene et al., 2015).
products.
Replacement of the ERG9 promoter with a repressive one also serves 3.5. Deletion of ROX1
as a promising alternative to ERG9 deletion. Asadollahi et al. (2008)
replaced the endogenous ERG9 promoter with the methionine repressive ROX1 is a transcriptional factor inhibiting the expression of hypoxia-
promoter MET3 in the S. cerevisiae cells, resulting in a decreased induced genes in MVA pathway and ergosterol biosynthesis (Henry,
ergosterol content and increased levels of FPP derived compounds like Nickels, & Edlind, 2002; Montañés, Pascual-Ahuir, & Proft, 2011),
sesquiterpenes. The yield of valencene and patchoulol separately which can be knocked out to enhance the mevalonate levels in cells and
increased by 50% and 70%, while the cubebol content remained nearly thus terpene production. In the study of Jakočiūnas et al. (2015), single
unchanged. The addition of higher amounts of methionine to the me­ deletion of ROX1 in S. cerevisiae can significantly boost valencene yield
dium can further increase the patchoulol content by 47% to 16.9 mg/L. compared to other single gene alterations of ERG9, BTS1, YPL062W and
Another study reported the same replacement strategy, which increased YJL064W. Under ROX1 deletion, the valencene production volume
the cubebol production more than threefold from 3.2 mg/L to 9.9 mg/L. reached 539.3 mg/L which was a 160-fold increase when additional
However, the simultaneous overexpression of tHMG1 in this background engineering strategies (overexpression of ERG10, ERG13, tHMG1,
decreased cubebol production by 10% (Asadollahi et al., 2010). Glucose- ERG12, ERG8, ERG19, ERG20 and IDI1, downregulation of ERG9,
regulated promoters such as HXT1 promoter can also be used to replace deletion of BTS1, DPP1, LPP1) were simultaneously employed in the
the ERG9 promoter. Under this condition, integration of the tHMG1 S. cerevisiae strain with valencene synthase from C. nootkatensis (Chen
gene, overexpression of ERG20, GGPPS gene (BTS1), carotenogenic et al., 2019). Similarly, a two-fold increase in bisabolene production was
genes (crtYB and crtI) and carotenoid cleavage dioxygenase gene reported in S. cerevisiae by expressing bisabolene synthase from
(CCD1), deletion of lipid phosphate phosphatase gene (LPP1) and A. grandis and deleting ROX1 gene. In addition, the deletion of YJL064W
diacylglycerol pyrophosphate phosphatase gene (DPP1) increased the and YPL062W genes can further increase the flux through the MVA
β-ionone production by 23-fold to 5 mg/L in the engineered S. cerevisiae pathway, thereby boosting the bisabolene yield from 400 mg/L to 800
strain (López et al., 2015). Another study replaced the ERG9 and ERG20 mg/L (Özaydın, Burd, Lee, & Keasling, 2013). According to Trikka et al.
promoters with ergosterol-sensitive ERG1 promoter and glucose- (2015), the combination of ROX1 deletion and the deletion of other five
sensitive HXT1 promoter respectively, together with the expression of genes (YGR259C, YNR063W, VBA5, YER134C and DOS2) which limit
truncated GGPPS from Phomopsis amygdali and casbene synthase from the productivity of carotenoid and potentially diterpene, induced a 40-
Ricinus communis, which successfully boosted the casbene production by fold increase in carotenoid, and consequently a 12-fold increase in
3.6-fold to 108.5 mg/L (Callari, Meier, Ravasio, & Heider, 2018). It is sclareol yield to 750 mg/L in S. cerevisiae with sclareol synthase from
reported that ERG9 can also be overexpressed to enrich the squalene Salvia sclarea. It is worth mentioning that ROX1 deletion in S. cerevisiae
pool for triterpene biosynthesis. In the study of Dai et al. (2013), a 262- can facilitate the expression of multiple ERG genes participating in
fold increase in protopanaxadiol production can be observed in ergosterol biosynthesis, leading to 2.5- to 16-fold-lower susceptibilities
S. cerevisiae with enhanced protopanaxadiol synthase from Panax to two sterol biosynthesis inhibitors, azoles and terbinafine (Henry et al.,
ginseng, when combining the overexpression of ERG9, tHMG1, ERG20 2002).
and squalene epoxidase (ERG1).
3.6. Recent advances on engineering strategies
3.4. Integration of extra IDI1 gene
Construction of orthogonal pathway is an emerging strategy to
The gene IDI1 catalyzes the isomerization of IPP to DMAPP by enhance monoterpene production in yeast by using neryl diphosphate
encoding for an essential isomerase IDI, contributing to the subsequent (NPP) as substrate for MTSs (Ignea et al., 2019). NPP is formed by the
generation of GPP and monoterpene. Therefore, the introduction of IDI1 condensation of IPP and DMAPP in the cis-configuration under the
gene into the yeast genome is well accepted as a promising strategy to catalysis of NPP synthase (NPPS) encoded by SlNDPS1 gene (Schilmiller
improve the monoterpene production which depends on the copy et al., 2009). Unlike GPP, NPP is not consumed by Erg20p for FPP
number of IDI1 (Ignea et al., 2011). According to Liu, Zhang, et al. conversion. MTSs can be engineered to specifically accept NPP as the
(2013), the integration of an extra copy of IDI1 into the genome of substrate for yielding different monoterpene products. The orthogonal
S. cerevisiae can lead to a 50% increase in geraniol yield, while the IDI1 route branches out from the MVA pathway at the point following the
overexpression using a multi-copy plasmid boosted geraniol production synthesis of IPP and DMAPP. The branch point functions as a metabolic
by 1.45-fold. A 6-fold increase can also be observed when combining the valve which can be controlled dynamically to regulate fluxes between
overexpression of IDI1 and tHMG1. In the study of Ignea et al. (2011), the canonical and orthogonal branches (Ignea et al., 2019). For instance,
the high copy number plasmid pYES2-myc was used to integrate the IDI1 Ignea et al. (2019) successfully installed dynamic control by introducing
gene under the control of the galactose promoter in the genome of ergosterol-repressed ERG1 promoter upstream of the ERG20 gene in
S. cerevisiae with cineole synthase from Salvia fruticosa. For the modified S. cerevisiae with NPP-specific variants S. pomifera sabinene synthase
strain, the overexpression of IDI1 boosted the cineole production to 250 (SpSabS) and C. limon limonene synthase (ClLimS), which directed
mg/L which was 5-fold higher than that of the unmodified one. Under fluxes to the orthogonal pathway due to the increased sterol levels
the HMG2 expression (K6R), the same study also cloned the IDI1 gene (Leber et al., 2001), and thus led to 7.1- and 6.8-fold increases in sabi­
into the COD4 cassette to obtain the COD40 construct which was then nene and limonene production respectively. Similarly, Cheng et al.
integrated into the yeast chromosome, producing cineole at the yield (2019) established an orthogonal biosynthetic pathway involving NPPS
level of 200 mg/L. A further increase to 485 mg/L can be achieved when from Solanum lycopersicum and N-terminal truncated C. limon limonene
a plasmid expressing IDI1 was introduced, confirming that the integra­ synthase. Under this condition, dynamic downregulation of the ERG20
tion of additional copies can significantly promote yields. It is worth gene by replacing the ERG20 promoter with the glucose-sensing HXT1

8
Z. Liang et al. Food Research International 147 (2021) 110487

promoter can boost the limonene yield by 6-fold to 917.7 mg/L in the STSs through investigating plant STSs (Zhang et al., 2020). In compar­
engineered S. cerevisiae strain. In the study of Hu et al. (2020), the N- ison with plants, fungi possess a higher TPSs diversity which out­
terminal truncated genes of (+)-limonene synthase (tLS) and NPPS numbers by at least 10 times (Blackwell, 2011). For instance, each
(tNDPS1) were codon-optimized and heterologously expressed to fungus in the phylum Basidiomycota was reported with an average of
reconstruct an orthogonal pathway in S. cerevisiae, resulting in a 17-fold 10–20 putative TPS homologues (Wawrzyn, Quin, Choudhary, López-
increase to 6.97 mg/L in (+)-limonene production. The yield can be Gallego, & Schmidt-Dannert, 2012). Therefore, the fungal TPSs and
further enhanced by 40% to 9.8 mg/L by adopting a tLS–tNDPS1 fusion their terpene products are abundant natural resources.
enzyme. Various fungal TPS genes have been identified through the heterol­
ogous expression in E. coli, but many of them remain largely untapped
3.7. Terpene product toxicity (Table 2). From Agrocybe aegerita, Zhang et al. (2020) identified 11 STSs
that were cloned and characterized in the engineered terpene-
Indeed, the engineering strategies can significantly promote mono­ overproducing E. coli chassis strain. Nine of the STSs produced at least
terpene synthesis in the MVA and orthogonal pathways in S. cerevisiae, one sesquiterpene except for Agr10 and Agr11, indicating that they were
yet the increased amounts of intermediates and monoterpenes are toxic not functional probably due to their incomplete metal-binding regions
and hazardous to the strain (Moser & Pichler, 2019). Monoterpenes can (NSE motif). δ-Cadinene and β-myrcene were generated by expressing
readily compromise the cell membrane integrity and function, as well as Agr4, which is a bifunctional enzyme responsible for the biosynthesis of
intracellular energy metabolism (Uribe, Ramirez, & Peña, 1985). both sesquiterpenes and monoterpenes. The expression of Agr1 also
Therefore, irreversible cell damage can be induced by high monoterpene synthesized δ-cadinene, which accounted for 60% of the total terpenes
levels. For instance, complete growth inhibition and substantial viability produced. Four major sesquiterpenes in similar proportions were pro­
loss were detected in both mutant and wild type S. cerevisiae strains in duced by Agr3-expressing E. coli, including α-muurolene (32%),
the presence of 60 mg/L limonene (Brennan, Turner, Krömer, & Nielsen, γ-muurolene (22%), δ-cadinol (21%) and δ-cadinene (24%). Agr3 is
2012). To improve S. cerevisiae tolerance toward diverse monoterpenes, closely related to CpSTS3 from Clitopilus pseudopinsitus and Cop3 from
heterologous expression of efflux pumps like ATP-binding cassette Coprinopsis Cinerea, both of which mainly produce α-muurolene. E. coli
(ABC) transporter can achieve expulsion and isolation of monoterpenes. expressing the Agr8 gene mainly produces γ-muurolene and β-cadinene.
For instance, GcABC-G1 is a specialized ABC transporter promoting the In other studies, the heterologous expression of the Copu3 and CpSTS3
survival rate of S. cerevisiae cells by 3, 7 and 30 times in the presence of genes from the Basidiomycota Coniophora puteana mainly yielded
(-)-β-pinene, (+)-limonene and (+)-3-carene, respectively. However, the cubebol and α-cadinol (Mischko, Hirte, Fuchs, Mehlmer, & Brück, 2018;
transformed yeast cells showed no increased tolerance to the racemic Nagamine et al., 2019), while the expression of ShSTS12 from Stereum
α-pinene (Wang et al., 2013). Monoterpenes are sparingly water-soluble hirsutum primarily produced α-cubebene and β-cubebene (Nagamine
oils that predominantly induce phase toxicity rather than molecular et al., 2019; Quin, Flynn, Wawrzyn, Choudhary, & Schmidt- Dannert,
toxicity. Therefore, the upregulation of ABC transporters can only 2013). Additionally, Nagamine et al. (2019) found that the E. coli
address low levels of molecular toxicity but no phase toxicity. For expression of CpSTS18, CpSTS5 and CpSTS11 from Clitopilus pseudo-
instance, ABC transporters were proved to fail in limonene toxicity pinsitus generated γ-cadinene, γ-murolene and alloaromadendrene,
alleviation since limonene interferes with cell wall integrity rather than respectively. α-Gurjunene, β-gurjunene and β-copaene are the major
membranes in S. cerevisiae (Brennan, Krömer, & Nielsen, 2013; Hu et al., products from the expression of PpSTS06, Galma_104215 and Copu2
2012). Truncation of tricalbin protein (tTcb3p1-989) was first reported as genes that respectively derive from Postia placenta, Galerina marginata
a successful engineering of phase tolerance in S. cerevisiae (Brennan and C. puteana (Ichinose & Kitaoka, 2018; Mischko et al., 2018; Zhang
et al., 2015). tTcb3p1-989 can improve tolerance toward limonene, et al., 2020). For the brown-rot basidiomycete P. placenta, myrcene and
β-pinene and myrcene by 9-, 11- and 8-fold. Under limonene stress, linalool are the main monoterpene products generated by the expression
tTcb3p1-989 served to maintain cell wall integrity and enhance cell wall of PpSTS25, while the expression of PpSTS06 mainly produced α-gur­
resilience. It also displayed negligible perturbations to strain growth junene (Ichinose & Kitaoka, 2018). From Daldinia eschscholzii and
since fewer genes involved in cell wall maintenance and biogenesis were Hypoxylon sp., Wu et al. (2016) separately identified EC12-PGS and
affected in the reconstructed strain. Interestingly, most monoterpenes CI4A-CPS genes by the heterologous expression in E. coli, producing the
have no inhibitory effect on strain growth at their aqueous saturation corresponding sesquiterpenes including α-guaiene, β-pinene and β-cha­
points that are well below their minimum inhibitory concentrations migrene. Similar researches reported that α-farnesene, nerolidol, and
(MIC) (Brennan et al., 2012). Therefore, the toxic level of monoterpene germacrene D were derived from C. pseudo-pinsitus, Hypoxylon sp., and
may be hard to achieve. In contrast to monoterpene, the toxic effect of S. hirsutum separately containing three STS genes encoding CpSTS14,
sesquiterpene product on yeast cells is negligible due to the low pro­ Hyp1 and ShSTS10 (Nagamine et al., 2019; Quin et al., 2013; Shaw
duction level and weak toxicity (Schempp, Drummond, Buchhaupt, & et al., 2015). It should be noted that the functions of fungi are different
Schrader, 2018). between ascomycetes and basidiomycetes, while basidiomycetes are
more dominant in the total number and variety of terpene products
4. Fungal terpene synthase genes as TPSs reservoir (Zhang et al., 2020).
Rational engineering of fungal TPSs is an effective strategy to
Fungi are well-known for their production of diverse terpenes. Spe­ enhance enzyme specificity and activity due to the plasticity of TPSs.
cifically, the identity and number of terpene products depend on the The TPSs plasticity refers to the capacity of TPSs to modify their func­
fungal TPSs active sites in which the substrate is activated by a metal- tions with a small number of amino acid substitutions (Yoshikuni, Fer­
binding motif to generate a carbocation intermediate and guided rin, & Keasling, 2006). Site-directed mutagenesis and contact mapping
through a series of cyclization reactions (Shaw et al., 2015). So far, mutagenesis are the recent methods to identify plasticity residues which
about 100 fungal TPSs had been functionally characterized and most of control the profiles of STSs that are then transformed into a variety of
them are sesquiterpene synthases (STSs). STSs from plants and fungi more specific synthases (Greenhagen, O’Maille, Noel, & Chappell, 2006;
conserve metal-binding motifs composed of an NSE/DTE and an Yoshikuni et al., 2006). For instance, site-directed mutagenesis can
asparagine-rich motif. The NSE motif is highly preserved in fungal STSs, successfully identify plasticity residues in trichodiene synthase from
while the asparagine-rich motif in fungal STSs [D(D/E/N)XX(D/E)] is Fusarium sporotrichioides and generate a different distribution of
different to some extent from that found in plants [DDXX(D/E)]. Outside sesquiterpene products including some newly identified sesquiterpenes
the motif, the TPSs sequences of plants and microorganisms have limited (Vedula, Jiang, Zakharian, Cane, & Christianson, 2008). The study of
similarity, indicating the difficulty to predict the functions of fungal Shaw et al. (2015) also showed that the mutation of one amino acid in

9
Z. Liang et al. Food Research International 147 (2021) 110487

Table 2
Selective studies of fungal terpene synthase (TPS) genes.
Terpene/Terpenoid Fungus TPS Plasmid/Vector Identification approach Reference

Monoterpene
β-Pinene Hypoxylon sp. CI4A-CPS pJBEI-3122, pBbE1a, pBbE2k Heterologous expression in E. coli (Wu et al., 2016)
Myrcene Postia placenta PpSTS25 pGYRG Heterologous expression in E. coli (Ichinose & Kitaoka, 2018)

Monoterpenoid
Linalool Postia placenta PpSTS25 pGYRG Heterologous expression in E. coli (Ichinose & Kitaoka, 2018)
1,8-Cineole Hypoxylon sp. Hyp3 pTrc99 Heterologous expression in E. coli (Shaw et al., 2015)

Sesquiterpene
α-Farnesene Clitopilus pseudo-pinsitus CpSTS14 pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Nagamine et al., 2019)
Germacrene D Stereum hirsutum ShSTS10 pUCBB, pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Quin et al., 2013)
(Nagamine et al., 2019)
α-Selinene Agrocybe aegerita Agr8 pET Heterologous expression in E. coli (Zhang et al., 2020)
β-Selinene Agrocybe aegerita Agr8 pET Heterologous expression in E. coli (Zhang et al., 2020)
δ-Cadinene Agrocybe aegerita Agr1/Agr4 pET Heterologous expression in E. coli (Zhang et al., 2020)
β-Cadinene Agrocybe aegerita Agr8 pET Heterologous expression in E. coli (Zhang et al., 2020)
γ-Cadinene Clitopilus pseudo-pinsitus CpSTS18 pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Nagamine et al., 2019)
γ-Muurolene Clitopilus pseudo-pinsitus CpSTS5 pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Nagamine et al., 2019)
α-Muurolene Agrocybe aegerita Agr3 pET Heterologous expression in E. coli (Zhang et al., 2020)
α-Guaiene Daldinia eschscholzii EC12-PGS pJBEI-3122, pBbE1a, pBbE2k Heterologous expression in E. coli (Wu et al., 2016)
β-Chamigrene Hypoxylon sp. CI4A-CPS pJBEI-3122, pBbE1a, pBbE2k Heterologous expression in E. coli (Wu et al., 2016)
Alloaromadendrene Clitopilus pseudo-pinsitus CpSTS11 pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Nagamine et al., 2019)
α-Gurjunene Postia placenta PpSTS06 pGYRG Heterologous expression in E. coli (Ichinose & Kitaoka, 2018)
β-Gurjunene Galerina marginata Galma_104215 pET Heterologous expression in E. coli (Zhang et al., 2020)
β-Copaene Coniophora puteana Copu2 pACYC Heterologous expression in E. coli (Mischko et al., 2018)
α-Cubebene Stereum hirsutum ShSTS12 pUCBB, pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Quin et al., 2013)
(Nagamine et al., 2019)
β-Cubebene Stereum hirsutum ShSTS12 pUCBB, pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Quin et al., 2013)
(Nagamine et al., 2019)

Sesquiterpenoid
Cubebol Coniophora puteana Copu3 pACYC Heterologous expression in E. coli (Mischko et al., 2018)
Nerolidol Hypoxylon sp. Hyp1 pTrc99 Heterologous expression in E. coli (Shaw et al., 2015)
α-Cadinol Clitopilus pseudo-pinsitus CpSTS3 pC9SC103, pDP1031, pDP1032 Heterologous expression in E. coli (Nagamine et al., 2019)

E. coli, Escherichia coli.

Hypoxylon sp. can lead to the conversation of Hyp3 from one 1,8-cineole long time required for the insertion of multiple genes. In response, a
synthase to one limonene, myrcene or farnesene synthase. Currently, the rapid reconstitution strategy was developed to introduce heterologous
heterologous genes for the functional expression of TPSs in S. cerevisiae aflatrem biosynthetic genes into A. oryzae for the biosynthesis of indole
are mainly derived from plant sources, or synthetically generated diterpenes aflatrem and β-aflatrem (Tagami et al., 2014). This strategy
(Paramasivan & Mutturi, 2017). Therefore, fungi might serve as a po­ requires two rounds of tandem transformation which use two plasmids
tential source of heterologous TPS genes with an enormous diversity, carrying the same selectable marker and promoter/terminator but
especially for sesquiterpenes. different target genes. Furthermore, it is possible to simultaneously
prepare several types of transformants with various genes in a single
5. Alternative microbial engineering platforms transformation, and introduce multiple genes with a single selectable
marker gene. The use of multiple plasmids with a small number of target
5.1. Filamentous fungi genes in a tandem transformation has contributed to the rapid and
simple reconstitution of fungal biosynthetic machinery. Fungal clus­
Similar to S. cerevisiae, filamentous fungi can function as a viable host tered regularly interspaced short palindromic repeat (CRISPR)/Cas9
for heterologous production of terpenes (Table 3), although relevant system is a recently developed genome editing technology for heterol­
research is still at the early stage. It should be noted that a filamentous ogous production of terpenes in A. oryzae. The CRISPR/Cas9 system
fungus of A. oryzae, which has been defined as a Generally Recognized as comprises only two components, the Cas9 nuclease of Streptococcus
Safe (GRAS) organism by US FDA (Food and Drug Administration, species and a single guide RNA (sgRNA). In the recipient genome, the
2018), has been adopted to reconstitute biosynthetic machinery, usually sgRNA can recognize the target sequence, which is subsequently cleaved
by introducing heterologous terpene biosynthetic genes using genome- by the Cas9 protein, creating a double-stranded break. When the break is
editing method, to yield various terpene products, including aphidico­ repaired by the cell via homologous recombination, gene integration can
lin (Fujii et al., 2011), pyripyropene (Itoh et al., 2010) and andrastin be introduced at the target locus (Katayama et al., 2016; Shi et al.,
(Matsuda, Awakawa, & Abe, 2013). 2019). In the study of Nagamine et al. (2019), A. oryzae can successfully
Generally, the terpene biosynthetic genes are amplified from the express 29 sesquiterpene synthase genes and diterpene biosynthetic
genomic DNA and cloned into the expression vectors to obtain expres­ genes from C. pseudo-pinsitus and S. hirsutum by introducing CRISPR/
sion plasmids before introducing into fungal host strains for over­ Cas9 plasmid and donor DNA plasmid with target genes. In addition,
expression (Nagamine et al., 2019). A. oryzae usually serves as a CRISPR/Cas9 system has a synergetic effect with forced recycling system
genetically tractable and efficient ascomycete host for basidiomycete which involves an autonomous maintenance in Aspergillus (AMA1)-
terpene gene expression via plasmid vectors. For instance, the heterol­ based replicating plasmid containing a drug resistance marker ptrA.
ogous expression of the seven pleuromutilin biosynthesis genes from Conditional expression of Aoace2 gene from AMA1-based plasmid is
Clitopilus passeckerianu (Pl-sdr, Pl-p450-2, Pl-p450-1, Pl-ggs, Pl-cyc, Pl-atf, detrimental to fungal growth, which allows for forced recycling of the
Pl-p450-3) in A. oryzae, can boost the production of diterpene product, plasmid and thus repeatable genetic engineering (Katayama et al.,
pleuromutilin by up to 20-fold compared to C. passeckerianu (Bailey 2019). For instance, Liu et al. (2019) reconstituted a Basidiomycota
et al., 2016). However, this bottom-up synthetic method is limited by a diterpene erinacine biosynthetic gene cluster in A. oryzae via a

10
Z. Liang et al. Food Research International 147 (2021) 110487

Table 3
Selective studies of terpenes/terpenoids from different engineered filamentous fungi.
Fungi Terpene/ Yield (mg/ Plasmid/Vector Biosynthetic gene Fold Reference
Terpenoid L) increase

Aspergillus oryzae Aphidicolin N/A pTAex3, pPTRI, pUSA, GGPP synthase (PbGGS), diterpene synthase (PbACS), N/A (Fujii et al.,
pAdeA two monooxygenases (PbP450-1 and PbP450-2) genes 2011)
Pyripyropene N/A pTAex3, pPTRI Polyketide synthase (PKS), prenyltransferase (PT), CoA N/A (Itoh et al.,
ligase, flavin adenine dinucleotide (FAD)-dependent 2010)
monooxygenase (FMO), cytochrome P450 (P450),
acetyltransferase (AT) genes
Andrastin N/A pTAex3, pPTRI, pAdeA, PKS, PT, FMO, methyltransferase (MT), terpene cyclase N/A (Matsuda
pUSA (AdrI) genes et al., 2013)
Aflatrem N/A pUARA2, pUSA2 GGPP synthase (PaxG), prenyltransferase (PaxC), N/A (Tagami
epoxidase (PaxM), cyclase (PaxB), cytochrome P450 et al., 2014)
(AtmP/Q), reverse-mode prenyltransferase (AtmD)
genes
Pleuromutilin 84.24 pYES2 Cytochrome P450, AT, terpene cyclase, GGPP 20 (Bailey et al.,
synthetase (GGS), P450 monooxygenase, short-chain 2016)
dehydrogenase/reductase (SDR) genes
N/A pC9SC103, pDP1031, Sesquiterpene synthase, glucose-methanol-choline N/A (Nagamine
pDP1032 (GMC) oxidase, oxidation/reduction enzyme, et al., 2019)
cytochrome P450, flavoprotein, GGPP synthase genes
Erinacine Erinacine pRGE-g801/g201, pC9pA- Cytochrome P450, short-chain dehydrogenase, N/A (Liu et al.,
Q: 4.7 g801, pC9pGg401/g601, polyprenyl synthase, UbiA prenyltransferase, enoyl- 2019)
Erinacine pDP201a, pDP401a, (acyl carrier protein) reductase, UDP-glucoronosyl and
Q2: 1.1 pDP601a UDP-glucosyl transferase, membrane bound O-acyl
transferase, GMC oxidase genes

Aspergillus nidulans Gamma- N/A pHHO5 Fusarium fujikuroi ent-kaurene synthase (cps/ks), N- N/A (Bromann
terpinene, ent- terminally truncated and hemagglutinin (HA)-tagged et al., 2016)
kaurene gamma-terpinene synthase from Citrus unshiu (gTerpS),
cluster-specific transcription factor (pbcR), ent-pimara-8
(14),15-diene synthase, GGPP synthase, cytochrome
P450 genes

Paraconiothyrium Taxol N/A pET32, pPK2, pBlueScript Taxus canadensis GGPP synthase gene 3 (Soliman
SSM001 spp. KS+ et al., 2017)
Alternaria alternata Taxadiene 0.0619 ± pGB123, pGB125, pGB127 IDI, tHMG1, taxadiene synthase (TS) genes N/A (Bian et al.,
0.0063 2017)

N/A, not available; GGPP, geranylgeranyl diphosphate; PKS, polyketide synthase; PT, prenyltransferase; FAD, flavin adenine dinucleotide; FMO, FAD-dependent
monooxygenase; AT, acetyltransferase; MT, methyltransferase; SDR, short-chain dehydrogenase/reductase; GMC, glucose-methanol-choline; UDP, uridine diphos­
phate; HA, hemagglutinin; TS, taxadiene synthase.

combination of CRISPR/Cas9 system and forced recycling system, pro­ pathway, which ensures FPP and/or GPP supply to activate GGPPS to
ducing erinacine Q and Q2 at the yield levels of 4.7 and 1.1 mg/L, improve its transcription and Taxol production (Soliman et al., 2017).
respectively. This combined strategy can eliminate the lengthy Bian et al. (2017) also developed an effective alternative to the semi-
screening step and use unlimited biosynthetic gene introduction because chemical synthesis of Taxol using endophytic fungus Alternaria alter­
of marker-free genome editing. nate TPF6. Specifically, a heterologous MVA pathway was transformed
Transgenic technology can also apply to other filamentous fungi to into A. alternata TPF6, then engineered by introducing the plant
improve terpene production. In the study of Bromann, Toivari, Viljanen, taxadiene-forming gene via ATMT method, resulting in the production
Ruohonen, and Nakari-Setälä (2016), Aspergillus nidulans was engi­ of 61.9 ± 6.3 μg/L taxadiene, which can be converted into Taxol by 19
neered to heterologously produce gamma-terpinene and ent-kaurene. enzymatic steps including hydroxylation and other oxygenation re­
Fusarium fujikuroi ent-kaurene synthase (cps/ks), and the N-terminally actions of the taxadiene skeleton (Hezari & Croteau, 1997). Up to date,
truncated and hemagglutinin (HA)-tagged gamma-terpinene synthase the potential of filamentous fungi as a terpene-producing platform has
from Citrus unshiu (gTerpS), were overexpressed in A. nidulans strain by not been fully exploited, particularly in the engineering and optimiza­
using a plasmid that incorporated the synthase genes under the control tion of endogenous MVA pathway.
of gpdA promoter. A plant GGPPS gene was also proved to be feasible for
integration into the Taxol-producing Paraconiothyrium SSM001 genome 5.2. Escherichia coli
via Agrobacterium tumefaciens-mediated transformation (ATMT) to in­
crease the GGPPS copy number, which enhanced the production level of With the advancement of metabolic engineering and transgenic
a diterpene anticancer drug, Taxol (paclitaxel) (Soliman, Mosa, El- technology, yeast is viewed as a convenient and efficient host for large-
Keblawy, & Husseiny, 2017). ATMT was carried out by delivering the scale terpene production (Paramasivan & Mutturi, 2017). However,
genetic components that are bordered by the left border (LB) and right some engineered yeasts are incapable of enhancing the yield of certain
border (RB) of its helper plasmid derived from three different terpene products such as cineole and cubebol (Asadollahi et al., 2008;
A. tumefaciens Ti plasmids (octopine, nopaline and L,L-succinamopine Asadollahi et al., 2010; Ignea et al., 2011). In response, E. coli serves as a
plasmids) (Hood, Gelvin, Melchers, & Hoekema, 1993). In addition, promising alternative host (Moser & Pichler, 2019). E. coli has a similar
feeding the GGPPS-transformed fungus with exogenous GGPP can genetic background to yeast, indicating its great potential in terpene
further improve the synthesis efficiency of both terpenes and Taxol. The synthesis using microbial-directed heterologous pathway (Pitera, Pad­
introduction of new copies of GGPPS requires more terpene precursors don, Newman, & Keasling, 2007).
such as GPP or FPP which are consumed by the original fungal GGPPS to The advantages of E. coli in terpene synthesis are mainly ascribed to
produce native GGPP for the synthesis of essential diterpenes. Therefore, two aspects. First, E. coli is a highly efficient host for terpene biosyn­
the addition of extra GGPP can be adopted by the original terpene thesis. E. coli can synthesize the basic structure units of terpenes via its

11
Z. Liang et al. Food Research International 147 (2021) 110487

inherent MEP pathway which surpasses the MVA pathway in stoichi­ the commercial production of terpenes, especially in precursor supply,
ometry and accumulates fewer byproducts (Dugar & Stephanopoulos, pathway optimization and microbial tolerance to terpene products. In
2011). Compared to S. cerevisiae, E. coli consumes less time when pro­ the future, more efforts should be invested toward identifying the bot­
ducing the same terpene due to its faster growth rate (Ingy & Wim, tlenecks of MVA or MEP pathways, so as to maximize the flux and in­
2017). Secondly, E. coli is more tolerant of monoterpene products such crease the precursor supply. Engineering of terpene synthase enzymes
as limonene and α-pinene (Himejima, Hobson, Otsuka, Wood, & Kubo, and introducing a heterologous pathway can be performed in parallel to
1992). According to Chubukov et al. (2015), the non-oxidized limonene test potential combinatorial effects on terpene production. Engineering
exhibited minimal toxicity to wild-type E. coli, while a point mutation in and optimization of orthogonal pathway could be a main direction of
alkyl hydroperoxidase (ahpC) can further alleviate the acute limonene future research on S. cerevisiae as a terpene-producing platform, which
toxicity caused by limonene hydroperoxide. E. coli also displayed can be attained by reducing the flux of competing pathway and chan­
enhanced tolerance to cyclohexane, pinene and limonene by expressing neling the metabolic flux of MVA pathway toward NPP. Filamentous
the AcrAB-TolC efflux proteins (Aono, Tsukagoshi, & Yamamoto, 1998; fungi as a potential microbial cell factory also expect the development of
Dunlop et al., 2011). However, E. coli is also reported with multiple engineering strategies to modify the endogenous MVA pathway for
drawbacks as a microbial host. So far, E. coli has not been classified as a enhancing terpene synthesis. In addition, the synergic effect of MVA and
GRAS organism by US FDA due to the possible contamination of the final MEP pathways overexpressing in E. coli on production of specific ter­
product by endotoxins (Ingy & Wim, 2017). Also, the MEP pathway in penes is worth further investigation. Future research should also
E. coli suffers from the natural cryptic regulation via various mechanisms consider the potential strategies in increasing microbial tolerance or
including cellular compartmentalization, gene duplication, bifunctional attenuate terpene toxicity in the presence of high levels of terpene
fusion proteins, transcriptional control and redox regulation. Introduc­ products. The effect of the fine-tuning of multi-gene expression on the
tion of the heterologous MVA pathway to E. coli might offer a bypass to synthesis of other isoprenoid-derived compounds also awaits further
the highly regulated MEP pathway (Davies, Jinkerson, & Posewitz, exploration.
2015). For instance, E. coli heterologously expressing a functional MVA While E. coli is the mostly used model bacterial platform, other
pathway, along with the overexpression of FPPS and integral membrane bacteria such as Bacillus subtilis has also been engineered for terpene
phosphatase PgpB, can produce up to 526.1 mg/L of farnesol that is production. B. subtilis is regarded as an attractive host for terpene bio­
higher than the reported yield (145.7 mg/L) in S. cerevisiae (Wang, Park, production due to its high isoprene productivity (Pramastya, Song,
Choi, & Kim, 2016). In an in silico comparison between the MEP and Elfahmi, Sukrasno, & Quax, 2020) and its generally recognized as safe
MVA pathways with different hosts, E. coli is superior to S. cerevisiae in (GRAS) status (Food and Drug Administration, 2018). In recent years,
the IPP production when using the heterologous MVA pathway, the B. subtilis has been reported effective in heterologous production of
native MEP pathway, and both pathways combined, respectively (Meng, terpenes and their intermediate via different engineering strategies
Wang, Hua, Zhang, & Wang, 2011). Further, the MVA and MEP path­ including CRISPR/Cas9 system, upregulation of MEP pathway genes
ways exist a synergic effect for terpene synthesis. In the study of Wang, (dxs, ispD, ispF, ispH, ispE, ispC, ispG, idi) and overexpression of GGPPS
Quan, et al. (2019), the flux of the MVA and MEP pathway in the dual and FPPS (Pramastya et al., 2020; Song et al., 2020; Song et al., 2021).
pathway overexpressing E. coli strain were 1.5- and 4.8-fold higher than Recent research also confirmed that portable bacterial CRISPR tran­
that of the strains overexpressing MEP and MVA pathway alone, scriptional activation method can be applied to the metabolic engi­
respectively. In addition, cytochrome P450 expression in E. coli is poor neering in Pseudomonas putida, which makes it a possible host for
compared with S. cerevisiae since most P450s are membrane-anchored to terpene production (Kiattisewee et al., 2021). Apart from bacteria,
the endoplasmic reticulum which are inserted by P450 reductases for co- another major prokaryotic host is cyanobacteria. Cyanobacteria are
expression (Moser & Pichler, 2019). Indeed, E. coli is a viable terpene- considered a potential microbial platform for sustainable bioproduction
producing host, but the efficiency of E. coli and S. cerevisiae in terpene because these photosynthetic prokaryotes require only CO2 and light for
production need to be further investigated and compared for selecting growth. Enhanced terpene production in these microorganisms can be
the most suitable microbial host. attained using a combinatorial metabolic engineering approach of mu­
Recent advances in the metabolic engineering of E. coli for terpene tation in GGPPS gene, modulation of GPPS expression, and optimization
production is supported by research efforts (Table 4). In the study of of the MEP pathway such as overexpression of IDI and 1-deoxy-D-xylu­
Jeong et al. (2020), E. coli HP variant obtained from parental E. coli lose-5-phosphate synthase (DXS) (Lin, Zhang, & Pakrasi, 2021; Rodri­
DH5α strain by adaptive evolution, displayed an increased cell fitness in gues & Lindberg, 2021). Further, other emerging bacterial platforms
terpene synthesis under adenylate cyclase (CyaA) mutation, as evi­ such as Rhodobacter capsulatus, Pseudomonas putida and Corynebacterium
denced by a five-fold increase to 193.2 mg/L in taxadiene production. glutamicum have been developed. Valencene, patchoulol and many other
Specifically, an insertion of mobile element IS10 in CyaA successfully terpenes have been successfully produced by these heterogeneous pro­
reduced the intracellular concentration of cyclic adenosine mono­ duction platforms (Kogure & Inui, 2018; Loeschcke & Thies, 2015;
phosphate (cAMP), which can regulate its receptor protein (CRP) to Troost et al., 2019).
rewire cell metabolism and thus improve cell fitness for terpene pro­ In addition to Saccharomyces as the model yeast platform, attempts
duction. In addition, Zong et al. (2019) reported a 29.51-fold increase in have also been made to other yeast species, such as Yarrowia lipolytica
nerol production in E. coli through a heterologous MVA pathway, when and Lipomyces starkeyi which can be used to produce (-)-α-bisabolol,
combining the overexpression of acetyl-CoA acetyltransferase (ERG10) linalool, α-farnesene, α-santalene, α‑zingiberene by overexpressing the
amplified from S. cerevisiae genomic DNA. An attempt of replacing rate-limiting enzymes (e.g. HMGR, FPPS, IDI) of the MVA pathway and
Erg20p with A. grandis geranyl diphosphate synthase (GPPS2) was also downregulating the competing squalene synthesis pathway (Cao, Wei,
observed in promoting linalool production in E. coli with Actinidia arguta Lin, & Hua, 2017; Dai et al., 2021; Jia et al., 2019; Ma et al., 2021; Yang,
linalool synthase by increasing the availability of GPP (Kong, Fu, Li, Pan, Nambou, Wei, & Hua, 2016). In addition to yeast and filamentous fungi,
& Guo, 2020). researchers have also explored other eukaryotic hosts such as Chlamy­
domonas reinhardtii and Phaeodactylum tricornutum as new microbial
5.3. Trends in microbial terpene production platforms, which can produce betulin, 13R(+) manoyl oxide, (E)-
α-bisabolene (D’Adamo et al., 2019; Lauersen et al., 2018; Wichmann,
Currently, plant-derived terpene products are unable to meet the Baier, Wentnagel, Lauersen, & Kruse, 2018). Overall, studies on these
gradually growing usage needs for commercial applications, rendering emerging platforms are still limited compared to S. cerevisiae, E. coli and
the microorganism as a feasible alternative terpene-producing platform. Aspergillus, thus it may have a long way to go to achieve their com­
However, microbial cell factories still demand further improvement for mercial applications.

12
Z. Liang et al. Food Research International 147 (2021) 110487

Table 4
Selective studies of recent advanced engineering strategies for terpene/terpenoid production in Escherichia coli.
Terpene/ Maximum yield Strain Engineering strategy Terpene synthase Reference
Terpenoid (mg/L)

Farnesol 526.1 DH5α a. Introduction of a heterologous MVA pathway N/A (Wang et al.,
b. Overexpression of FPPS and integral membrane 2016)
phosphatases PgpB and YbjG
Taxadiene 193.2 Adaptive HP variant a. Introduction of a heterologous MVA pathway Synthetic taxadiene (Jeong et al.,
of DH5α b. CyaA mutation synthase 2020)
Nerol 1.564 ± 0.102 K-12 MG1655 (DE3) a. Introduction of a heterologous MVA pathway Nerol synthase from Glycine (Zong et al.,
b. Overexpression of IDI1, MVD1, ERG8, ERG12, tHMG1 and max 2019)
ERG13
c. Overexpression of acetyl-CoA acetyltransferase (ERG10)
Linalool 63 ± 5.6 MG165 a. Introduction of a heterologous MVA pathway Linalool synthase from (Kong et al.,
b. Expression of Abies grandis geranyl diphosphate synthase Actinidia arguta 2020)
(GPPS2) instead of Erg20p
c. Overexpression of IDI1

N/A, not available; MVA, mevalonate; FPPS, farnesyl diphosphate (FPP) synthase; CyaA, adenylate cyclase.

6. Potential food applications of terpene products from monoterpenoids with a pleasant rose-like aroma, which can be
engineered microorganisms massively produced in an engineered S. cerevisiae strain by deploying
amino acid mutations in ERG20, replacing the ERG20 promoter with
Up to date, metabolic engineering of S. cerevisiae and E. coli are HXT1 promoter, or integrating extra IDI1 gene into the yeast genome
gradually applied in food and beverage industries for the production of (Fischer et al., 2011; Liu, Zhang, et al., 2013; Zhao et al., 2017). It is a
various terpene products potentially acting as flavoring agents, sweet­ flavoring additive applied in chewing gum, candy, ice cream, beverages,
ening agents, preservatives and functional food ingredients (Table 5). and many other products (Kutyna & Borneman, 2018). During the
S. cerevisiae is more attractive for food applications due to its safety alcoholic fermentation of non-aromatic Parellada grape must, the
which promotes approval of new food products (Lyu et al., 2019). In expression of the Ocimum basilicum (sweet basil) geraniol synthase gene
contrast, filamentous fungi as a terpene-producing platform have not in a S. cerevisiae wine strain can successfully yield 750 μg/L of geraniol,
been fully explored for food application yet, thus a promising new area which is higher than its olfactory sensory threshold and its quantities in
for food industry. Geraniol is one of the most in-demand wines obtained from the highly aromatic Muscat grapes. Further,

Table 5
Bioactivities and food applications of terpene products from engineered microorganisms.
Food category Terpene product Engineered Bioactivity Food application Reference
microorganism

Flavoring agent Geraniol S. cerevisiae Natural aroma/flavor a. Aroma/flavor additive in (Kutyna &
chewing gum, candy, ice Borneman,
cream, beverages 2018)
b. Aroma and monoterpene (Pardo et al.,
enhancement in wine 2015)
c. Replacing flavor hop (Denby et al.,
addition in classical 2018)
brewing
Linalool S. cerevisiae Natural aroma/flavor Replacing flavor hop (Denby et al.,
addition in classical 2018)
brewing
Limonene S. cerevisiae, E. coli Natural aroma/flavor Aroma/flavor additive in (Duetz et al.,
candy and soft drinks 2003)
Nerolidol S. cerevisiae Natural aroma/flavor N/A (Kutyna &
Borneman,
2018)
Valencene S. cerevisiae Natural aroma/flavor Aroma/flavor additive in (Beekwilder
food and drinks et al., 2014)

Sweetening Steviol glycosides S. cerevisiae, E. coli, Sweet taste Alternative and low-caloric (Prakash et al.,
agent A. nidulans sweetener 2014)
Mogroside V S. cerevisiae Sweet taste Sweetener in low-calorie (Zhao & Li,
sweet beverages 2018)

Preservative Monoterpenoids (geraniol, linalool, S. cerevisiae, E. coli Antimicrobial activity N/A (Lyu et al.,
limonene, etc.), sesquiterpenoids 2019)
(farnesene, cubebol, farnesol, etc.)

Functional food Limonene S. cerevisiae, E. coli Gastroprotective, antiviral, antihyperalgesic, Imparting health benefits to (Vieira et al.,
ingredients antidiabetic, anticancer, antinociceptive, Chinese Baijiu 2018)
antioxidant and anti-inflammatory activities (Hu et al.,
2020)
Steviol glycosides S. cerevisiae, E. coli, Anti-hyperglycemic, antihypertensive, anti- N/A (Yılmaz et al.,
A. nidulans inflammatory, anti-carcinogenic and 2020)
antioxidant activities
Geraniol, linalool, nerol S. cerevisiae Antioxidant activity Formulation of antioxidant (Wang, Chen
rich functional food et al., 2019)

N/A, not available; S. cerevisiae, Saccharomyces cerevisiae; E. coli, Escherichia coli; A. nidulans, Aspergillus nidulans.

13
Z. Liang et al. Food Research International 147 (2021) 110487

geraniol can be metabolized by yeast enzymes to excess monoterpenes, costs from feedstock supply (Lyu et al., 2019). It is worth mentioning
leading to a significant increase in total monoterpene concentration in that the food additives produced with genetically engineered microbes
wine. Therefore, the geraniol-engineered yeast can positively affect the usually get low consumer acceptance. Therefore, sustained information
terpene profile of wine, resulting in a monoterpene enhanced wine with and education are required in parallel with the application of the mi­
fruity and flowery aromas (Pardo et al., 2015). In specific types of beer, crobial technology to overcome the negative public perception
the dry-hopped flavor and aroma are mostly ascribed to the presence of (Schempp et al., 2018).
geraniol and linalool, which originate from the hop flowers added to the Several microbial engineering derived terpene products can poten­
wort during fermentation in classical brewing. In the study of Denby tially serve as bioactive components of functional food due to their
et al. (2018), S. cerevisiae as a brewing strain can be metabolically multiple health benefits. For instance, limonene is reported as being
engineered by overexpressing tHMGR and mutated FPPS (FPPS*), so as gastroprotective, antiviral, antihyperalgesic, antidiabetic, anticancer,
to increase GPP accumulation. This strain was also equipped with a full- antinociceptive, antioxidant and anti-inflammatory, indicating its po­
length geraniol synthase from O. basilicum (ObGES) and truncated tential application in functional food and high-value food products
linalool synthase from Mentha citrata (t67-McLIS) to produce linalool (Vieira, Beserra, Souza, Totti, & Rozza, 2018). During Chinese Baijiu
and geraniol, thereby replacing flavor hop addition. Limonene is fermentation, Hu et al. (2020) successfully established an orthogonal
another monoterpene frequently used as a flavoring agent in candy and pathway in S. cerevisiae for elevating the heterologous production of
soft drinks (Duetz, Bouwmeester, van Beilen, & Witholt, 2003), which (+)-limonene which can impart potential health benefits to Chinese
can be yielded in large amount in engineered S. cerevisiae by expressing Baijiu. Steviol glycosides are diterpenoid sweeteners derived from
tHMG1 mutant (Behrendorff et al., 2013) or ERG20 mutant (Jongedijk engineered microorganisms, which could be another bioactive compo­
et al., 2015), or by establishing an NPP-based orthogonal pathway nent of functional food by possessing anti-hyperglycemic, antihyper­
(Cheng et al., 2019; Hu et al., 2020; Ignea et al., 2019), or in engineered tensive, anti-inflammatory, anti-carcinogenic and antioxidant activities
E. coli with a heterologous MVA pathway (Alonso-Gutierrez et al., 2013). (Yılmaz, Görgüç, Uygun, & Bircan, 2020). Likewise, geraniol, linalool
Nerolidol is a typical sesquiterpene officially permitted as food flavoring and nerol with effective antioxidant activities, which are monoterpenoid
additive by US FDA (Food and Drug Administration, 2020), giving a products produced from engineered microorganisms, may also
wood- and fresh bark-like scent (Kutyna & Borneman, 2018). The het­ contribute to the formulation of antioxidant rich functional food (Wang,
erologous production of this sesquiterpene by S. cerevisiae can be largely Chen, & Hou, 2019).
enhanced in the background of ERG9 downregulation and/or tHMG1 More research efforts need to be invested in terpene production using
overexpression (Jackson, 2005). Another sesquiterpene valencene is microbial engineering platforms in food industry. Future research could
also commercially used as an additive to impart or reinforce fruity and explore the potential of filamentous fungi in the production of desired
woody flavor in food and drinks with a market volume of about 10,000 terpenes for food application. For instance, A. oryzae is a promising
kg per year (Beekwilder et al., 2014). This bicyclic sesquiterpene can be terpene-producing platform for this purpose due to its GRAS status. It is
massively produced in engineered S. cerevisiae via different engineering also worthy of exploring the production of valuable terpenes by engi­
strategies including tHMG1 overexpression (Farhi et al., 2011), ERG9 neering S. cerevisiae during wine fermentation, which can modify or
downregulation (Asadollahi et al., 2008; Chen et al., 2019) and ROX1 improve the aroma/flavor of wine. In addition, the technical obstacles in
deletion (Chen et al., 2019; Jakočiūnas et al., 2015). food application of microbial engineering derived terpene products, the
At present, there is a growing consumer demand for natural sweet­ safety concern regarding to the microbial engineering in food process­
ening agents, which can offer the similar sweetness of common sugar ing, and the influence of the processing parameters on the terpene
and restrict the high calorie intake. Therefore, S. cerevisiae is recom­ product quality also deserve further investigation.
mended as a microbial cell factory to manufacture alternative and low-
caloric sweetening agents. Steviol glycosides are typical sweeteners that 7. Conclusion
are a group of diterpene glycosides (rubusoside, steviolbioside, dulco­
side A, stevioside, rebaudioside-A, B, C, D, E, F, and M). They can be de Terpene production via microbial cell factories is of great signifi­
novo synthesized from GGPP with five heterologous enzymes in cance to substitute the traditional supply way. This review introduces
S. cerevisiae, conferring characteristic sweet tastes that are about 30 to the biosynthetic pathways of terpenes in different organisms. Both MVA
250 times more potent than sucrose (Gold et al., 2018; Prakash, Mar­ and MEP pathways exist in plant cells, while S. cerevisiae and other fungi
kosyan, & Bunders, 2014). E. coli is also a microbial chassis for the high- only harbor the MVA pathway. Bacteria such as E. coli only have
level production of steviol glycosides. The ent-copalyl diphosphate inherent MEP pathway and can be introduced a heterologous MVA
synthase (CPPS) and ent-kaurene synthase (KS) from Stevia rebaudiana pathway through engineering. Engineering of S. cerevisiae for promoting
can be heterologously expressed in E. coli to produce ent-kaurene, which terpene synthesis are described comprehensively, where overexpression
is a dedicated precursor responsible for steviol glycoside biosynthesis of HMGR variant, ERG20 upregulation, ERG9 downregulation, IDI1 gene
(Kong, Kang, Kim, Oh, & Lee, 2015). It is worth mentioning that ent- integration, ROX1 deletion and orthogonal pathway construction
kaurene can also be produced in engineered A. nidulans by over­ constitute the main engineering strategies to enhance terpene produc­
expressing F. fujikuroi ent-kaurene synthase (Bromann et al., 2016), tion. In recent years, numerous TPS genes have been identified in fungi,
indicating the vast potential of filamentous fungi as an engineering serving as potential heterologous TPS genes for the functional TPSs
platform for desired terpene products in food industry. Similarly, mog­ expression in S. cerevisiae. Apart from S. cerevisiae, filamentous fungi and
roside V also serves as a sweetener in low-calorie sweet beverages since E. coli also serve as efficient hosts for terpene synthesis. Nowadays, the
it is nearly 300 times sweeter than sucrose. It is a cucurbitane-type tri­ diverse terpene products from engineered microorganisms have been
terpenoid saponin derived from the introduction of Arabidopsis thaliana gradually applied in food and beverage industries as flavoring agents,
cytochrome P450 reductase 1 (AtCPR1) and various genes encoding key sweetening agents, preservatives and functional food ingredients.
enzymes in the biosynthesis pathway in S. cerevisiae (Zhao & Li, 2018). Future studies are required to investigate the heterologous production of
Multiple monoterpenoids (geraniol, linalool, limonene, etc.) and fungi-derived terpenes in S. cerevisiae and to improve the efficiency of
sesquiterpenoids (farnesene, cubebol, farnesol, etc.) generated through S. cerevisiae, filamentous fungi and E. coli in terpene production, so as to
metabolic engineering of S. cerevisiae and E. coli, are potential food expand their production scale and promote industrial applications.
preservatives due to their high antimicrobial properties. However, the
microbial technology has not been engaged in the industrial production Declaration of Competing Interest
of food preservatives, which is mainly due to the low yield, poor toxicity
resistance of yeast to terpene products, as well as the high production The authors declare that they have no known competing financial

14
Z. Liang et al. Food Research International 147 (2021) 110487

interests or personal relationships that could have appeared to influence Cao, X., Wei, L.-J., Lin, J.-Y., & Hua, Q. (2017). Enhancing linalool production by
engineering oleaginous yeast Yarrowia lipolytica. Bioresource Technology, 245,
the work reported in this paper.
1641–1644. https://doi.org/10.1016/j.biortech.2017.06.105.
Chen, H., Zhu, C., Zhu, M., Xiong, J., Ma, H., Zhuo, M., & Li, S. (2019). High production
of valencene in Saccharomyces cerevisiae through metabolic engineering. Microbial
Acknowledgement Cell Factories, 18(1), 1–14. https://doi.org/10.1186/s12934-019-1246-2.
Chen, Y., Partow, S., Scalcinati, G., Siewers, V., & Nielsen, J. (2012). Enhancing the copy
number of episomal plasmids in Saccharomyces cerevisiae for improved protein
This review research was supported by the food science research production. FEMS Yeast Research, 12(5), 598–607. https://doi.org/10.1111/j.1567-
group of the Faculty of Veterinary and Agricultural Sciences, University 1364.2012.00809.x.
Cheng, S., Liu, X., Jiang, G., Wu, J., Zhang, J.-L., Lei, D., … Zhao, G.-R. (2019).
of Melbourne. Orthogonal engineering of biosynthetic pathway for efficient production of limonene
in Saccharomyces cerevisiae. ACS Synthetic Biology, 8(5), 968–975. https://doi.org/
10.1021/acssynbio.9b00135.
Chubukov, V., Mingardon, F., Schackwitz, W., Baidoo, E. E., Alonso-Gutierrez, J., Hu, Q.,
Author Contribution
… Mukhopadhyay, A. (2015). Acute limonene toxicity in Escherichia coli is caused by
limonene hydroperoxide and alleviated by a point mutation in alkyl
Zijian Liang and Hang Zhi drafted the original manuscript. Critical hydroperoxidase AhpC. Applied Environmental Microbiology, 81(14), 4690–4696.
inputs and corrections were successively provided by Zhongxiang Fang, https://doi.org/10.1128/AEM.01102-15.
D’Adamo, S., Schiano di Visconte, G., Lowe, G., Szaub-Newton, J., Beacham, T.,
and Pangzhen Zhang during the preparation process. Pangzhen Zhang Landels, A., … Matthijs, M. (2019). Engineering the unicellular alga Phaeodactylum
helped in the conception and structure design of the manuscript and tricornutum for high-value plant triterpenoid production. Plant Biotechnology Journal,
final proofing of the manuscript for submission. 17(1), 75–87. https://doi.org/10.1111/pbi.12948.
Dai, Z., Liu, Y., Zhang, X., Shi, M., Wang, B., Wang, D., … Zhang, X. (2013). Metabolic
engineering of Saccharomyces cerevisiae for production of ginsenosides. Metabolic
References Engineering, 20, 146–156. https://doi.org/10.1016/j.ymben.2013.10.004.
Dai, Z., Pomraning, K. R., Panisko, E. A., Hofstad, B. A., Campbell, K. B., Kim, J., …
Magnuson, J. K. (2021). Genetically engineered oleaginous yeast Lipomyces starkeyi
Alonso-Gutierrez, J., Chan, R., Batth, T. S., Adams, P. D., Keasling, J. D., Petzold, C. J., &
for sesquiterpene α-zingiberene production. ACS Synthetic Biology. https://doi.org/
Lee, T. S. (2013). Metabolic engineering of Escherichia coli for limonene and perillyl
10.1021/acssynbio.0c00503.
alcohol production. Metabolic Engineering, 19, 33–41. https://doi.org/10.1016/j.
Davies, F. K., Jinkerson, R. E., & Posewitz, M. C. (2015). Toward a photosynthetic
ymben.2013.05.004.
microbial platform for terpenoid engineering. Photosynthesis Research, 123(3),
Aono, R., Tsukagoshi, N., & Yamamoto, M. (1998). Involvement of outer membrane
265–284. https://doi.org/10.1007/s11120-014-9979-6.
protein TolC, a possible member of the mar-sox regulon, in maintenance and
DeBose-Boyd, R. A. (2008). Feedback regulation of cholesterol synthesis: Sterol-
improvement of organic solvent tolerance of Escherichia coli K-12. Journal of
accelerated ubiquitination and degradation of HMG CoA reductase. Cell Research, 18
Bacteriology, 180(4), 938–944. https://doi.org/10.1128/JB.180.4.938-944.1998.
(6), 609–621. https://doi.org/10.1038/cr.2008.61.
Asadollahi, M. A., Maury, J., Møller, K., Nielsen, K. F., Schalk, M., Clark, A., & Nielsen, J.
Denby, C. M., Li, R. A., Vu, V. T., Costello, Z., Lin, W., Chan, L. J. G., … Keasling, J. D.
(2008). Production of plant sesquiterpenes in Saccharomyces cerevisiae: Effect of
(2018). Industrial brewing yeast engineered for the production of primary flavor
ERG9 repression on sesquiterpene biosynthesis. Biotechnology and Bioengineering, 99
determinants in hopped beer. Nature Communications, 9(1), 965. https://doi.org/
(3), 666–677. https://doi.org/10.1002/bit.21581.
10.1038/s41467-018-03293-x.
Asadollahi, M. A., Maury, J., Schalk, M., Clark, A., & Nielsen, J. (2010). Enhancement of
Duetz, W. A., Bouwmeester, H., van Beilen, J. B., & Witholt, B. (2003). Biotransformation
farnesyl diphosphate pool as direct precursor of sesquiterpenes through metabolic
of limonene by bacteria, fungi, yeasts, and plants. Applied Microbiology and
engineering of the mevalonate pathway in Saccharomyces cerevisiae. Biotechnology
Biotechnology, 61(4), 269–277. https://doi.org/10.1007/s00253-003-1221-y.
and Bioengineering, 106(1), 86–96. https://doi.org/10.1002/bit.22668.
Dugar, D., & Stephanopoulos, G. (2011). Relative potential of biosynthetic pathways for
Bach, T. (1986). Hydroxymethylglutaryl-CoA reductase, a key enzyme in phytosterol
biofuels and bio-based products. Nature Biotechnology, 29(12), 1074–1078. https://
synthesis? Lipids, 21(1), 82–88. https://doi.org/10.1007/BF02534307.
doi.org/10.1038/nbt.2055.
Bailey, A. M., Alberti, F., Kilaru, S., Collins, C. M., de Mattos-Shipley, K., Hartley, A. J., …
Dunlop, M. J., Dossani, Z. Y., Szmidt, H. L., Chu, H. C., Lee, T. S., Keasling, J. D., …
Foster, G. D. (2016). Identification and manipulation of the pleuromutilin gene
Mukhopadhyay, A. (2011). Engineering microbial biofuel tolerance and export using
cluster from Clitopilus passeckerianus for increased rapid antibiotic production.
efflux pumps. Molecular Systems Biology, 7(1), 487. https://doi.org/10.1038/
Scientific Reports, 6(1), 25202. https://doi.org/10.1038/srep25202.
msb.2011.21.
Beekwilder, J., van Houwelingen, A., Cankar, K., van Dijk, A. D., de Jong, R. M.,
Farhi, M., Marhevka, E., Masci, T., Marcos, E., Eyal, Y., Ovadis, M., … Vainstein, A.
Stoopen, G., … Bosch, D. (2014). Valencene synthase from the heartwood of Nootka
(2011). Harnessing yeast subcellular compartments for the production of plant
cypress (Callitropsis nootkatensis) for biotechnological production of valencene. Plant
terpenoids. Metabolic Engineering, 13(5), 474–481. https://doi.org/10.1016/j.
Biotechnology Journal, 12(2), 174–182. https://doi.org/10.1111/pbi.12124.
ymben.2011.05.001.
Behrendorff, J. B., Vickers, C. E., Chrysanthopoulos, P., & Nielsen, L. K. (2013). 2, 2-
Fischer, M. J., Meyer, S., Claudel, P., Bergdoll, M., & Karst, F. (2011). Metabolic
Diphenyl-1-picrylhydrazyl as a screening tool for recombinant monoterpene
engineering of monoterpene synthesis in yeast. Biotechnology Bioengineering, 108(8),
biosynthesis. Microbial Cell Factories, 12(1), 1–12. https://doi.org/10.1186/1475-
1883–1892. https://doi.org/10.1002/bit.23129.
2859-12-76.
Food and Drug Administration (2018). Microorganisms & microbial-derived ingredients
Bian, G., Yuan, Y., Tao, H., Shi, X., Zhong, X., Han, Y., & Liu, T. (2017). Production of
used in food (partial list). Retrieved from https://www.fda.gov/food/generally-rec
taxadiene by engineering of mevalonate pathway in Escherichia coli and endophytic
ognized-safe-gras/microorganisms-microbial-derived-ingredients-used-food-partia
fungus Alternaria alternata TPF6. Biotechnology Journal, 12(4), 1600697. https://doi.
l-list. Accessed January 3, 2020.
org/10.1002/biot.201600697.
Food and Drug Administration (2020). Code of federal regulations (CFR). Title 21: Food
Blackwell, M. (2011). The Fungi: 1, 2, 3 … 5.1 million species? American Journal of
and drugs. Chapter I - food and drug administration, department of health and
Botany, 98(3), 426-438. https://doi.org/10.3732/ajb.1000298.
human services, subchapter B - food for human consumption (continued), Part 172 -
Brennan, T. C., Williams, T. C., Schulz, B. L., Palfreyman, R. W., Krömer, J. O., &
food additives permitted for direct addition to food for human consumption, subpart
Nielsen, L. K. (2015). Evolutionary engineering improves tolerance for replacement
f - flavoring agents and related substances, subpart 172.510 - natural flavoring
jet fuels in Saccharomyces cerevisiae. Applied Environmental Microbiology, 81(10),
substances and natural substances used in conjunction with flavors. Retrieved from
3316–3325. https://doi.org/10.1128/AEM.04144-14.
https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?
Brennan, T. C. R., Krömer, J. O., & Nielsen, L. K. (2013). Physiological and
fr=172.510. Accessed September 10, 2020.
transcriptional responses of Saccharomyces cerevisiae to d-limonene show changes to
Fujii, R., Minami, A., Tsukagoshi, T., Sato, N., Sahara, T., Ohgiya, S., … Oikawa, H.
the cell wall but not to the plasma membrane. Applied and Environmental
(2011). Total biosynthesis of diterpene aphidicolin, a specific inhibitor of DNA
Microbiology, 79(12), 3590–3600. https://doi.org/10.1128/AEM.00463-13.
polymerase α: Heterologous expression of four biosynthetic genes in Aspergillus
Brennan, T. C. R., Turner, C. D., Krömer, J. O., & Nielsen, L. K. (2012). Alleviating
oryzae. Bioscience, Biotechnology, and Biochemistry, 75(9), 1813–1817. https://doi.
monoterpene toxicity using a two-phase extractive fermentation for the
org/10.1271/bbb.110366.
bioproduction of jet fuel mixtures in Saccharomyces cerevisiae. Biotechnology and
Gold, N. D., Fossati, E., Hansen, C. C., DiFalco, M., Douchin, V., & Martin, V. J. J. (2018).
Bioengineering, 109(10), 2513–2522. https://doi.org/10.1002/bit.24536.
A combinatorial approach to study cytochrome P450 enzymes for de novo
Bromann, K., Toivari, M., Viljanen, K., Ruohonen, L., & Nakari-Setälä, T. (2016).
production of steviol glucosides in baker’s yeast. ACS Synthetic Biology, 7(12),
Engineering Aspergillus nidulans for heterologous ent-kaurene and gamma-terpinene
2918–2929. https://doi.org/10.1021/acssynbio.8b00470.
production. Applied Microbiology and Biotechnology, 100(14), 6345–6359. https://
Greenhagen, B. T., O’Maille, P. E., Noel, J. P., & Chappell, J. (2006). Identifying and
doi.org/10.1007/s00253-016-7517-5.
manipulating structural determinates linking catalytic specificities in terpene
Burg, J. S., & Espenshade, P. J. (2011). Regulation of HMG-CoA reductase in mammals
synthases. Proceedings of the National Academy of Sciences, 103(26), 9826. https://
and yeast. Progress in Lipid Research, 50(4), 403–410. https://doi.org/10.1016/j.
doi.org/10.1073/pnas.0601605103.
plipres.2011.07.002.
Henry, K. W., Nickels, J. T., & Edlind, T. D. (2002). ROX1 and ERG regulation in
Callari, R., Meier, Y., Ravasio, D., & Heider, H. (2018). Dynamic control of ERG20 and
Saccharomyces cerevisiae: Implications for antifungal susceptibility. Eukaryotic Cell, 1
ERG9 expression for improved casbene production in Saccharomyces cerevisiae.
(6), 1041–1044. https://doi.org/10.1128/EC.1.6.1041-1044.2002.
Frontiers in Bioengineering and Biotechnology, 6, 160. https://doi.org/10.3389/
fbioe.2018.00160.

15
Z. Liang et al. Food Research International 147 (2021) 110487

Hezari, M., & Croteau, R. (1997). Taxol biosynthesis: An update. Planta Medica, 63(04), Kong, M. K., Kang, H.-J., Kim, J. H., Oh, S. H., & Lee, P. C. (2015). Metabolic engineering
291–295. https://doi.org/10.1055/s-2006-957684. of the Stevia rebaudiana ent-kaurene biosynthetic pathway in recombinant Escherichia
Himejima, M., Hobson, K. R., Otsuka, T., Wood, D. L., & Kubo, I. (1992). Antimicrobial coli. Journal of Biotechnology, 214, 95–102. https://doi.org/10.1016/j.
terpenes from oleoresin of ponderosa pine tree Pinus ponderosa: A defense jbiotec.2015.09.016.
mechanism against microbial invasion. Journal of Chemical Ecology, 18(10), Kong, S., Fu, X., Li, X., Pan, H., & Guo, D. (2020). De novo biosynthesis of linalool from
1809–1818. https://doi.org/10.1007/BF02751105. glucose in engineered Escherichia coli. Enzyme and Microbial Technology, 140,
Hood, E. E., Gelvin, S. B., Melchers, L. S., & Hoekema, A. (1993). New Agrobacterium 109614. https://doi.org/10.1016/j.enzmictec.2020.109614.
helper plasmids for gene transfer to plants. Transgenic Research, 2(4), 208–218. Kuranda, K., François, J., & Palamarczyk, G. (2009). The isoprenoid pathway and
https://doi.org/10.1007/BF01977351. transcriptional response to its inhibitors in the yeast Saccharomyces cerevisiae. FEMS
Hu, F., Liu, J., Du, G., Hua, Z., Zhou, J., & Chen, J. (2012). Key cytomembrane ABC Yeast Research, 10(1), 14–27. https://doi.org/10.1111/j.1567-1364.2009.00560.x.
transporters of Saccharomyces cerevisiae fail to improve the tolerance to d-limonene. Kutyna, D. R., & Borneman, A. (2018). Heterologous production of flavour and aroma
Biotechnology Letters, 34(8), 1505–1509. https://doi.org/10.1007/s10529-012-0931- compounds in Saccharomyces cerevisiae. Genes, 9(7), 326. https://doi.org/10.3390/
6. genes9070326.
Hu, Z., Lin, L., Li, H., Li, P., Weng, Y., Zhang, C., … Xiao, D. (2020). Engineering Lauersen, K. J., Wichmann, J., Baier, T., Kampranis, S. C., Pateraki, I., Møller, B. L., &
Saccharomyces cerevisiae for production of the valuable monoterpene d-limonene Kruse, O. (2018). Phototrophic production of heterologous diterpenoids and a
during Chinese Baijiu fermentation. Journal of Industrial Microbiology and hydroxy-functionalized derivative from Chlamydomonas reinhardtii. Metabolic
Biotechnology, 47(6–7), 511–523. https://doi.org/10.1007/s10295-020-02284-6. Engineering, 49, 116–127. https://doi.org/10.1016/j.ymben.2018.07.005.
Ichinose, H., & Kitaoka, T. (2018). Insight into metabolic diversity of the brown-rot Leber, R., Zenz, R., Schröttner, K., Fuchsbichler, S., Pühringer, B., & Turnowsky, F.
basidiomycete Postia placenta responsible for sesquiterpene biosynthesis: Semi- (2001). A novel sequence element is involved in the transcriptional regulation of
comprehensive screening of cytochrome P450 monooxygenase involved in expression of the ERG1 (squalene epoxidase) gene in Saccharomyces cerevisiae.
protoilludene metabolism. Microbial Biotechnology, 11(5), 952–965. https://doi.org/ European Journal of Biochemistry, 268(4), 914–924. https://doi.org/10.1046/j.1432-
10.1111/1751-7915.13304. 1327.2001.01940.x.
Ignea, C., Cvetkovic, I., Loupassaki, S., Kefalas, P., Johnson, C. B., Kampranis, S. C., & Li, Z., Howell, K., Fang, Z., & Zhang, P. (2020). Sesquiterpenes in grapes and wines:
Makris, A. M. (2011). Improving yeast strains using recyclable integration cassettes, Occurrence, biosynthesis, functionality, and influence of winemaking processes.
for the production of plant terpenoids. Microbial Cell Factories, 10(1), 4. https://doi. Comprehensive Reviews in Food Science and Food Safety, 19(1), 247–281. https://doi.
org/10.1186/1475-2859-10-4. org/10.1111/1541-4337.12516.
Ignea, C., Pontini, M., Maffei, M. E., Makris, A. M., & Kampranis, S. C. (2014). Liang, Z., Fang, Z., Pai, A., Luo, J., Gan, R., Gao, Y., … Zhang, P. (2020). Glycosidically
Engineering monoterpene production in yeast using a synthetic dominant negative bound aroma precursors in fruits: A comprehensive review. Critical Reviews in Food
geranyl diphosphate synthase. ACS Synthetic Biology, 3(5), 298–306. https://doi.org/ Science and Nutrition, 1–29. https://doi.org/10.1080/10408398.2020.1813684.
10.1021/sb400115e. Liang, Z., Zhang, P., & Fang, Z. (2020). Modern technologies for extraction of aroma
Ignea, C., Raadam, M. H., Motawia, M. S., Makris, A. M., Vickers, C. E., & compounds from fruit peels: A review. Critical Reviews in Food Science and Nutrition,
Kampranis, S. C. (2019). Orthogonal monoterpenoid biosynthesis in yeast 1–24. https://doi.org/10.1080/10408398.2020.1840333.
constructed on an isomeric substrate. Nature Communications, 10(1), 3799. https:// Lin, P.-C., Zhang, F., & Pakrasi, H. B. (2021). Enhanced limonene production in a fast-
doi.org/10.1038/s41467-019-11290-x. growing cyanobacterium through combinatorial metabolic engineering. Metabolic
Ignea, C., Trikka, F. A., Kourtzelis, I., Argiriou, A., Kanellis, A. K., Kampranis, S. C., & Engineering Communications, 12, e00164. https://doi.org/10.1016/j.mec.2021.
Makris, A. M. (2012). Positive genetic interactors of HMG2 identify a new set of e00164.
genetic perturbations for improving sesquiterpene production in Saccharomyces Liscum, L., Finer-Moore, J., Stroud, R., Luskey, K., Brown, M. S., & Goldstein, J. L.
cerevisiae. Microbial Cell Factories, 11(1), 162. https://doi.org/10.1186/1475-2859- (1985). Domain structure of 3-hydroxy-3-methylglutaryl coenzyme A reductase, a
11-162. glycoprotein of the endoplasmic reticulum. Journal of Biological Chemistry, 260(1),
Ignea, C., Trikka, F. A., Nikolaidis, A. K., Georgantea, P., Ioannou, E., Loupassaki, S., … 522–530.
Kampranis, S. C. (2015). Efficient diterpene production in yeast by engineering Liu, C., Minami, A., Ozaki, T., Wu, J., Kawagishi, H., Maruyama, J.-I., & Oikawa, H.
Erg20p into a geranylgeranyl diphosphate synthase. Metabolic Engineering, 27, (2019). Efficient reconstitution of Basidiomycota diterpene erinacine gene cluster in
65–75. https://doi.org/10.1016/j.ymben.2014.10.008. Ascomycota host Aspergillus oryzae based on genomic DNA sequences. Journal of the
Ingy, I. A., & Wim, J. Q. (2017). A glimpse into the biosynthesis of terpenoids. KnE Life American Chemical Society, 141(39), 15519–15523. https://doi.org/10.1021/
Sciences, 3(5), 81–98. https://doi.org/10.18502/kls.v3i5.981. jacs.9b08935.
Itoh, T., Tokunaga, K., Matsuda, Y., Fujii, I., Abe, I., Ebizuka, Y., & Kushiro, T. (2010). Liu, H., Sun, Y., Ramos, K. R. M., Nisola, G. M., Valdehuesa, K. N. G., Lee, W. K., …
Reconstitution of a fungal meroterpenoid biosynthesis reveals the involvement of a Chung, W.-J. (2013). Combination of Entner-Doudoroff pathway with MEP increases
novel family of terpene cyclases. Nature Chemistry, 2(10), 858–864. https://doi.org/ isoprene production in engineered Escherichia coli. Plos One, 8(12), Article e83290.
10.1038/nchem.764. https://doi.org/10.1371/journal.pone.0083290.
Jackson, B. E. (2005). Yeast as a host for sesquiterpene production ((Ph.D)). Houston, Texas, Liu, J., Zhang, W., Du, G., Chen, J., & Zhou, J. (2013). Overproduction of geraniol by
USA: Rice University. enhanced precursor supply in Saccharomyces cerevisiae. Journal of Biotechnology, 168
Jakočiūnas, T., Bonde, I., Herrgård, M., Harrison, S. J., Kristensen, M., Pedersen, L. E., … (4), 446–451. https://doi.org/10.1016/j.jbiotec.2013.10.017.
Keasling, J. D. (2015). Multiplex metabolic pathway engineering using CRISPR/Cas9 Loeschcke, A., & Thies, S. (2015). Pseudomonas putida—a versatile host for the
in Saccharomyces cerevisiae. Metabolic Engineering, 28, 213–222. https://doi.org/ production of natural products. Applied Microbiology and Biotechnology, 99(15),
10.1016/j.ymben.2015.01.008. 6197–6214. https://doi.org/10.1007/s00253-015-6745-4.
Jeong, S.-H., Park, J.-B., Wang, Y., Kim, G.-H., Zhang, G., Wei, G., … Kim, S.-W. (2020). López, J., Essus, K., Kim, I.-K., Pereira, R., Herzog, J., Siewers, V., … Agosin, E. (2015).
Regulatory molecule cAMP changes cell fitness of the engineered Escherichia coli for Production of β-ionone by combined expression of carotenogenic and plant CCD1
terpenoids production. Metabolic Engineering, 65, 178–184. https://doi.org/10.1016/ genes in Saccharomyces cerevisiae. Microbial Cell Factories, 14(1), 84. https://doi.org/
j.ymben.2020.11.009. 10.1186/s12934-015-0273-x.
Jia, D., Xu, S., Sun, J., Zhang, C., Li, D., & Lu, W. (2019). Yarrowia lipolytica construction Lu, Z., Peng, B., Ebert, B. E., Dumsday, G., & Vickers, C. E. (2021). Auxin-mediated
for heterologous synthesis of α-santalene and fermentation optimization. Applied protein depletion for metabolic engineering in terpene-producing yeast. Nature
Microbiology and Biotechnology, 103(8), 3511–3520. https://doi.org/10.1007/ Communications, 12(1), 1051. https://doi.org/10.1038/s41467-021-21313-1.
s00253-019-09735-w. Lyu, X., Lee, J., & Chen, W. N. (2019). Potential natural food preservatives and their
Jones, K. L., Kim, S.-W., & Keasling, J. (2000). Low-copy plasmids can perform as well as sustainable production in yeast: Terpenoids and polyphenols. Journal of Agricultural
or better than high-copy plasmids for metabolic engineering of bacteria. Metabolic and Food Chemistry, 67(16), 4397–4417. https://doi.org/10.1021/acs.jafc.8b07141.
Engineering, 2(4), 328–338. https://doi.org/10.1006/mben.2000.0161. Ma, Y., Li, W., Mai, J., Wang, J., Wei, Y., Ledesma-Amaro, R., & Ji, X.-J. (2021).
Jongedijk, E., Cankar, K., Ranzijn, J., van der Krol, S., Bouwmeester, H., & Beekwilder, J. Engineering Yarrowia lipolytica for sustainable production of the chamomile
(2015). Capturing of the monoterpene olefin limonene produced in Saccharomyces sesquiterpene (-)-α-bisabolol. Green Chemistry, 23(2), 780–787. https://doi.org/
cerevisiae. Yeast, 32(1), 159–171. https://doi.org/10.1002/yea.3038. 10.1039/D0GC03180A.
Katayama, T., Nakamura, H., Zhang, Y., Pascal, A., Fujii, W., & Maruyama, J.-I. (2019). Matsuda, Y., Awakawa, T., & Abe, I. (2013). Reconstituted biosynthesis of fungal
Forced recycling of an AMA1-based genome-editing plasmid allows for efficient meroterpenoid andrastin A. Tetrahedron, 69(38), 8199–8204. https://doi.org/
multiple gene deletion/integration in the industrial filamentous fungus Aspergillus 10.1016/j.tet.2013.07.029.
oryzae. Applied and Environmental Microbiology, 85(3), e01896–01818. https://doi. Meadows, A. L., Hawkins, K. M., Tsegaye, Y., Antipov, E., Kim, Y., Raetz, L., …
org/10.1128/AEM.01896-18. Tsong, A. E. (2016). Rewriting yeast central carbon metabolism for industrial
Katayama, T., Tanaka, Y., Okabe, T., Nakamura, H., Fujii, W., Kitamoto, K., & isoprenoid production. Nature, 537(7622), 694–697. https://doi.org/10.1038/
Maruyama, J.-I. (2016). Development of a genome editing technique using the nature19769.
CRISPR/Cas9 system in the industrial filamentous fungus Aspergillus oryzae. Meng, H., Wang, Y., Hua, Q., Zhang, S., & Wang, X. (2011). In silico analysis and
Biotechnology Letters, 38(4), 637–642. https://doi.org/10.1007/s10529-015-2015-x. experimental improvement of taxadiene heterologous biosynthesis in Escherichia coli.
Kiattisewee, C., Dong, C., Fontana, J., Sugianto, W., Peralta-Yahya, P., Carothers, J. M., & Biotechnology Bioprocess Engineering, 16(2), 205–215. https://doi.org/10.1007/
Zalatan, J. G. (2021). Portable bacterial CRISPR transcriptional activation enables s12257-010-0329-z.
metabolic engineering in Pseudomonas putida. Metabolic Engineering. https://doi.org/ Mischko, W., Hirte, M., Fuchs, M., Mehlmer, N., & Brück, T. B. (2018). Identification of
10.1016/j.ymben.2021.04.002. sesquiterpene synthases from the Basidiomycota Coniophora puteana for the efficient
Kogure, T., & Inui, M. (2018). Recent advances in metabolic engineering of and highly selective β-copaene and cubebol production in E. coli. Microbial Cell
Corynebacterium glutamicum for bioproduction of value-added aromatic chemicals Factories, 17(1), 1–13. https://doi.org/10.1186/s12934-018-1010-z.
and natural products. Applied Microbiology and Biotechnology, 102(20), 8685–8705. Montañés, F. M., Pascual-Ahuir, A., & Proft, M. (2011). Repression of ergosterol
https://doi.org/10.1007/s00253-018-9289-6. biosynthesis is essential for stress resistance and is mediated by the Hog1 MAP

16
Z. Liang et al. Food Research International 147 (2021) 110487

kinase and the Mot3 and Rox1 transcription factors. Molecular Microbiology, 79(4), Trikka, F. A., Nikolaidis, A., Athanasakoglou, A., Andreadelli, A., Ignea, C., Kotta, K., …
1008–1023. https://doi.org/10.1111/j.1365-2958.2010.07502.x. Makris, A. M. (2015). Iterative carotenogenic screens identify combinations of yeast
Moser, S., & Pichler, H. (2019). Identifying and engineering the ideal microbial terpenoid gene deletions that enhance sclareol production. Microbial Cell Factories, 14(1), 60.
production host. Applied Microbiology Biotechnology, 103(14), 5501–5516. https:// https://doi.org/10.1186/s12934-015-0246-0.
doi.org/10.1007/s00253-019-09892-y. Troost, K., Loeschcke, A., Hilgers, F., Özgür, A. Y., Weber, T. M., Santiago-Schübel, B., …
Nagamine, S., Liu, C., Nishishita, J., Kozaki, T., Sogahata, K., Sato, Y., … Maruyama, J.-I. Drepper, T. (2019). Engineered Rhodobacter capsulatus as a phototrophic platform
(2019). Ascomycete Aspergillus oryzae is an efficient expression host for production organism for the synthesis of plant sesquiterpenoids. Frontiers in Microbiology, 10
of basidiomycete terpenes by using genomic DNA sequences. Applied Environmental (1998). https://doi.org/10.3389/fmicb.2019.01998.
Microbiology, 85(15), e00409–00419. https://doi.org/10.1128/AEM.00409-19. Uribe, S., Ramirez, J., & Peña, A. (1985). Effects of beta-pinene on yeast membrane
Özaydın, B., Burd, H., Lee, T. S., & Keasling, J. D. (2013). Carotenoid-based phenotypic functions. Journal of Bacteriology, 161(3), 1195. https://doi.org/10.1128/
screen of the yeast deletion collection reveals new genes with roles in isoprenoid JB.161.3.1195-1200.1985.
production. Metabolic Engineering, 15, 174–183. https://doi.org/10.1016/j. Vedula, L. S., Jiang, J., Zakharian, T., Cane, D. E., & Christianson, D. W. (2008).
ymben.2012.07.010. Structural and mechanistic analysis of trichodiene synthase using site-directed
Paramasivan, K., & Mutturi, S. (2017). Progress in terpene synthesis strategies through mutagenesis: Probing the catalytic function of tyrosine-295 and the asparagine-225/
engineering of Saccharomyces cerevisiae. Critical Reviews in Biotechnology, 37(8), serine-229/glutamate-233–Mg2+B motif. Archives of Biochemistry Biophysics, 469(2),
974–989. https://doi.org/10.1080/07388551.2017.1299679. 184–194. https://doi.org/10.1016/j.abb.2007.10.015.
Pardo, E., Rico, J., Gil, J. V., & Orejas, M. (2015). De novo production of six key grape Vieira, A. J., Beserra, F. P., Souza, M. C., Totti, B. M., & Rozza, A. L. (2018). Limonene:
aroma monoterpenes by a geraniol synthase-engineered S. cerevisiae wine strain. Aroma of innovation in health and disease. Chemico-Biological Interactions, 283,
Microbial Cell Factories, 14(1), 136. https://doi.org/10.1186/s12934-015-0306-5. 97–106. https://doi.org/10.1016/j.cbi.2018.02.007.
Peralta-Yahya, P. P., Ouellet, M., Chan, R., Mukhopadhyay, A., Keasling, J. D., & Wang, C.-Y., Chen, Y.-W., & Hou, C.-Y. (2019). Antioxidant and antibacterial activity of
Lee, T. S. (2011). Identification and microbial production of a terpene-based seven predominant terpenoids. International Journal of Food Properties, 22(1),
advanced biofuel. Nature Communications, 2(1), 1–8. https://doi.org/10.1038/ 230–238. https://doi.org/10.1080/10942912.2019.1582541.
ncomms1494. Wang, C., Park, J. E., Choi, E. S., & Kim, S. W. (2016). Farnesol production in Escherichia
Pitera, D. J., Paddon, C. J., Newman, J. D., & Keasling, J. D. (2007). Balancing a coli through the construction of a farnesol biosynthesis pathway–application of PgpB
heterologous mevalonate pathway for improved isoprenoid production in Escherichia and YbjG phosphatases. Biotechnology Journal, 11(10), 1291–1297. https://doi.org/
coli. Metabolic Engineering, 9(2), 193–207. https://doi.org/10.1016/j. 10.1002/biot.201600250.
ymben.2006.11.002. Wang, K., & Ohnuma, S.-I. (1999). Chain-length determination mechanism of isoprenyl
Prakash, I., Markosyan, A., & Bunders, C. (2014). Development of next generation stevia diphosphate synthases and implications for molecular evolution. Trends in
sweetener: Rebaudioside M. Foods, 3(1), 162–175. https://doi.org/10.3390/ Biochemical Sciences, 24(11), 445–451. https://doi.org/10.1016/S0968-0004(99)
foods3010162. 01464-4.
Pramastya, H., Song, Y., Elfahmi, E. Y., Sukrasno, S., & Quax, W. J. (2020). Positioning Wang, Q., Quan, S., & Xiao, H. (2019). Towards efficient terpenoid biosynthesis:
Bacillus subtilis as terpenoid cell factory. Journal of Applied Microbiology. https://doi. Manipulating IPP and DMAPP supply. Bioresources and Bioprocessing, 6(1), 6. https://
org/10.1111/jam.14904. doi.org/10.1186/s40643-019-0242-z.
Quin, M. B., Flynn, C. M., & Schmidt-Dannert, C. (2014). Traversing the fungal Wang, Y., Lim, L., DiGuistini, S., Robertson, G., Bohlmann, J., & Breuil, C. (2013).
terpenome. Natural Product Reports, 31(10), 1449–1473. https://doi.org/10.1039/ A specialized ABC efflux transporter GcABC-G1 confers monoterpene resistance to
C4NP00075G. Grosmannia clavigera, a bark beetle-associated fungal pathogen of pine trees. New
Quin, M. B., Flynn, C. M., Wawrzyn, G. T., Choudhary, S., & Schmidt- Dannert, C. (2013). Phytologist, 197(3), 886–898. https://doi.org/10.1111/nph.12063.
Mushroom hunting by using bioinformatics: Application of a predictive framework Wawrzyn, G. T., Quin, M. B., Choudhary, S., López-Gallego, F., & Schmidt-Dannert, C.
facilitates the selective identification of sesquiterpene synthases in Basidiomycota. (2012). Draft genome of Omphalotus olearius provides a predictive framework for
ChemBioChem, 14(18), 2480–2491. https://doi.org/10.1002/cbic.201300349. sesquiterpenoid natural product biosynthesis in Basidiomycota. Chemistry & Biology,
Rodrigues, J. S., & Lindberg, P. (2021). Metabolic engineering of Synechocystis sp. PCC 19(6), 772–783. https://doi.org/10.1016/j.chembiol.2012.05.012.
6803 for improved bisabolene production. Metabolic Engineering. Communications, Wichmann, J., Baier, T., Wentnagel, E., Lauersen, K. J., & Kruse, O. (2018). Tailored
12, Article e00159. https://doi.org/10.1016/j.mec.2020.e00159. carbon partitioning for phototrophic production of (E)-α-bisabolene from the green
Schempp, F. M., Drummond, L., Buchhaupt, M., & Schrader, J. (2018). Microbial cell microalga Chlamydomonas reinhardtii. Metabolic Engineering, 45, 211–222. https://
factories for the production of terpenoid flavor and fragrance compounds. Journal of doi.org/10.1016/j.ymben.2017.12.010.
Agricultural and Food Chemistry, 66(10), 2247–2258. https://doi.org/10.1021/acs. Wu, W., Tran, W., Taatjes, C. A., Alonso-Gutierrez, J., Lee, T. S., & Gladden, J. M. (2016).
jafc.7b00473. Rapid discovery and functional characterization of terpene synthases from four
Schilmiller, A. L., Schauvinhold, I., Larson, M., Xu, R., Charbonneau, A. L., Schmidt, A., endophytic xylariaceae. Plos One, 11(2), e0146983. https://doi.org/10.1371/
… Pichersky, E. (2009). Monoterpenes in the glandular trichomes of tomato are journal.pone.0146983.
synthesized from a neryl diphosphate precursor rather than geranyl diphosphate. Yang, X., Nambou, K., Wei, L., & Hua, Q. (2016). Heterologous production of α-farnesene
Proceedings of the National Academy of Sciences, 106(26), 10865–10870. https://doi. in metabolically engineered strains of Yarrowia lipolytica. Bioresource Technology,
org/10.1073/pnas.0904113106. 216, 1040–1048. https://doi.org/10.1016/j.biortech.2016.06.028.
Shaw, J. J., Berbasova, T., Sasaki, T., Jefferson-George, K., Spakowicz, D. J., Yılmaz, F. M., Görgüç, A., Uygun, Ö., & Bircan, C. (2020). Steviol glycosides and
Dunican, B. F., … Strobel, S. A. (2015). Identification of a fungal 1, 8-cineole polyphenols extraction from Stevia rebaudiana Bertoni leaves using maceration,
synthase from Hypoxylon sp. with specificity determinants in common with the plant microwave-, and ultrasound-assisted techniques. Separation Science and Technology,
synthases. Journal of Biological Chemistry, 290(13), 8511–8526. https://doi.org/ 1–13. https://doi.org/10.1080/01496395.2020.1743311.
10.1074/jbc.M114.636159. Yoshikuni, Y., Ferrin, T. E., & Keasling, J. D. (2006). Designed divergent evolution of
Shi, T.-Q., Gao, J., Wang, W.-J., Wang, K.-F., Xu, G.-Q., Huang, H., & Ji, X.-J. (2019). enzyme function. Nature, 440(7087), 1078–1082. https://doi.org/10.1038/
CRISPR/Cas9-based genome editing in the filamentous fungus Fusarium fujikuroi and nature04607.
its application in strain engineering for gibberellic acid production. ACS Synthetic Zhang, C., Chen, X., Orban, A., Shukal, S., Birk, F., Too, H.-P., & Rühl, M. (2020).
Biology, 8(2), 445–454. https://doi.org/10.1021/acssynbio.8b00478. Agrocybe aegerita serves as a gateway for identifying sesquiterpene biosynthetic
Soliman, S. S. M., Mosa, K. A., El-Keblawy, A. A., & Husseiny, M. I. (2017). Exogenous enzymes in higher fungi. ACS Chemical Biology, 15(5), 1268–1277. https://doi.org/
and endogenous increase in fungal GGPP increased fungal Taxol production. Applied 10.1021/acschembio.0c00155.
Microbiology and Biotechnology, 101(20), 7523–7533. https://doi.org/10.1007/ Zhang, D. L., Jennings, S. M., Robinson, G. W., & Poulter, C. D. (1993). Yeast squalene
s00253-017-8509-9. synthase: Expression, purification, and characterization of soluble recombinant
Song, Y., Guan, Z., van Merkerk, R., Pramastya, H., Abdallah, I. I., Setroikromo, R., & enzyme. Archives of Biochemistry and Biophysics, 304(1), 133–143. https://doi.org/
Quax, W. J. (2020). Production of squalene in Bacillus subtilis by squalene synthase 10.1006/abbi.1993.1331.
screening and metabolic engineering. Journal of Agricultural and Food Chemistry, 68 Zhang, G., Cao, Q., Liu, J., Liu, B., Li, J., & Li, C. (2015). Refactoring β-amyrin synthesis
(15), 4447–4455. https://doi.org/10.1021/acs.jafc.0c00375. in Saccharomyces cerevisiae. AIChE Journal, 61(10), 3172–3179. https://doi.org/
Song, Y., He, S., Abdallah, I. I., Jopkiewicz, A., Setroikromo, R., van Merkerk, R., … 10.1002/aic.14950.
Quax, W. J. (2021). Engineering of multiple modules to improve amorphadiene Zhang, P., Fuentes, S., Siebert, T., Krstic, M., Herderich, M., Barlow, E. W. R., &
production in Bacillus subtilis using CRISPR-Cas9. Journal of Agricultural and Food Howell, K. (2016). Terpene evolution during the development of Vitis vinifera L. cv.
Chemistry, 69(16), 4785–4794. https://doi.org/10.1021/acs.jafc.1c00498. Shiraz grapes. Food Chemistry, 204, 463–474. https://doi.org/10.1016/j.
Sun, W., Zhao, Y.-J., & Li, C. (2019). De novo synthesis of plant natural products in yeast. foodchem.2016.02.125.
In T. P. Basso (Ed.), Yeasts in Biotechnology (pp. 105–122). IntechOpen. Zhang, Y., Nielsen, J., & Liu, Z. (2017). Engineering yeast metabolism for production of
Tagami, K., Minami, A., Fujii, R., Liu, C., Tanaka, M., Gomi, K., … Oikawa, H. (2014). terpenoids for use as perfume ingredients, pharmaceuticals and biofuels. FEMS Yeast
Rapid reconstitution of biosynthetic machinery for fungal metabolites in Aspergillus Research, 17(8), fox080. https://doi.org/10.1093/femsyr/fox080.
oryzae: Total biosynthesis of aflatrem. ChemBioChem, 15(14), 2076–2080. https:// Zhao, J., Bao, X., Li, C., Shen, Y., & Hou, J. (2016). Improving monoterpene geraniol
doi.org/10.1002/cbic.201402195. production through geranyl diphosphate synthesis regulation in Saccharomyces
Tholl, D. (2006). Terpene synthases and the regulation, diversity and biological roles of cerevisiae. Applied Microbiology Biotechnology, 100(10), 4561–4571. https://doi.org/
terpene metabolism. Current Opinion in Plant Biology, 9(3), 297–304. https://doi.org/ 10.1007/s00253-016-7375-1.
10.1016/j.pbi.2006.03.014. Zhao, J., Li, C., Zhang, Y., Shen, Y., Hou, J., & Bao, X. (2017). Dynamic control of ERG20
Tippmann, S., Ferreira, R., Siewers, V., Nielsen, J., & Chen, Y. (2017). Effects of expression combined with minimized endogenous downstream metabolism
acetoacetyl-CoA synthase expression on production of farnesene in Saccharomyces contributes to the improvement of geraniol production in Saccharomyces cerevisiae.
cerevisiae. Journal of Industrial Microbiology & Biotechnology, 44(6), 911–922. https:// Microbial Cell Factories, 16(1), 17. https://doi.org/10.1186/s12934-017-0641-9.
doi.org/10.1007/s10295-017-1911-6.

17
Z. Liang et al. Food Research International 147 (2021) 110487

Zhao, Y.-J., & Li, C. (2018). Biosynthesis of plant triterpenoid saponins in microbial cell in Saccharomyces cerevisiae. Enzyme and Microbial Technology, 100, 28–36. https://
factories. Journal of Agricultural and Food Chemistry, 66(46), 12155–12165. https:// doi.org/10.1016/j.enzmictec.2017.02.006.
doi.org/10.1021/acs.jafc.8b04657. Zhuang, X. (2013). Engineering novel terpene production platforms in the yeast
Zhou, F., & Pichersky, E. (2020). More is better: The diversity of terpene metabolism in Saccharomyces cerevisiae ((Ph.D)). Lexington, Kentucky, USA: University of
plants. Current Opinion in Plant Biology, 55, 1–10. https://doi.org/10.1016/j. Kentucky.
pbi.2020.01.005. Zong, Z., Hua, Q., Tong, X., Li, D., Wang, C., Guo, D., & Liu, Z. (2019). Biosynthesis of
Zhou, P., Xie, W., Li, A., Wang, F., Yao, Z., Bian, Q., … Ye, L. (2017). Alleviation of nerol from glucose in the metabolic engineered Escherichia coli. Bioresource
metabolic bottleneck by combinatorial engineering enhanced astaxanthin synthesis Technology, 287, 121410. https://doi.org/10.1016/j.biortech.2019.121410.

18

También podría gustarte