You are on page 1of 13

Received: 8 February 2021 | Revised: 20 June 2021 | Accepted: 27 August 2021

DOI: 10.1002/bit.27935

REVIEW

Sortase A: A chemoenzymatic approach for the labeling of cell


surfaces

Poonam Kumari1 | Sujoy Bowmik1 | Sudipto Kumar Paul1 | Bidisha Biswas1 |


1 1
Sanjay K. Banerjee | Upadhyayula Surayanarayana Murty |
Velayutham Ravichandiran2 | Utpal Mohan2

1
Department of Biotechnology, National
Institute of Pharmaceutical Education & Abstract
Research (NIPER), Guwahati, Assam, India
Sortase A, a transpeptidase enzyme is present in many Gram‐positive bacteria and
2
Department of Medicinal Chemistry, National
Institute of Pharmaceutical Education &
helps in the recruitment of the cell surface proteins. Over the last two decades,
Research (NIPER), Kolkata, West Bengal, India Sortase A has become an attractive tool for performing in vivo and in vitro ligations.
Sortase A‐mediated ligation has continuously been used for its specificity, robust-
Correspondence
Utpal Mohan, Department of Medicinal ness, and highly efficient nature. These properties make it a popular choice among
Chemistry, National Institute of protein engineers as well as researchers from different fields. In this review, we give
Pharmaceutical Education & Research
(NIPER), Kolkata, WB 700054, India. an overview of Sortase A‐mediated ligation of various molecules on the cell surfaces,
Email: mohan.utpal@gmail.com which can have diverse applications in interdisciplinary fields.

KEYWORDS
enzyme, ligation, S. aureus, Sortase A

1 | INTRODUCTION chemically modifying the tyrosine, aspartate, or glutamate amino


acids (Hermanson, 2013). With time, other approaches are also been
Cell surface modification is one of the powerful tools in bioengi- discovered, to attain specificity, greater yield, sensitivity, and reaction
neering to conjugate tag/functional molecules on the cell surface rate (Tiefenbrunn & Dawson, 2010). All these methods have their
(Fox & Sarkar, 2014). These modifications are essential to carry out own limitations. Genetic‐based methods are highly selective and ef-
biophysical and structural studies. Protein labeling with synthetic fective. The genetic‐based fluorescent labeling is widely used for the
molecules opens up a new approach to study the specific function of different molecules like proteins, peptide, or even single amino acid
the proteins in the living system, cel–cell interaction as well as the but the use of fluorescent proteins sometimes lead to a conforma-
interaction of cells with extracellular environment (Abbina et al., tional change of the target protein and large size of these fluorescent
2017; George et al., 2004). Cell surface modification is quite a chal- proteins may alter the functionality of the target proteins (Zacharias
lenging task due to the presence of side‐chain groups present in the et al., 2002). Therefore, among site‐specific modifications, enzyme‐
surface proteins. The prerequisites of cell surface labeling are the based methods are robust, highly efficient and the major advantage
specificity and versatility, which aids in manipulating cell surfaces. of enzyme‐based methods is that it requires mild conditions (Zhang
Wide array of methods are available for the cell surface labeling et al., 2018). Different classes of enzymes have been used, among
such as chemical‐based, genetic‐based, and enzyme‐based (Sahoo, which Sortase A is a transpeptidase enzyme present in Gram‐positive
2012; Sijbrandij et al., 2013; Wu et al., 2009). Chemical‐based bacteria that helps in the recruitment of cell surface proteins
methods are the oldest and most used, exploited both covalent as (Mazmanian et al., 1999; Ton‐That et al., 1999). It is a valuable bio-
well as noncovalent approaches. Most common methods to func- chemical reagent as it is able to ligate the molecules together in vitro
tionalize proteins utilize either lysine or cysteine amino acid reside (Jacobitz et al., 2017). Sortase A is a robust enzyme that is com-
with thiol or amine derivates such as N‐hydroxysuccinimidyl ester or mercially available and can be produced in moderate yields (>40 mg/
maleimides. In addition, other methods have been developed by L) (Nuijens et al., 2019). The Sortagging reaction has been used for

Biotechnol Bioeng. 2021;118:4577–4589. wileyonlinelibrary.com/journal/bit © 2021 Wiley Periodicals LLC | 4577


10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4578 | KUMARI ET AL.

different applications from protein cyclization to cell surface mod- NHS (N‐hydroxyl‐succinimidyl ester), maleimide and pyridyldithiol
ification. The easy availability of the enzyme makes the approach (Cabric et al., 2007; Digilio et al., 2010; Huang et al., 2015; Lee et al.,
quite a popular tool in chemical biology. 2006, 2007; Torres & Gait, 2012). The NHS uses the exposed amine
In this review, we discuss the role of Sortase A in cell surface group, maleimide‐conjugated molecules using exposed thiol group
modifications. We discuss the Sortase A‐based cell surface labeling in while pyridyldithiol attaches through free thiol. Covalent modification
both prokaryotic and eukaryotic cells. We also try to discuss the with a higher degree may cause significant alteration on the basis of
advantages and shortcomings of the various methods. physiology, membrane mobility, and diffusion kinetics of the re-
engineered cells (Paulick et al., 2007; Rabuka et al., 2008; Teramura &
Iwata, 2010).
2 | C E L L S U R F A C E MO D I F I C A T I O N S Electrostatic interactions use negatively charged cell surface and
positively charged polymers to modify the cell surface structure. The
main advantage of the electrostatic interactions is the protection of
Cell surface modifications are one of the significant tools to be used in the cells from sheer stress and immune response while biocompat-
biomedical engineering. The cell surface modifications can be done by ibility is a major concern to be used in therapeutics (Lee et al., 2018).
various strategies, which are mainly divided into three categories de- The other main strategy in the chemical‐based modifications is the
pending on the different approaches: (a) genetic‐based modifications, (b) hydrophobic insertions and provides noninvasive modifications. Dif-
chemical‐based modifications, and (c) enzyme‐based modifications. ferent lipophilic dyes (Dil, DiD, DiR, and DiO) that are currently
available in the market modifies cell surfaces through hydrophobic
interactions. Polyethylene glycols (PEGs) and polyvinyl alcohol (PVA)
2.1 | Genetic‐based modifications are two amphiphilic polymers used in cell surface modifications. PEGs
interact with the lipid bilayer and also been used for surface plasmon
Genetic‐based modifications are well established and one of the resonance (SPR) spectroscopy (Yamamoto et al., 2016). The ad-
highly versatile methods for the cell surface modifications. The basic vantage of hydrophobic insertion over other chemical‐based mod-
step in genetic engineering is the insertion of exogenous genetic ifications is less toxicity and the ability of modified cells to resume
material in the cell to express or suppress the expression of a specific their normal activity (Carlos et al., 2009; Fujita et al., 2008; Miura
cell surface molecule (Kellam et al., 2003; Zhao et al., 2010). Many et al., 2006). The lipid‐conjugated biomolecules' fate is not fully un-
studies were done that focused on the expression of receptors in- derstood and it requires further investigation (Inui et al., 2010).
volved in the cell surface recruitment and migration, to study the In covalent conjugation, synthetic glycans are also used for the
ligand–receptor interaction, incorporated molecules to label the cells, modification of cell surfaces. Cell surface display of synthetic glyco-
and many more (Howarth et al., 2005; Kato & Mrksich, 2004; Penn & lipids has been utilized to investigate the impact of specific glycan
Mangi, 2008). Even though genetic engineering is quite a popular tool structures on cell differentiation and behavior. Pulsipher et al. gener-
but still there are few challenges related to regulatory and safety ated synthetic glycolipids and used liposomal delivery strategy, to
issues (Custódio & Mano, 2016). The other issues associated with incorporate these glycolipids onto neuronal cell surfaces (Pulsipher
genetic engineering approaches are the use of vectors for gene de- et al., 2014). Huang et al. conjugated heparin sulfate glycans to a
livery. Viral vectors have higher transfection efficiency but they are phospholipid tail (Huang et al., 2014), to control the differentiation of
associated with immunogenic response and may lead to tumorigen- embryonic stem cells. In another example, Paszek et al. generated
esis. While other nonviral vectors have low transfection efficiency polymers modified with glycan structures found in natural mucin O‐
(Liu et al., 2019), it leads to permanent modifications in cells, which glycans (Paszek et al., 2014). The utility of synthetic glycolipids has
limits its applicability. Thus, other methods including chemical‐based been limited by their rapid removal from the cell surface by inter-
and enzyme‐based methods have been introduced. nalization. The ability to generate long‐lived synthetic glycolipids will
expand the future applications of synthetic glycolipid engineering
(Woods et al., 2015).
2.2 | Chemical‐based modifications

Chemical‐based cell surface modification is the oldest and one of the 2.3 | Enzyme‐based modifications
most used approaches. Various synthetic and natural molecules are at-
tached to the cell surface mainly by covalent bonding, hydrophobic in- Enzymatic labeling techniques in living cells have gained attention
teractions, and electrostatic interactions (Figure 1). Covalent attachment due to their high specificity, expeditious, mild conditions, efficient
of chemical probes has dominance over noncovalent binding in terms of reaction catalyzing “site‐specific” protein modification, and have
handling, efficiency, and robustness (Sahoo, 2012). higher supremacy over nonenzymatic methods (Sahoo, 2012) (-
Covalent conjugation is one of the straightforward approaches Figure 2). Different classes of peptidases, transferases, ligases, and
that uses the exposed functional groups of proteins for grafting of oxidoreductases are the major ones used in protein labeling. Among
various chemical entities. The commonly used crosslinkers are the all the classes, few are quite popular which are described in Table 1.
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
KUMARI ET AL. | 4579

F I G U R E 1 Chemical labeling strategies for cell surface modification. Covalent conjugation uses N‐hydroxyl‐succinimidyl ester, maleimide
and pyridyldithiol for cell surface engineering. Electrostatic interactions using cationic and anionic polymers to allow modification by creating
different layers. Hydrophobic interactions uses polyethylene glycols and polyvinyl alcohol conjugates with alkyl tails embedded within cell
surface through hydrophobic interaction

In peptidase class, Sortase A and subtiligase are the most common Microbial transglutaminase, farnesyltransferases, N‐
example. Another peptidase enzyme, subtiligase is the engineered en- myristoytransferase, and phosphopantetheinyl transferase are the well‐
zyme derived from subtilisin present in the Bacillus amyloliquefaciens known enzymes in the transferases class. Microbial transglutaminase has
(Wells & Estell, 1988). It catalyzes the reaction between acyl donor the ability to catalyze the reaction between γ‐carboxamide groups of the
peptide ester to the N‐terminal α‐amine of the acceptor of the peptide on glutamine residues and ε‐amino group of lysine and forms an amide bond.
the N‐terminus. The major difference between subtiligase and subtilisin is It is extracted from Streptomyces mobaraensis. The enzyme activity is also
the change in its active site. S221 in the subtilisin is changed to cysteine in affected by the residues, which surround lysine. So, it is not able to
the engineered enzyme that leads to shifts in the reaction from hydrolysis modify all glutamine or lysine residues (Spolaore et al., 2012). The ability
to aminolysis (Abrahmsen et al., 1991). Even though it has ligase activity of the enzyme to modify the intrinsic residues is an advantage but be-
but still excess of acyl acceptor fragment is required to suppress the cause of its broad specificity, it is difficult to predict whether protein is
hydrolysis reaction, therefore, affecting the efficacy (Nuijenset al. 2019). modified or not (Zhang et al., 2018). Farnesyltransferase, another
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4580 | KUMARI ET AL.

F I G U R E 2 Systemic representation of
enzyme‐based labeling for cell surface
modifications. Enzymes recognize specific
substrate sequence and assist in the ligating
molecules onto the cell surface

transferase recognizes CaaX motif and transfers an isoprenoid from its Among all the enzymes, Sortase A, microbial transglutaminase,
substate farnesyl diphosphate. In CaaX, C stands for cysteine, a stands for and lipoic acids are the ones to perform labeling at any site, which are
small aliphatic group and X decides the substrate specificity toward other the major advantages of these three enzymes when compared to
enzymes in the family (Jiang et al., 2018). As the enzyme uses only four others. But as we already discussed, in the case of microbial trans-
amino acids as its motif, it helps in minimizing the potential disruption of glutaminase its activity is hindered by the surrounding residues to the
target protein structure. The other enzyme from the transferase is N‐ modification site. As it can modify glutamine or lysine residue, non-
myristoyltransferase, important for the lipid‐proteins cotranslationally/ specific modifications might be a problem. While lipoic acid ligase has
post‐translationally modifications in eukaryotes (Beld et al., 2014). It a recognition sequence of around 13 amino acids, which might dis-
transfers the myristate from myristoyl‐CoA to the glycine at the N‐ rupt the structure of the protein, Sortase A has a small recognition
terminal and aids in catalyzing an amide bond. Phosphopantethyeinyl motif composed of only five amino acids.
transferase catalyzes the reaction by transferring the Ppant (phospho-
pantetheine) group to PCP (peptiyl) or acyl carrier protein domain (Da
Silva Freitas et al., 2013). The enzyme is quite versatile in its applications 3 | SORTASE A OF GRAM‐ P O S I T I V E
but the major disadvantage is its solubility (Zhang et al., 2018). BAC TERIA
Ligases are the family that has the potential to catalyze the reaction
by fusing two large molecules. Tubulin tyrosine ligase belongs to this Sortase A (SrtA) is a member of the Sortases family, which aids in the
family and helps in the tyrosine addition to the C‐terminal of tubulin by covalent recruitment of cell surface proteins on Gram‐positive bac-
catalyzing peptide bond using ATP (Ersfeld et al., 1993). Lipoic acid ligases teria. In 1990, the Schneewind group has discovered the activity of
identify Lp1A acceptor peptide (LAP) and catalyze the reaction by at- Sortase enzymes. The same group also isolated the Sortase A enzyme
tachment of lipoate to lysine amino acid in LAP. While biotin ligase in 1999 (Ton‐That et al., 2000). After the discovery, the Sortase family
conjugates the reaction by attachment of biotin onto protein using ATP. was vigorously studied. Sortase family has a total of six members
All these enzymes catalyze protein modifications site‐specifically. Lipoic based on their recognition motif as well as their substrates and clas-
acid ligase has also opened new possibilities as it can perform modifica- sified as A–F (Bradshaw et al., 2015; Clancy et al., 2010; Spirig et al.,
tions at the intrinsic sites making the enzyme‐based modifications at- 2011). Sortase A (SrtA) is the intensively studied and best known
tractive tool in protein engineering. among the Sortases family. In the early years of Sortase A discovery,
Oxidoreductases family is also one of the predominant family researchers realized that it could be an alternative target for anti-
uses in the protein labeling. The formylglycine generating enzyme bacterial therapy because it helps in the attachment of virulence
was first discovered by the Figura group while studying sulfatase proteins on the cell surface of Gram‐positive bacteria (Mandlik et al.,
deficiency‐related diseases (Dierks et al., 2003). It performs the 2008; Ton‐That et al., 2000). Inhibition of Sortase A will not create
modification of sulfatases type I by oxidation and hydrolysis of thiol pressure on bacteria as it is not essential for the growth of bacteria
group of cysteine. Recombinant enzymes of the class were expressed (Guo et al., 2015). In 2004, Mao et al. demonstrated that SrtA has the
in Escherichia coli. Formylglycine generating enzymes‐mediated pro- capability to perform in vitro protein ligation, which is more robust,
tein modifications have been developed as a commercial platform site‐specific, and reliable (Mao et al., 2004). The group performed the
called as SMARTag technology by Redwood Biosciences/Catalent. peptide–peptide, protein–peptide, and protein–protein ligation. After
Among all these enzymes, Sortase A, N‐myristoyltransferase, phos- this study, the SrtA application in biomolecular engineering was ex-
phopantetheinyl transferases, and formylglycine generating enzymes tensively studied in the following years (Li et al., 2017; Reed et al.,
are able to perform protein modification in E. coli (Zhang et al., 2018). 2020; Xu & Moyle, 2020).
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
KUMARI ET AL. | 4581

Staphylococcus aureus Sortase A (SaSrtA) has eight stranded β‐


barrel sheets with hydrophobic pockets for the substrate binding.

Grunewald et al. (2015); Pippig et al. (2014); Sunbul


Apart from SaSrtA, structure of other SrtA from Staphylococcus

Weeks and Wells (2018); Henager et al. (2016)

Ho and Tirrell (2016); Kulkarni et al. (2013)


pyogenes, Bacillus anthracis, Streptococcus agalactiae, and Strepto-

Dennler et al. (2014); Mero et al. (2016)


Pasqual et al., 2018; Wu et al. (2017)

Sueda et al. (2011); Kim et al. (2016)


Plaks et al. (2015); Gray et al. (2016)

Drake et al. (2014); Wu et al. (2009)


coccus mutans also has been reported (Khare et al., 2011; Race et al.,

Schumacher et al. (2017)


2009; Wallock‐Richards et al., 2015; Weiner et al., 2010). A common

Zhang et al. (2015)


characteristic of SrtA enzyme is the presence of short helix present

et al. (2008)
References

within β6/β7loop, which contacts the sorting signal LPXTG, upon


substrate binding (Chan et al., 2015; Suree et al., 2009).
SrtA recognize its substrate by LPXTG motif (X‐ any amino acid) and
performs the housekeeping role (Figure 3). Cell surface proteins are an-
chored by two‐step transpeptidation reaction (Bradshaw et al., 2015;
Clancy et al., 2010). First, the SrtA recognizes its motif and cysteine
residue in the active site nucleophilically attacks and cleaves between
threonine and glycine. Due to this, a thioacyl bond is established between
LPET‐SrtA. In the second step, the N‐terminal of oligoglycine present at
the lipid II of the cell‐wall nucleophilically attacks the thioacyl inter-
mediate, thus a peptide bond is formed between the carboxylic acid
Labeling terminal

N or C‐terminus

N or C‐terminus

group of threonine present in LPXT and the amino group of oligoglycine.


N‐terminus

N‐terminus
C‐terminus

C‐terminus

C‐terminus
Any site

Any site

Any site

By this mechanism, it recruits various cell surface proteins on the cell wall.
The main advantage of the SrtA is its ability to perform reactions in
Different enzymatic labeling methods with their recognition sequence, substrate and labeling terminal

nonnatural environment, its specificity toward its motif as well as it has


less impact on protein folding and functionality (Dai et al., 2019). But the
Tyrosine analogues Functionalized‐glycine

major pitfall of SrtA is its low catalytic efficiency, reversible nature of SrtA
reaction, and its Ca2+ dependence. Even though the low catalytic effi-
Lysine peptides, Primary amines

ciency and dependence on Ca2+ was partially resolved by the develop-


Myristicacid analogues

Lipoic acid analogues


Isoprenoid analogues
Glutamine Peptide

ment of SrtA variants having 140 times higher activity and variant, which
Biotin analogues
Peptide esters

Coderivatives

are Ca2+ independent (Antos et al., 2017). The reversibility of the reaction
Substrate

G(n)

can be resolved by using the excess of one substrate and shifting the

equilibrium by removing the by‐products G‐XX (Antos et al., 2016;


Schmohl & Schwarzer, 2014). The latest strategy to tackle the reversibility
issue was resolved by the Antos group. They used metal‐coordinating
peptides. LPXTGGH is used instead of LPXTG as a recognition motif for
Sortase A. After cleaving by Sortase A, released GGH shows a higher
affinity towards Ni+2, therefore, deactivates the GGH peptide (Reed
Tub tag (VDSVEGEGEEEGEE)
Qtag (LLQG) Ktag (MKHKGS)

et al., 2020).
Recognition Sequence

GLNDIFEAQKIEWHE

wWhile performing the Sortase A‐based ligation, the accessi-


GFEIDKVWYDLDA
GDSLSWLLRLLN

bility of protein‐reactive motif to the enzyme is essential. Accessing


GNEASYPL

CXPXR
LPXTG

CaaX

the solvent availability NMR and protein crystal structure will be


useful in assessing the success of sortagging. In the case of a steric


interface, a flexible linker can be used. A flexible linker can be kept
between the protein of interest and Sortase A reactive motif (Antos
et al., 2009).
Formylglycine generating enzyme
Phosphopantetheinyl transferase

4 | SORTASE A MEDIATED CELL SURFACE


Microbial transglutaminase

MODIFICA TION
N‐Myristoyltransferase

Tubulin tyrosine ligase


Farnesyltransferase

Lipoic acid ligase

Sortase, a special transpeptidase present in Gram‐positive bacteria


attaches many secreted proteins to the cell wall. The enzyme syn-
Biotin ligase
Subtiligase
TABLE 1

Sortase A

thesized recombinant protein with higher yield and stability. The


Enzyme

unique mechanism of Sortase A has opened a new avenue in protein


engineering, like protein–protein bioconjugation, circularization of
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4582 | KUMARI ET AL.

F I G U R E 3 Sortase A‐mediated recruitment of cell surface proteins on cell wall of Gram‐positive bacteria. Sortase A recognizes LPXTG motif
present at the C‐terminal of the cell surface protein. It forms thio‐acyl bond with the enzyme and the intermediate is resolved by the amine
which act as a nucleophile from the lipid II pentaglycine

FIGURE 4 Redecorated the cell surface of Staphylococcus aureus using small molecules having LPXTG motif via Sortase A (Nelson
et al., 2010)

proteins, peptide–nucleic acid hybrids preparation, labeling of the cell phase‐dependent manner, which is peaked at the late stationary phase.
surface, protein immobilization on a solid surface. They also found that incorporation can be inhibited by hydroxylamine
For several years, researchers are successfully using Sortase A for (Maňásková et al., 2014). In a different study to further extend the
decorating various living cell surfaces with LPXTG‐tagged small che- efficiency of Sortase‐mediated ligation, they have introduced three
mical probes, recombinant proteins and peptides, and polymers. types of modifications in SrtA substrate. The presence of positively
Nelson et al. documented that the cell surface of S. aureus can be charged amino acid at C‐terminus increases the incorporation rate 20
engineered using its endogenous Sortase A with a chemical or fluor- folds, whereas methionine in place of aspartic acid increases in-
escence probe (Figure 4). They have incubated LPXTG tagged biotin corporation threefolds. Same incorporation efficiency was observed
with wild type S. aureus, resulting in covalent attachment of biotiny- after addition of glycopeptide vancomycin with LPXTG motif, which
lated probe on the cell surface, which was confirmed by MALDI‐TOF occurs at 500 folds low substrate concentration (Maňásková et al.,
mass spectrum (Nelson et al., 2010). In another example, Manaskova 2016). In search of an environmentally friendly feedstock for microbial
et al. modified the carboxy terminus of LPXTG with an amide group as fermentation, Willson et al. have engineered Clostridium acetobutylicum
it may perturb the catalytic function of Sortase A. They have en- as a consolidated bioprocessing organism so that it can hydrolyze
gineered S. aureus cell wall with FITC tagged LPXTG amide and re- lignocellulose and ferment the released sugar. By sortagging, they have
ported that the incorporation of exogenous substrate of SrtA is attached a cellulosome so that it could remain in close vicinity to the
negatively correlated with SrtA mRNA expression and in a growth released sugars (Willson et al., 2016).
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
KUMARI ET AL. | 4583

Apart from bacteria, researchers also tried to engineer cell sur- RBC by CRISPR/Cas9 system. With SrtA around 9000 payloads (self‐
face protein of mammalian cells using different suitable recombinant antigen) were attached to Kell‐LPETGG RBCs. This strategy shows
proteins, fluorescent, or chemical probes. Popp et al. reported positive results in curing multiple sclerosis and type I diabetes in a
mammalian cell surface labeling with the chemical probe by sortag- mouse model (Pishesha et al., 2017). With the help of SrtA‐mediated
ging. LPETG tagged CD40L was expressed on human embryonic transpeptidation, Tan et al. monitored intracellular trafficking of cell
kidney (HEK) 293T cells. Cells were labeled with a biotinylated surface receptors on a real‐time basis. By using the same labeling
pentaglycine probe by incubating in a sortase‐containing serum method, the pH‐dependent dynamic distribution of OGR1 receptor
medium. In this same study, they have also engineered influenza A/ on the cell surface was visualized from the cell surface to various
WSN/33 NA with biotin (Popp et al., 2007). In a similar study, Tanaka intracellular compartments (Tan et al., 2018).
et al. has chosen osteoclast differentiation factor (ODF), a type II Research in different labs further extend the scope of sortase‐
membrane protein as their model cell surface protein. They have mediated ligation, where they have engineered the virus capsid
transfected C terminus LPETG tagged ODF construct into HEK 293T protein. To observe the influenza virus budding in real time, Popp
cell. The selective attachment of triglycine conjugated biotin and et al. labeled two membrane proteins, hemagglutinin (HA) and
EGFP on the cell surface occurs in the presence of 30 µM SrtA. The neuraminidase (NA), with biotin and Alexa Fluor 647 using extra-
labeling was confirmed by western blot using SAv‐HRP and anti‐ cellular sortase. This study enables to monitor birth of new viruses in
FLAG antibody (Tanaka et al., 2008). Nagamune and coworkers infected cells (Popp et al., 2012). To make biological tools capable of
showed that not only on C terminal, N terminal of cell surface protein displaying a wide range of synthetic protein, Hess et al. engineered
can also be labeled with SrtA. They have expressed a transmembrane M13 bacteriophage capsid protein pIII, pVIII, pIX with large molecules
protein cyan fluorescence protein (CFP) on HEK 293T cell surface like GFP and biotin with higher efficiency by using sortase A. They
and subsequently tagged with Alexa Fluor 647 by SrtA. They ana- have also shown that capsid proteins of the same bacteriophage can
lyzed the labeling efficiency by using immunofluorescence staining display two or more different moieties (Hess et al., 2012). In a similar
(Yamamoto & Nagamune, 2009). approach, Schoonen et al. have experimentally proved that proteins,
To attach the labeling probe, cell surface protein has to be fluorophore, and biotin can be attached selectively in cowpea
modified with recognition motif, which may hamper the functional chlorotic mottle virus (CCMV) capsid protein by sortagging, which is
activity of the protein. To overcome this problem, Park et al. de- capable to encapsulate large cargos compared to VLPs from other
monstrated a one‐step method for labeling living cells without the viruses (Schoonen et al., 2015). All the cell surface modifications have
modification of membrane proteins. They have expressed opt‐ been described in Table 2.
SrtAΔ59 containing a signal peptide from platelet‐derived growth Wide varieties of substrates have been used by the Sortase A
factor receptor (opt‐SrtAD59–TM) and labeled with EGFP and in all these reports like capsid proteins of M13 bacteriophage,
TAMRA as fluorescent proteins and chemical dyes respectively (Park capsid of CCMV, mammalian cell surface proteins, glycoproteins,
et al., 2013). and many more. Using the HA protein (hemagglutination), the la-
Sortagging can also be a useful tool for modulating cell–cell in- beling efficiency was found to be 70%–80% while 68 ± 9% label-
teraction. In their study, Tomita et al. directly incorporated a PEGy- ing efficiency was seen after 3 h at 37°C for pIII M13 capsid
lated triglycine on murine EL4 thymoma cell line (E.G7 cells). Next, protein (Hess et al., 2012; Popp et al., 2012). While using CCMV
they optimized the concentration of sortase (12 µM) and substrate capsid protein, the labeling efficiency was around 50%–60%
(50 µM) for maximum ligation efficiency followed by displaying Fc (Schoonen et al., 2015). In the case of mammalian cells, the re-
domains of mouse IgG1 and IgG2a (Fc1‐LPETG and Fc2a‐LPETG) by searchers evaluated the time for the labeling reaction. Park et al.,
sortagging, which was confirmed by CLSM and FACS analysis after group has found that 15 min is sufficient to monitor the labeling
immunostaining. Tagged cells were cocultured with the dendritic cells (Park et al., 2013). Other groups used the ODF and suggested that
at 37°C. Results showed a display of Fc2a‐LPETG increases the labeling could be monitored by 5 min incubation. They also
phagocytosis rate by fourfold (Tomita et al., 2013). Swee et al. in- checked the labeling effect on the viability of cells and did not find
corporated a single domain antibody on the surface of the activated any effect. Therefore, this technique can be used in the future for
CD8 T cell by sortagging and redirected its cytotoxic activity speci- receptor‐trafficking and pulse‐chase labeling (Tanaka et al., 2008).
fically toward Class II MHC positive B cell. The cell surface of the The labeling onto the cell surface has been done using en-
parasite Toxoplasma gondi can also be labeled with a mouse Class II dogenous Sortase A as well as exogenous Sortase A. As most of the
MHC‐specific (VHH7) single‐domain antibody, which resulted in a gram‐positive bacteria have their own Sortase A (endogenous Sor-
significant increase of B cell infection and subsequent lysis by this tase A) on its cell wall, they do not require the enzyme/s from the
parasite in comparison to non‐B cells (Swee et al., 2015). SML has outside. As the presence of Sortase A has been limited to only gram‐
been exploited to overcome the risk related to the immune‐ positive cells, the expressed and purified Sortase A (exogenous Sor-
suppressive treatment of the autoimmune disorder. To suppress tase A) can be used for modifying different surfaces and cells in which
autoimmunity for a self‐antigen, Pishesha et al. made use of the the enzyme is absent. Although the expression and purification of the
natural route of the destruction of RBC by phagocytic cells present in enzyme hardly take 1–2 days, it will slightly affect the time for the
the spleen. The LPETGG tag was introduced on the Kell protein of overall reaction.
| 4584

TABLE 2 Cell surface modification of prokaryotic and eukaryotic cells using Sortase A
Cells Molecule Sortase used Work done Reference

Staphylococcus aureus Fluorescein, biotin and azide Endogenous First example of bacterial cell surface modification Nelson et al. (2010)

Staphylococcus aureus Chemically modified FITC LPETG‐ amide Endogenous Studied the effect of modified LPXTG domain and SrtA mRNA Manaskova et al. (2014;
expression on exogenous incorporation of chemical probe 2016)

Clostridium acetobutylicum Cellulosome Endogenous Engineered C. acetobutylicum to a consolidated bioprocessing Willson et al. (2016)
organism

Staphylococcus aureus and Peptides Endogenous Have seen the recruitment of peptide onto the cell surface of Kumari et al. (2020)
Enterococcus faecalis Gram‐positive bacteria leads to decrease in biofilm formation

HEK 293T cells Biotin, Alexa Fluor 647 and Osteoclast differentiate factor Exogenous Exploited sortase mediated ligation for labeling of cell surface Tanaka et al. (2008)
(ODF) labeled with Biotin, FLAG, HA tag, EGFP in protein in both carboxy and amino terminal.
different experiment

Hela cell EGFP Endogenous Demonstrated a method of cell surface labeling without attaching Park et al. (2013)
recognition motif for sortase A on surface protein.

Murine EL4 thymoma Fc region of mouse IgG1 and IgG2a Exogenous Used sortagging to study cell‐cell interaction. Tomita et al. (2013)
cell (E.G7)

Mouse hematopoietic cells, Biotin probe in all. Single domain antibodies at T cell surface Exogenous Redirected immunity towards a specific cell Swee et al. (2015)
yeast cells, 293 T
cells and Toxoplasma gondii

Human and mouse RBCs Peptides from disease‐relevant autoantigens like Exogenous Illustrated SML of self‐antigen on cell surface as an alternative of Pishesha et al. (2017)
MOG, OVA immune suppressive treatment
and insulin‐derived peptide

Leukocytes Hemagglutinin (HA) Exogenous Monitored intracellular trafficking of cell surface receptor on real Tan et al. (2018)
time basis.

Influenza A/WSN/33 strain Alexa Fluor 647, Biotin Exogenous Exemplify a sortase based ligation strategy to observe growth of Popp et al. (2012)
influenza virus.

M13 bacteriophage Biotin, GFP Exogenous Sortagging can be harnessed to engineer bacteriophage capsid Hess et al. (2012)
protein for higher labeling efficiency and to make them as a
CCMV viral capsid protein GFP, FITC Exogenous Schoonen et al. (2015)
tool to carry large molecules.
KUMARI
ET AL.

10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
KUMARI ET AL. | 4585

5 | C ONC LUS I ON We expect that endogenous and exogenous Sortase A will be


widely used to perform epitope‐grafting on bacterial cell surfaces as
Various strategies have been developed and optimized for cell sur- well as cancer cells to reroute the immune system. The ease of this
face modification. Chemical‐based approaches allow the ease in cell method and combination of the sortase‐based labeling with other
surface modification without performing any genetic engineering on labeling methods for a wide array of molecules and surfaces makes
the cells but it suffers from the off‐site modifications. But the de- the Sortase A more promising candidate in the field of protein en-
velopment of affinity‐based labeling performs the reaction in a con- gineering. We envisage that the field of bioconjugation using Sortase
trolled manner (Tsukiji & Hamachi, 2014; Wang et al., 2011). As an A will further evolve and will contribute immensely to biomaterial
alternative to chemical‐based techniques, researchers are quite in- synthesis.
terested in enzymatic‐based labeling because of the requirement of
mild conditions. Among these, Sortase A is quite a popular and well‐ ACKNOWLEDGME NT
studied enzyme for cell surface modifications. Sortase A has been We thank NIPER Guwahati and NIPER Kolkata for their constant support.
exploited since 2004 and researchers have found diverse applications
in different fields. The review gives useful insight on the Sortase A‐ CONFLIC T OF INTERESTS
mediated cell surface labeling. Sortase A is not only used for the The authors declare there are no conflict of interests.
prokaryotic cells but has also been used for the modification of
mammalian cell surfaces, which makes it an interesting choice. A UT H O R C O N T R I B U TI O N
Sortase A‐based labeling can be used for both proteins and un- The manuscript was written through the contribution of all the
natural moieties. Thus, we can play with a diverse number of mole- authors.
cules. Sortase A cannot only perform the reaction in solution but it
can also catalyze the reaction even when immobilized on the solid DATA AVAILABILITY STATEMENT
support (Steinhagen et al., 2013). They can perform modifications on Data sharing is not applicable to this article as no new data were
N‐terminal (Antos et al., 2009; Williamson et al., 2012), C‐terminal created or analysed in this study.
(Antos et al., 2008; Popp et al., 2007; Popp & Ploegh, 2011), internal
loop regions (Guimaraes et al., 2011; Popp et al., 2009), which pro- ORC I D
vides an additional advantage over other labeling methods. Time is Utpal Mohan http://orcid.org/0000-0002-0256-7305
also an important parameter for this labeling. Despite being an or-
thogonal reaction, the cell surface labeling time of 45 min is enough RE F ER EN CES
for robust labeling (Antos et al., 2009). Even though Sortase A has its Abbina, S., Siren, E. M., Moon, H., & Kizhakkedathu, J. N. (2017). Surface
own limitations such as reaction reversibility and low catalytic ac- engineering for cell‐based therapies: Techniques for manipulating
mammalian cell surfaces. ACS Biomaterials Science & Engineering,
tivity, different genetically engineered Sortase A are available to
4(11), 3658–3677. https://doi.org/10.1021/acsbiomaterials.
tackle its low catalytic activity. Engineered sortases such as hepta- 7b00514
mutant and pentamutant do not require Ca2+ ions. The other major Abrahmsen, L., Tom, J., Burnier, J., Butcher, K. A., Kossiakoff, A., &
limitation, reversibility of the reaction is tackled by removal of some Wells, J. A. (1991). Engineering subtilisin and its substrates for
efficient ligation of peptide bonds in aqueous solution. Biochemistry,
of the reactants, using the excess of one of the reactants more as well
30(17), 4151–4159. https://doi.org/10.1021/bi00231a007
as use of metal peptide which has a high affinity toward GGG‐ Antos, J. M., Ingram, J., Fang, T., Pishesha, N., Truttmann, M. C., &
fragment (Antos et al., 2017; Schmohl & Schwarzer, 2014). Different Ploegh, H. L. (2017). Site‐specific protein labeling via sortase‐
substrates can be used with Sortase A such as peptides, membrane mediated transpeptidation. Current Protocols in Protein Science, 89(1),
15–3. https://doi.org/10.1002/cpps.38
proteins, soluble proteins, antibodies, bacterial toxins, and many
Antos, J. M., Miller, G. M., Grotenbreg, G. M., & Ploegh, H. L. (2008). Lipid
more (Popp et al., 2009; Popp & Ploegh, 2011; Sinisi et al., 2012; modification of proteins through sortase‐catalyzed transpeptidation.
Witte et al., 2013). Although the accessibility and the flexibility of the Journal of the American Chemical Society, 130(48), 16338–16343.
region that is to be labeled is a crucial factor for successful trans- https://doi.org/10.1021/ja806779e
Antos, J. M., Popp, M. W. L., Ernst, R., Chew, G. L., Spooner, E., &
peptidation. A flexible linker (Gly4Ser repeats) is needed but the exact
Ploegh, H. L. (2009). A straight path to circular proteins. Journal of
length of the linker cannot be determined and has to be determined
Biological Chemistry, 284(23), 16028–16036. https://doi.org/10.
empirically. In most cases, efficient transpeptidation can be achieved 1074/jbc.M901752200
using the above solution but still to some extent the intrinsic struc- Antos, J. M., Truttmann, M. C., & Ploegh, H. L. (2016). Recent advances in
tures of the proteins may inflict limitations. This case might be true sortase‐catalyzed ligation methodology. Current Opinion in Structural
Biology, 38, 111–118. https://doi.org/10.1016/j.sbi.2016.05.021
for labeling the internal part of the proteins, where modifying the
Beld, J., Sonnenschein, E. C., Vickery, C. R., Noel, J. P., & Burkart, M. D.
internal loop cannot be admissible. (2014). The phosphopantetheinyl transferases: Catalysis of a post‐
The labeling can be combined with other methods like genetic‐ translational modification crucial for life. Natural Product Reports,
based to incorporate the LPXTG motif to the proteins. Click chem- 31(1), 61–108. https://doi.org/10.1039/C3NP70054B
Bradshaw, W. J., Davies, A. H., Chambers, C. J., Roberts, A. K.,
istry can also be used for the addition of the LPXTG motif or GGG at
Shone, C. C., & Acharya, K. R. (2015). Molecular features of the
the peptide and unnatural moieties.
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4586 | KUMARI ET AL.

sortase enzyme family. The FEBS Journal, 282(11), 2097–2114. George, N., Pick, H., Vogel, H., Johnsson, N., & Johnsson, K. (2004).
https://doi.org/10.1111/febs.13288 Specific labeling of cell surface proteins with chemically diverse
Cabric, S., Sanchez, J., Lundgren, T., Foss, A., Felldin, M., Källen, R., compounds. Journal of the American Chemical Society, 126(29),
Salmela, K., Tibell, A., Tufveson, G., Larsson, R., Korsgren, O., & 8896–8897. https://doi.org/10.1021/ja048396s
Nilsson, B. (2007). Islet surface heparinization prevents the instant Gray, M. A., Tao, R. N., DePorter, S. M., Spiegel, D. A., &
blood‐mediated inflammatory reaction in islet transplantation. McNaughton, B. R. (2016). A nanobody activation
Diabetes, 56(8), 2008–2015. https://doi.org/10.2337/db07‐0358 immunotherapeutic that selectively destroys HER2‐positive breast
Carlos, A. G., Teramura, Y., & Iwata, H. (2009). Cryopreserved agarose‐ cancer cells. ChemBioChem, 17(2), 155–158. https://doi.org/10.
encapsulated islets as bioartificial pancreas: A feasibility study. 1002/cbic.201500591
Transplantation, 87(1), 29–34. https://doi.org/10.1097/TP. Guimaraes, C. P., Carette, J. E., Varadarajan, M., Antos, J., Popp, M. W.,
0b013e318191b24b Spooner, E., Brummelkamp, T. R., & Ploegh, H. L. (2011).
Chan, A. H., Yi, S. W., Terwilliger, A. L., Maresso, A. W., Jung, M. E., & Identification of host cell factors required for intoxication through
Clubb, R. T. (2015). Structure of the Bacillus anthracis Sortase A use of modified cholera toxin. Journal of Cell Biology, 195(5),
enzyme bound to its sorting signal: A flexible amino‐terminal 751–764. https://doi.org/10.1083/jcb.201108103
appendage modulates substrate access. Journal of Biological Grünewald, J., Klock, H. E., Cellitti, S. E., Bursulaya, B., McMullan, D.,
Chemistry, 290(42), 25461–25474. https://doi.org/10.1074/jbc. Jones, D. H., Chiu, H. P., Wang, X., Patterson, P., Zhou, H., Vance, J.,
M115.670984 Nigoghossian, E., Tong, H., Daniel, D., Mallet, W., Ou, W., Uno, T.,
Clancy, K. W., Melvin, J. A., & McCafferty, D. G. (2010). Sortase Brock, A., Lesley, S. A., & Geierstanger, B. H. (2015). Efficient
transpeptidases: Insights into mechanism, substrate specificity, and preparation of site‐specific antibody–drug conjugates using
inhibition. Peptide Science, 94(4), 385–396. https://doi.org/10.1002/ phosphopantetheinyl transferases. Bioconjugate Chemistry, 26(12),
bip.21472 2554–2562. https://doi.org/10.1021/acs.bioconjchem.5b00558
Custódio, C. A., & Mano, J. F. (2016). Cell surface engineering to control Guo, Y., Cai, S., Gu, G., Guo, Z., & Long, Z. (2015). Recent progress in the
cellular interactions. ChemNanoMat, 2(5), 376–384. https://doi.org/ development of sortase A inhibitors as novel anti‐bacterial virulence
10.1002/cnma.201600047 agents. RSC Advances, 5(62), 49880–49889. https://doi.org/10.
Dai, X., Böker, A., & Glebe, U. (2019). Broadening the scope of sortagging. 1039/C8RA06705H
RSC Advances, 9(9), 4700–4721. https://doi.org/10.1039/ Henager, S. H., Chu, N., Chen, Z., Bolduc, D., Dempsey, D. R., Hwang, Y.,
C8RA06705H Wells, J., & Cole, P. A. (2016). Enzyme‐catalyzed expressed protein
Da Silva Freitas, D., Mero, A., & Pasut, G. (2013). Chemical and enzymatic ligation. Nature Methods, 13(11), 925–927. https://doi.org/10.1038/
site specific PEGylation of hGH. Bioconjugate Chemistry, 24(3), nmeth.4004
456–463. https://doi.org/10.1021/bc300594y Hermanson, G. T. (2013). Bioconjugate techniques (3rd ed.). Academic
Dennler, P., Chiotellis, A., Fischer, E., Brégeon, D., Belmant, C., Press. https://doi.org/10/1016/C2009‐0‐64240‐9
Gauthier, L., Lhospice, F., Romagne, F., & Schibli, R. (2014). Hess, G. T., Cragnolini, J. J., Popp, M. W., Allen, M. A., Dougan, S. K.,
Transglutaminase‐based chemo‐enzymatic conjugation approach Spooner, E., Ploegh, H. L., Belcher, A. M., & Guimaraes, C. P. (2012).
yields homogeneous antibody–drug conjugates. Bioconjugate M13 bacteriophage display framework that allows sortase‐mediated
Chemistry, 25(3), 569–578. https://doi.org/10.1021/bc400574z modification of surface‐accessible phage proteins. Bioconjugate
Dierks, T., Schmidt, B., Borissenko, L. V., Peng, J., Preusser, A., Chemistry, 23(7), 1478–1487. https://doi.org/10.1021/bc300130z
Mariappan, M., & von Figura, K. (2003). Multiple sulfatase deficiency Ho, S. H., & Tirrell, D. A. (2016). Chemoenzymatic labeling of proteins for
is caused by mutations in the gene encoding the human Cα‐ imaging in bacterial cells. Journal of the American Chemical Society,
formylglycine generating enzyme. Cell, 113(4), 435–444. https://doi. 138(46), 15098–15101. https://doi.org/10.1021/jacs.6b07067
org/10.1016/S0092‐8674(03)00347‐7 Howarth, M., Takao, K., Hayashi, Y., & Ting, A. Y. (2005). Targeting
Digilio, G., Menchise, V., Gianolio, E., Catanzaro, V., Carrera, C., quantum dots to surface proteins in living cells with biotin ligase.
Napolitano, R., Fedeli, F., & Aime, S. (2010). Exofacial protein thiols Proceedings of the National Academy of Sciences, 102(21),
as a route for the internalization of Gd (III)‐based complexes for 7583–7588. https://doi.org/10.1073/pnas.0503125102
magnetic resonance imaging cell labeling. Journal of Medicinal Huang, B., Abraham, W. D., Zheng, Y., López, S. C. B., Luo, S. S., &
Chemistry, 53(13), 4877–4890. https://doi.org/10.1021/jm901876r Irvine, D. J. (2015). Active targeting of chemotherapy to
Drake, P. M., Albers, A. E., Baker, J., Banas, S., Barfield, R. M., Bhat, A. S., disseminated tumors using nanoparticle‐carrying T cells. Science
de Hart, G. W., Garofalo, A. W., Holder, P., Jones, L. C., Kudirka, R., Translational Medicine, 7(291), 291ra94‐291ra94. https://doi.org/10.
McFarland, J., Zmolek, W., & Rabuka, D. (2014). Aldehyde tag 1126/scitranslmed.aaa5447
coupled with HIPS chemistry enables the production of ADCs Huang, M. L., Smith, R. A., Trieger, G. W., & Godula, K. (2014). Glycocalyx
conjugated site‐specifically to different antibody regions with remodeling with proteoglycan mimetics promotes neural
distinct in vivo efficacy and PK outcomes. Bioconjugate Chemistry, specification in embryonic stem cells. Journal of the American
25(7), 1331–1341. https://doi.org/10.1021/bc500189z Chemical Society, 136(30), 10565–10568. https://doi.org/10.1021/
Ersfeld, K., Wehland, J., Plessmann, U., Dodemont, H., Gerke, V., & ja505012a
Weber, K. (1993). Characterization of the tubulin‐tyrosine ligase. Inui, O., Teramura, Y., & Iwata, H. (2010). Retention dynamics of
Journal of Cell Biology, 120(3), 725–732. https://doi.org/10.1083/ amphiphilic polymers PEG‐lipids and PVA‐alkyl on the cell surface.
jcb.120.3.725 ACS Applied Materials & Interfaces, 2(5), 1514–1520. https://doi.org/
Fox, W. M., & Sarkar, D. (2014). Cell surface engineering by chemical 10.1021/am100134v
reaction and remodeling, In Micro‐ and Nanoengineering of the Cell Jacobitz, A. W., Kattke, M. D., Wereszczynski, J., & Clubb, R. T. (2017).
Surface (pp. 27–41). William Andrew Publishing. https://doi.org/10. Sortase transpeptidases: structural biology and catalytic mechanism.
1016/B978‐1‐4557‐3146‐6.00002‐7 Advances in pRotein Chemistry and Structural Biology, 109, 223–264.
Fujita, E., Teramura, Y., Shiraga, T., Yoshioka, S. I., Iwatsubo, T., Kawamura, A., https://doi.org/10.1016/bs.apcsb.2017.04.008
& Kamimura, H. (2008). Pharmacokinetics and tissue distribution of Jiang, H., Zhang, X., Chen, X., Aramsangtienchai, P., Tong, Z., & Lin, H.
tacrolimus (FK506) after a single or repeated ocular instillation in rabbits. (2018). Protein lipidation: Occurrence, mechanisms, biological
Journal of Ocular Pharmacology and Therapeutics, 24(3), 309–319. https:// functions, and enabling technologies. Chemical Reviews, 118(3),
doi.org/10.1089/jop.2007.0083 919–988. https://doi.org/10.1021/acs.chemrev.6b00750
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
KUMARI ET AL. | 4587

Kato, M., & Mrksich, M. (2004). Rewiring cell adhesion. Journal of the Chemistry, 7(42), 6545–6553. https://doi.org/10.1039/
American Chemical Society, 126(21), 6504–6505. https://doi.org/10. C6PY01616B
1021/ja039058e Miura, S., Teramura, Y., & Iwata, H. (2006). Encapsulation of islets with ultra‐
Kellam, B., Paul, A., & Shakesheff, K. M. (2003). Chemical modification of thin polyion complex membrane through poly (ethylene glycol)‐
mammalian cell surfaces. Chemical Society Reviews, 32(6), 327–337. phospholipids anchored to cell membrane. Biomaterials, 27(34),
https://doi.org/10.1039/B211643J 5828–5835. https://doi.org/10.1016/j.biomaterials.2006.07.039
Khare, B., Krishnan, V., Rajashankar, K. R., I‐Hsiu, H., Xin, M., Ton‐That, H., Nelson, J. W., Chamessian, A. G., McEnaney, P. J., Murelli, R. P.,
& Narayana, S. V. (2011). Structural differences between the Kazmiercak, B. I., & Spiegel, D. A. (2010). A biosynthetic strategy for
Streptococcus agalactiae housekeeping and pilus‐specific sortases: re‐engineering the Staphylococcus aureus cell wall with non‐native
SrtA and SrtC1. PLoS One, 6(8), e22995. https://doi.org/10.1371/ small molecules. ACS Chemical Biology, 5(12), 1147–1155. https://
journal.pone.0022995 doi.org/10.1021/cb100195d
Kim, D. I., Jensen, S. C., Noble, K. A., Kc, B., Roux, K. H., Nguyen, H. D., Phan, T. T. P., & Schumann, W. (2011). Analysis and
Motamedchaboki, K., & Roux, K. J. (2016). An improved smaller application of Bacillus subtilis sortases to anchor recombinant
biotin ligase for BioID proximity labeling. Molecular Biology of the proteins on the cell wall. AMB Express, 1(1), 1–11. https://doi.org/
Cell, 27(8), 1188–1196. https://doi.org/10.1091/mbc.E15‐12‐0844 10.1186/2191‐0855‐1‐22
Kulkarni, C., Kinzer‐Ursem, T. L., & Tirrell, D. A. (2013). Selective Nuijens, T., Toplak, A., Schmidt, M., Ricci, A., & Cabri, W. (2019). Natural
functionalization of the protein N Terminus with N‐myristoyl occurring and engineered enzymes for peptide ligation and
transferase for bioconjugation in cell lysate. ChemBioChem, 14(15), cyclization. Frontiers in Chemistry, 7, 829. https://doi.org/10.3389/
1958–1962. https://doi.org/10.1002/cbic.201300453 fchem.2019.00829
Kumari, P., Nath, Y., Murty, U. S., Ravichandiran, V., & Mohan, U. (2020). Ott, W., Nicolaus, T., Gaub, H. E., & Nash, M. A. (2016). Sequence‐
Sortase A mediated bioconjugation of common epitopes decreases independent cloning and post‐translational modification of
biofilm formation in Staphylococcus aureus. Frontiers in Microbiology, repetitive protein polymers through sortase and Sfp‐mediated
11, 1702. https://doi.org/10.3389/fmicb.2020.01702 enzymatic ligation. Biomacromolecules, 17(4), 1330–1338. https://
Lee, D. Y., Cha, B. H., Jung, M., Kim, A. S., Bull, D. A., & Won, Y. W. (2018). doi.org/10.1021/acs.biomac.5b01726
Cell surface engineering and application in cell delivery to heart Park, K., Jung, J., Son, J., Kim, S. H., & Chung, B. H. (2013). Anchoring
diseases. Journal of Biological Engineering, 12(1), 1–11. https://doi. foreign substances on live cell surfaces using Sortase A specific
org/10.1186/s13036‐018‐0123‐6 binding peptide. Chemical Communications, 49(83), 9585–9587.
Lee, D. Y., Lee, S., Nam, J. H., & Byun, Y. (2006). Minimization of https://doi.org/10.1039/c3cc44753g
immunosuppressive therapy after islet transplantation: Combined Pasqual, G., Chudnovskiy, A., Tas, J., Agudelo, M., Schweitzer, L. D.,
action of hemeoxygenase‐1 and PEGylation to islet. American Cui, A., Hacohen, N., & Victora, G. D. (2018). Monitoring T
Journal of Transplantation, 6(8), 1820–1828. https://doi.org/10. cell–dendritic cell interactions in vivo by intercellular enzymatic
1111/j.1600‐6143.2006.01414.x labelling. Nature, 553(7689), 496–500. https://doi.org/10.1038/
Lee, D. Y., Nam, J. H., & Byun, Y. (2007). Functional and histological nature25442
evaluation of transplanted pancreatic islets immunoprotected by Paszek, M. J., DuFort, C. C., Rossier, O., Bainer, R., Mouw, J. K., Godula, K.,
PEGylation and cyclosporine for 1 year. Biomaterials, 28(11), Hudak, J. E., Lakins, J. N., Wijekoon, A. C., Cassereau, L.,
1957–1966. https://doi.org/10.1016/j.biomaterials.2006.12.015 Rubashkin, M. G., Magbanua, M. J., Thorn, K. S., Davidson, M. W.,
Li, K., Zhang, R., Xu, Y., Wu, Z., Li, J., Zhou, X., Jiang, J., Liu, H., & Xiao, R. Rugo, H. S., Park, J. W., Hammer, D. A., Giannone, G., Bertozzi, C. R.,
(2017). Sortase A‐mediated crosslinked short‐chain & Weaver, V. M. (2014). The cancer glycocalyx mechanically primes
dehydrogenases/reductases as novel biocatalysts with improved integrin‐mediated growth and survival. Nature, 511(7509), 319–325.
thermostability and catalytic efficiency. Scientific Reports, 7(1), 1–11. https://doi.org/10.1038/nature13535
https://doi.org/10.1038/s41598‐017‐03168‐z Paulick, M. G., Forstner, M. B., Groves, J. T., & Bertozzi, C. R. (2007). A
Liu, L., He, H., & Liu, J. (2019). Advances on non‐genetic cell membrane chemical approach to unraveling the biological function of the
engineering for biomedical applications. Polymers, 11(12), 2017. glycosylphosphatidylinositol anchor. Proceedings of the National
https://doi.org/10.3390/polym11122017 Academy of Sciences, 104(51), 20332–20337. https://doi.org/10.
Mandlik, A., Swierczynski, A., Das, A., & Ton‐That, H. (2008). Pili in Gram‐ 1073/pnas.0710139104
positive bacteria: Assembly, involvement in colonization and biofilm Penn, M. S., & Mangi, A. A. (2008). Genetic enhancement of stem cell
development. Trends in Microbiology, 16(1), 33–40. https://doi.org/ engraftment, survival, and efficacy. Circulation Research, 102(12),
10.1016/j.tim.2007.10.010 1471–1482. https://doi.org/10.1161/CIRCRESAHA.108.175174
Mao, H., Hart, S. A., Schink, A., & Pollok, B. A. (2004). Sortase‐mediated Pippig, D. A., Baumann, F., Strackharn, M., Aschenbrenner, D., &
protein ligation: A new method for protein engineering. Journal of Gaub, H. E. (2014). Protein–DNA chimeras for nano assembly. ACS
the American Chemical Society, 126(9), 2670–2671. https://doi.org/ Nano, 8(7), 6551–6555. https://doi.org/10.1021/nn501644w
10.1021/acs.bioconjchem.0c00486 Pishesha, N., Bilate, A. M., Wibowo, M. C., Huang, N. J., Li, Z.,
Maňásková, S. H., Nazmi, K., Belkum, A., Bikker, F. J., Wamel, W. J., & Deshycka, R., Bousbaine, D., Li, H., Patterson, H. C., Dougan, S. K.,
Veerman, E. C. (2014). Synthetic LPETG‐containing peptide Maruyama, T., Lodish, H. F., & Ploegh, H. L. (2017). Engineered
incorporation in the Staphylococcus aureus cell‐wall in a sortase A‐ erythrocytes covalently linked to antigenic peptides can protect
and growth phase‐dependent manner. PLoS One, 9(2), e89260. against autoimmune disease. Proceedings of the National Academy of
https://doi.org/10.1371/journal.pone.0089260 Sciences, 114(12), 3157–3162.
Maňásková, S. H., Nazmi, K., 't Hof, W., Belkum, A., Martin, N. I., Plaks, J. G., Falatach, R., Kastantin, M., Berberich, J. A., & Kaar, J. L. (2015).
Bikker, F. J., Wamel, W. J., & Veerman, E. C. (2016). Staphylococcus Multisite clickable modification of proteins using lipoic acid ligase.
aureus sortase A‐mediated incorporation of peptides: Effect of Bioconjugate Chemistry, 26(6), 1104–1112. https://doi.org/10.1021/
peptide modification on incorporation. PLoS One, 11(1), e0147401. acs.bioconjchem.5b00161
https://doi.org/10.1371/journal.pone.0147401 Popp, M. W., Antos, J. M., Grotenbreg, G. M., Spooner, E., & Ploegh, H. L.
Mero, A., Grigoletto, A., Maso, K., Yoshioka, H., Rosato, A., & Pasut, G. (2007). Sortagging: A versatile method for protein labeling. Nature
(2016). Site‐selective enzymatic chemistry for polymer conjugation Chemical Biology, 3(11), 707–708. https://doi.org/10.1038/
to protein lysine residues: PEGylation of G‐CSF at lysine‐41. Polymer nchembio.2007.31
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4588 | KUMARI ET AL.

Popp, M. W. L., Antos, J. M., & Ploegh, H. L. (2009). Site‐specific protein Sueda, S., Yoneda, S., & Hayashi, H. (2011). Site‐specific labeling of
labeling via sortase‐mediated transpeptidation. Current Protocols in proteins by using biotin protein ligase conjugated with fluorophores.
Protein Science, 56(1), 15–3. https://doi.org/10.1002/0471140864. ChemBioChem, 12(9), 1367–1375. https://doi.org/10.1002/cbic.
ps1503s56 201000738
Popp, M. W. L., Karssemeijer, R. A., & Ploegh, H. L. (2012). Sunbul, M., Yen, M., Zou, Y., & Yin, J. (2008). Enzyme catalyzed site‐
Chemoenzymatic site‐specific labeling of influenza glycoproteins as specific protein labeling and cell imaging with quantum dots.
a tool to observe virus budding in real time. PLoSPathog, 8(3), Chemical Communications, 45, 5927–5929. https://doi.org/10.
e1002604. https://doi.org/10.1371/journal.ppat.1002604 1039/B812162A
Popp, M. W. L., & Ploegh, H. L. (2011). Making and breaking peptide Suree, N., Liew, C. K., Villareal, V. A., Thieu, W., Fadeev, E. A.,
bonds: Protein engineering using sortase. AngewandteChemie Clemens, J. J., Jung, M. E., & Clubb, R. T. (2009). The structure of the
International Edition, 50(22), 5024–5032. https://doi.org/10.1002/ Staphylococcus aureus sortase‐substrate complex reveals how the
anie.201008267 universally conserved LPXTG sorting signal is recognized. Journal of
Pulsipher, A., Griffin, M. E., Stone, S. E., Brown, J. M., & Hsieh‐Wilson, L. C. Biological Chemistry, 284(36), 24465–24477. https://doi.org/10.
(2014). Directing neuronal signaling through cell‐surface glycan 1074/jbc.M109.022624
engineering. Journal of the American Chemical Society, 136(19), Swee, L. K., Lourido, S., Bell, G. W., Ingram, J. R., & Ploegh, H. L. (2015).
6794–6797. https://doi.org/10.1021/ja5005174 One‐step enzymatic modification of the cell surface redirects cellular
Rabuka, D., Forstner, M. B., Groves, J. T., & Bertozzi, C. R. (2008). cytotoxicity and parasite tropism. ACS Chemical Biology, 10(2),
Noncovalent cell surface engineering: Incorporation of bioactive 460–465. https://doi.org/10.1021/cb500462t
synthetic glycopolymers into cellular membranes. Journal of the Tan, M., Yamaguchi, S., Nakamura, M., & Nagamune, T. (2018). Real‐time
American Chemical Society, 130(18), 5947–5953. https://doi.org/10. monitoring of pH‐dependent intracellular trafficking of ovarian
1021/ja710644g cancer G protein‐coupled receptor 1 in living leukocytes. Journal of
Race, P. R., Bentley, M. L., Melvin, J. A., Crow, A., Hughes, R. K., Bioscience and Bioengineering, 126(3), 363–370. https://doi.org/10.
Smith, W. D., Sessions, R. B., Kehoe, M. A., McCafferty, D. G., & 1016/j.jbiosc.2018.03.012
Banfield, M. J. (2009). Crystal structure of Streptococcus pyogenes Tanaka, T., Yamamoto, T., Tsukiji, S., & Nagamune, T. (2008). Site‐specific
sortase A. Journal of Biological Chemistry, 284(11), 6924–6933. protein modification on living cells catalyzed by sortase.
https://doi.org/10.1074/jbc.M805406200 ChemBioChem, 9(5), 802–807. https://doi.org/10.1002/cbic.
Reed, S. A., Brzovic, D. A., Takasaki, S. S., Boyko, K. V., & Antos, J. M. 200700614
(2020). Efficient sortase‐mediated ligation using a common c‐ Teramura, Y., & Iwata, H. (2010). Cell surface modification with polymers
terminal fusion tag. Bioconjugate Chemistry, 31(5), 1463–1473. for biomedical studies. Soft Matter, 6(6), 1081–1091. https://doi.
https://doi.org/10.1021/acs.bioconjchem.0c00156 org/10.1039/B913621E
Sahoo, H. (2012). Fluorescent labeling techniques in biomolecules: A Tiefenbrunn, T. K., & Dawson, P. E. (2010). Chemoselective ligation
flashback. RSC Advances, 2(18), 7017–7029. https://doi.org/10. techniques: Modern applications of time‐honored chemistry. Peptide
1039/C2RA20389H Science, 94(1), 95–106. https://doi.org/10.1002/bip.21337
Schmohl, L., & Schwarzer, D. (2014). Sortase‐mediated ligations for the Tomita, U., Yamaguchi, S., Maeda, Y., Chujo, K., Minamihata, K., &
site‐specific modification of proteins. Current Opinion in Chemical Nagamune, T. (2013). Protein cell‐surface display through in situ
Biology, 22, 122–128. https://doi.org/10.1016/j.cbpa.2014.09.020 enzymatic modification of proteins with a poly (Ethylene glycol)‐
Schoonen, L., Pille, J., Borrmann, A., Nolte, R. J., & van Hest, J. C. (2015). lipid. Biotechnology and Bioengineering, 110(10), 2785–2789. https://
Sortase A‐mediated N‐terminal modification of cowpea chlorotic doi.org/10.1002/bit.24933
mottle virus for highly efficient cargo loading. Bioconjugate Ton‐That, H., Mazmanian, S. K., Faull, K. F., & Schneewind, O. (2000).
Chemistry, 26(12), 2429–2434. https://doi.org/10.1021/bc300130z Anchoring of surface proteins to the cell wall of Staphylococcus aureus:
Schumacher, D., Lemke, O., Helma, J., Gerszonowicz, L., Waller, V., Sortase catalyzed in vitro transpeptidation reaction using LPXTG peptide
Stoschek, T., Durkin, P. M., Budisa, M., Leonhardt, H., Keller, B. G., and NH2‐Gly3 substrates. Journal of Biological Chemistry, 275(13),
Hackenberge, C. P. R. (2017). Broad substrate tolerance of tubulin 9876–9881. https://doi.org/10.1074/jbc.275.13.9876
tyrosine ligase enables one‐step site‐specific enzymatic protein Torres, A. G., & Gait, M. J. (2012). Exploiting cell surface thiols to enhance
labeling. Chemical Science, 8(5), 3471–3478. https://doi.org/10. cellular uptake. Trends in Biotechnology, 30(4), 185–190. https://doi.
1039/B812162A org/10.1016/j.tibtech.2011.12.002
Sijbrandij, T., Cukkemane, N., Nazmi, K., Veerman, E. C., & Bikker, F. J. Tsukiji, S., & Hamachi, I. (2014). Ligand‐directed tosyl chemistry for in situ
(2013). Sortase A as a tool to functionalize surfaces. Bioconjugate native protein labeling and engineering in living systems: From basic
Chemistry, 24(5), 828–831. https://doi.org/10.1021/bc4000146 properties to applications. Current Opinion in Chemical Biology, 21,
Sinisi, A., Popp, M. W., Antos, J. M., Pansegrau, W., Savino, S., Nissum, M., 136–143. https://doi.org/10.1016/j.cbpa.2014.07.012
Rappuoli, R., Ploegh, H. L., & Buti, L. (2012). Development of an Wallock‐Richards, D. J., Marles‐Wright, J., Clarke, D. J., Maitra, A.,
influenza virus protein array using sortagging technology. Dodds, M., Hanley, B., & Campopiano, D. J. (2015). Molecular basis
Bioconjugate Chemistry, 23(6), 1119–1126. https://doi.org/10. of Streptococcus mutans sortase A inhibition by the flavonoid natural
1021/bc200577u product trans‐chalcone. Chemical Communications, 51(52),
Spirig, T., Weiner, E. M., & Clubb, R. T. (2011). Sortase enzymes in Gram‐ 10483–10485. https://doi.org/10.1039/C5CC01816A
positive bacteria. Molecular Microbiology, 82(5), 1044–1059. https:// Wang, H., Koshi, Y., Minato, D., Nonaka, H., Kiyonaka, S., Mori, Y.,
doi.org/10.1111/j.1365‐2958.2011.07887.x Tsukiji, S., & Hamachi, I. (2011). Chemical cell‐surface receptor
Spolaore, B., Raboni, S., Ramos Molina, A., Satwekar, A., Damiano, N., & engineering using affinity‐guided, multivalent organocatalysts.
Fontana, A. (2012). Local unfolding is required for the site‐specific Journal of the American Chemical Society, 133(31), 12220–12228.
protein modification by transglutaminase. Biochemistry, 51(43), https://doi.org/10.1021/ja204422r
8679–8689. https://doi.org/10.1021/bi301005z Weeks, A. M., & Wells, J. A. (2018). Engineering peptide ligase specificity
Steinhagen, M., Zunker, K., Nordsieck, K., & Beck‐Sickinger, A. G. (2013). by proteomic identification of ligation sites. Nature Chemical Biology,
Large scale modification of biomolecules using immobilized sortase 14(1), 50–57. https://doi.org/10.1038/nchembio.2521
A from Staphylococcus aureus. Bioorganic & Medicinal Chemistry, Weiner, E. M., Robson, S., Marohn, M., & Clubb, R. T. (2010). The Sortase
21(12), 3504–3510. https://doi.org/10.1016/j.bmc.2013.03.039 A enzyme that attaches proteins to the cell wall of Bacillus anthracis
10970290, 2021, 12, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/bit.27935 by Universite Paris-Saclay, Wiley Online Library on [04/09/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
KUMARI ET AL. | 4589

contains an unusual active site architecture. Journal of Biological recombinant protein antigens. ChemPlusChem, 85(1), 227–236.
Chemistry, 285(30), 23433–23443. https://doi.org/10.1074/jbc. https://doi.org/10.1002/cplu.201900687
M110.135434 Yamamoto, T., & Nagamune, T. (2009). Expansion of the sortase‐mediated
Wells, J. A., & Estell, D. A. (1988). Subtilisin—an enzyme designed to be labeling method for site‐specific N‐terminal labeling of cell surface
engineered. Trends in Biochemical Sciences, 13(8), 291–297. https:// proteins on living cells. Chemical Communications, 9, 1022–1024.
doi.org/10.1016/0968‐0004(88)90121‐1 https://doi.org/10.1039/b818792d
Williamson, D. J., Fascione, M. A., Webb, M. E., & Turnbull, W. B. (2012). Yamamoto, T., Teramura, Y., Itagaki, T., Arima, Y., & Iwata, H. (2016).
Efficient N‐terminal labeling of proteins by use of sortase. Interaction of poly (ethylene glycol)‐conjugated phospholipids with
AngewandteChemie, 124(37), 9511–9514. https://doi.org/10.1002/ supported lipid membranes and their influence on protein
anie.201204538 adsorption. Science and Technology of Advanced Materials, 17(1),
Willson, B. J., Kovács, K., Wilding‐Steele, T., Markus, R., Winzer, K., & 677–684. https://doi.org/10.1080/14686996.2016.1240006
Minton, N. P. (2016). Production of a functional cell wall‐anchored Zacharias, D. A., Violin, J. D., Newton, A. C., & Tsien, R. Y. (2002).
minicellulosome by recombinant Clostridium acetobutylicum ATCC Partitioning of lipid‐modified monomeric GFPs into membrane
824. Biotechnology for Biofuels, 9(1), 1–22. https://doi.org/10.1186/ microdomains of live cells. Science, 296(5569), 913–916. https://
s13068‐016‐0526‐x doi.org/10.1126/science.1068539
Witte, M. D., Theile, C. S., Wu, T., Guimaraes, C. P., Blom, A. E., & Zhang, Y., Blanden, M. J., Sudheer, C., Gangopadhyay, S. A., Rashidian, M.,
Ploegh, H. L. (2013). Production of unnaturally linked chimeric Hougland, J. L., & Distefano, M. D. (2015). Simultaneous site‐specific
proteins using a combination of sortase‐catalyzed transpeptidation dual protein labeling using protein prenyltransferases. Bioconjugate
and click chemistry. Nature Protocols, 8(9), 1808–1819. https://doi. Chemistry, 26(12), 2542–2553. https://doi.org/10.1021/acs.
org/10.1038/nprot.2013.103 bioconjchem.5b00553
Woods, E. C., Yee, N. A., Shen, J., & Bertozzi, C. R. (2015). Glycocalyx Zhang, Y., Park, K. Y., Suazo, K. F., & Distefano, M. D. (2018). Recent
engineering with a recycling glycopolymer that increases cell progress in enzymatic protein labelling techniques and their
survival. in vivo. AngewandteChemie International Edition, 54(52), applications. Chemical Society Reviews, 47(24), 9106–9136. https://
15782–15788. https://doi.org/10.1002/anie.201508783 doi.org/10.1039/C8CS00537K
Wu, P., Shui, W., Carlson, B. L., Hu, N., Rabuka, D., Lee, J., & Bertozzi, C. R. Zhao, W., Teo, G. S. L., Kumar, N., & Karp, J. M. (2010). Chemistry and
(2009). Site‐specific chemical modification of recombinant proteins material science at the cell surface. Materials Today, 13(4), 14–21.
produced in mammalian cells by using the genetically encoded https://doi.org/10.1016/S1369‐7021(10)70056‐0
aldehyde tag. Proceedings of the National Academy of Sciences,
106(9), 3000–3005. https://doi.org/10.1073/pnas.0807820106
Wu, Q., Ploegh, H. L., & Truttmann, M. C. (2017). Hepta‐mutant
Staphylococcus aureus sortase A (SrtA7m) as a tool for in vivo How to cite this article: Kumari, P., Bowmik, S., Paul, S. K.,
protein labeling in Caenorhabditis elegans. ACS Chemical Biology, Biswas, B., Banerjee, S. K., Murty, U. S., Ravichandiran, V., &
12(3), 664–673. https://doi.org/10.1021/acschembio.6b00998
Mohan, U. (2021). Sortase A: A chemoenzymatic approach for
Xu, Z., & Moyle, P. M. (2020). A self‐adjuvanting vaccine platform:
Optimization of site‐specific sortasea mediated conjugation of toll‐ the labeling of cell surfaces. Biotechnology and Bioengineering,
like receptor 2 ligands onto the carboxyl or amino terminus of 118, 4577–4589. https://doi.org/10.1002/bit.27935

You might also like