Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

REvIEWS

Carbohydrate-​active enzymes
(CAZymes) in the gut microbiome
Jacob F. Wardman 1,2,4, Rajneesh K. Bains2,3,4, Peter Rahfeld2,3 and
Stephen G. Withers1,2,3 ✉
Abstract | The 1013–1014 microorganisms present in the human gut (collectively known as the human
gut microbiota) dedicate substantial percentages of their genomes to the degradation and uptake
of carbohydrates, indicating the importance of this class of molecules. Carbohydrates function
not only as a carbon source for these bacteria but also as a means of attachment to the host, and
a barrier to infection of the host. In this Review, we focus on the diversity of carbohydrate-​active
enzymes (CAZymes), how gut microorganisms use them for carbohydrate degradation, the
different chemical mechanisms of these CAZymes and the roles that these microorganisms and
their CAZymes have in human health and disease. We also highlight examples of how enzymes
from this treasure trove have been used in manipulation of the microbiota for improved health
and treatment of disease, in remodelling the glycans on biopharmaceuticals and in the potential
production of universal O-​type donor blood.

Short-​chain fatty acids


Humans have an intimate relationship with their micro­ on by excessive anaerobic fermentation of dietary oli­
Fatty acids containing up to six biota. The community of 1013–1014 bacteria in our gut1 is gosaccharides present in legumes, brassicas and dairy
carbon atoms (predominantly well known for its many roles in human health and phys­ products by gas-​producing bacteria of the colon10.
acetate, propionate and iology. These roles range from immune system develop­ The enzymes that act on carbohydrates — carbohydrate-
butyrate in the human gut)
ment2 and nutrient acquisition3 to drug metabolism4 and active enzymes (CAZymes) — operate on a diverse range
that can be produced by
gut microorganisms during modulation of host behaviour5. Accordingly, there has of substrates. Several distinct enzyme classes and asso­
fermentation. been increased interest in understanding and manipu­ ciated modules are included in the CAZy database and
lating the gut microbiota. The global market for human the associated CAZypedia11, as summarized in Fig. 1.
microbiota therapies, profiling and diagnostics was esti­ CAZymes involved in carbohydrate degradation include
mated to be around US $275–400 million in 2019 and is the glycoside hydrolases, polysaccharide lyases and carbo­
expected to rise to US $750–1,500 million by 2024 (ref.6). hydrate esterases12–14. Additionally, auxiliary activity
Carbohydrates have an important role in shaping the enzymes perform various redox transformations, which
microbiota. The gut is a competitive environment for in many cases are degradative in nature. The classes of
microorganisms, and the ability to degrade carbohy­ enzymes, and the chemical mechanisms used, are highly
1
Department of Biochemistry drates (whether found in food or derived from the host dependent upon the structure of the carbohydrate sub­
and Molecular Biology, or other members of the human gut microbiota) provides strate to be cleaved. A further limitation imposed by
University of British a competitive advantage to certain bacteria3. Microbial the anaerobic environment of the gut and the oxygen
Columbia, Vancouver,
British Columbia, Canada.
carbohydrate-​degrading activities provide substantial depen­dence of many auxiliary activity enzymes is that
benefits to the host as well. Humans lack most of the auxiliary activity enzymes do not seem to be widely
2
Michael Smith Laboratories,
University of British
enzymes necessary to degrade dietary carbohydrates7. encoded in the human gut microbiome11,15, although
Columbia, Vancouver, Instead, we outsource these enzymatic needs to our res­ exceptions do exist16. Meanwhile, the primary enzymes
British Columbia, Canada. ident gut bacteria3,7. Although the bacteria degrade these involved in carbo­hydrate synthesis are classified as gly­
3
Department of Chemistry, carbohydrates to fuel their own needs, in return the host cosyltransferases17. Often, CAZymes are appended with
University of British receives energy, typically up to 10% of the bodily energy carbohydrate-binding modules (CBMs), which have
Columbia, Vancouver,
budget8, in the form of short-​chain fatty acids. Short-​chain an important role in enhancing catalytic activity of the
British Columbia, Canada.
fatty acids also have anti-​inflammatory activities, can appended CAZyme but contain no catalytic activity of
4
These authors contributed
equally: Jacob F. Wardman,
modulate epigenetic remodelling and influence host their own18.
Rajneesh K. Bains. metabolism9. For the most part, this interplay between Because the evolution and acquisition of particular
✉e-​mail: withers@chem.ubc.ca carbohydrates, the microbiota and human health pro­ CAZymes provide certain bacteria with competitive
https://doi.org/10.1038/ ceeds smoothly and quietly, but occasionally it becomes advantages, bacteria of the human gut dedicate large
s41579-022-00712-1 evident. An example of this being the flatulence brought proportions of their genomes towards CAZymes7,19.

NATURe RevIeWS | Microbiology

0123456789();:
Reviews

Glycoside hydrolases Glycosyltransferases

O O
O O OR1 + R2 R2
OR + H2O OH R1
ROH Donor Acceptor

R = Monosaccharide, oligosacccharide,
polysaccharide or aglycone R1 (donor) R2 (acceptor)
Exo-glycoside
hydrolase
Reducing end O
Nucleoside Glycan assembly
O O O O O O P O
O O O O O OH or lipid

O
n
Non-reducing end
Endo-glycoside hydrolase n = 1 or 2 Protein glycosylation

O
Polysaccharide lyases GlcNAc Man
NH
O


Glycolipids OH
O
O–
O
O
RO O O Carbohydrate binding modules
OR′ OR′
ROH
CBM Associated with CAZymes

R′
Directs CAZymes

O2C

O2C

O2C to substrate
O O O O O
O O OH
O O O O
O O O O O O
O O O O O OH

R
Point of cleavage CBM binds to specific
carbohydrate motif

Carbohydrate esterases Auxiliary activities


OH O Various redox
XCOR XH OH Galactose OH transformations
O O O oxidase
X = NH or O OH
O
OH
catalysed by
HO HO
H2O RCOOH O2 H2O2
auxiliary activity
OH OH enzymes

Fig. 1 | overview of the cAZyme classes and associated modules. among the most frequently encountered substrates of polysaccharide
Glycoside hydrolases catalyse hydrolysis of glycosidic linkages within a lyases in the human gut microbiota. Carbohydrate-​binding modules
wide range of carbohydrate substrates and can be classified as either (CBMs) are domains without catalytic activity that are often appended
exo-​glycoside hydrolases or endo-​glycoside hydrolases depending to carbohydrate-​active enzymes (CAZymes). CBMs bind a range of
upon where they cleave within the carbohydrate substrate. Glycosyl­ carbohydrate motifs of varying sizes and compositions and aid in directing
transferases form glycosidic linkages using activated donors to transfer the associated CAZyme to the substrate. Carbohydrate esterases catalyse
sugars onto specific acceptors such as proteins, lipids or other glycans. de-​esterification of various carbohydrate substrates. Auxiliary activity
Polysaccharide lyases utilize a distinct mechanism to break down enzymes encompass a large class of redox active enzymes that function
polysaccharides containing uronic acids. Glycos­a minoglycans are upon carbohydrates.

Glycoside hydrolases are the largest and best-​studied class highest densities of glycoside hydrolases per kilobase out
of CAZymes responsible for carbohydrate degradation of all environments on earth21. Accordingly, gut residents
and will be the primary focus of this Review11. Despite such as Bacteroides thetaiotaomicron and Bacteroides
their vast substrate scope, diverse 3D structures and evo­ ovatus dedicate remarkable proportions of their genomes
lutionary origins, the majority of glycoside hydrolases to glycoside hydrolases and polysaccharide lyases (~6%
use one of the two chemical mechanisms that were first of all genes encoded, whereas most organisms dedi­
proposed by Koshland almost 70 years ago based on con­ cate 1–3% of their genes to CAZymes)7,19. In particular,
temporary understandings of acid-​catalysed glycoside CAZymes involved in the degradation of dietary poly­
hydrolysis20 (Box 1). However, there are some intriguing saccharides and human glycans are likely to be especially
adaptations of these mechanisms to match peculiarities of abundant7. Using this as a resource, enzymes involved
substrate structures, many of which are described later7,13. in the preparation of high-​value biopharmaceuticals22,23,
The sheer abundance and diversity of the CAZymes and even blood type remodelling24,25, have been identified
within the gut microbiome thus makes the gut an appeal­ from the gut microbiome.
ing starting point in CAZyme discovery for a range of This Review will focus on essentials of the enzymol­
applications7,21. Metagenomic sequencing has revealed ogy of CAZymes, common themes in bacterial carbo­
that the human gut microbiome as a whole has one of the hydrate degradation (with a particular focus on the

www.nature.com/nrmicro

0123456789();:
Reviews

well-​studied Bacteroidetes phylum) and the physiologi­ diet and the microbiome composition and functional­
cal roles of these enzymes in the deconstruction of car­ ities are also present across animal species26,28. As pro­
bohydrates by the human gut microbiota. Moreover, by posed by Freter et al. in 1983, a large but finite number
discussing the chemical mechanisms of these CAZymes, of ecological niches are available to microorganisms
as well as their applications, we aim to provide a unique in the gut29. One of the methods by which microor­
perspective into the field. Such work will highlight the ganisms establish themselves in the gut is through
importance of connecting our understandings of funda­ evolution of CAZymes capable of degrading specific
mental glycobiology of the human gut microbiota with polysaccharides3. Accordingly, different types of bacteria
our knowledge of CAZyme mechanisms to yield further have differing CAZyme contents7. Generally in humans,
advances in medicine and biotechnology. diets richer in plant-​based carbohydrates enrich for
Prevotella spp., whereas Bacteroides spp. are more abun­
Food, drugs and the microbiota dant with diets in which fewer dietary carbohydrates
Diet and the microbiota reach the gut30–34. However, species-​level and strain-​level
Across cultures and dietary habits, microbial compo­ differences in genome content as well as other dietary
sition and the associated consortiums of CAZymes factors also affect microbial abundance in the gut35,36.
within the human gut are specialized towards dietary Introduction of different dietary carbohydrates drives
polysaccharides26,27. Moreover, relationships between changes to the microbiome and the associated CAZyme
composition on various time scales. For example, within
a day of diet modification, small changes in resident
Box 1 | The classical Koshland mechanisms microbial diversity and metabolism are observed, along
The glycoside linkages found in starch and cellulose are among the most stable linkages with the introduction of metabolically active bacteria
found in nature166. Fortunately, nature has done an exquisite job at evolving glycoside from the new food sources35. The Hadza hunter–gatherers
hydrolases to hydrolyse such linkages. In a now seminal paper, Daniel Koshland Jr undergo seasonal changes in their diet as they alternate
proposed that glycoside hydrolases could follow two mechanisms on the basis of whether between a meat-​rich diet in the dry season and a honey
the stereochemistry at the anomeric centre is retained or inverted upon hydrolysis20. and berry-​rich diet during the wet season34. These die­
As shown in the figure (panel a), cleavage of the glycosidic linkage can be achieved via tary changes result in changes in both the microbiome
a two-​step double displacement mechanism with net retention of stereochemistry12,167.
composition (for example, decline of Prevotella spp.
The first step implicates two catalytic residues (typically Asp or Glu) acting in a concerted
manner. One residue acts as the nucleophile, attacking at the anomeric centre, whereas
during the wet season34) and in associated CAZyme
the other donates a proton to the departing leaving group. Next, the acid or base residue abundance (for example, an increased abundance of
deprotonates water as it attacks the glycosyl enzyme and releases the sugar product12,167. animal-​derived sialic acid (Neu5Gc)-​cleaving GH33
Alternatively, glycoside hydrolases can proceed through a single displacement family members during the dry season37). It should
mechanism with inversion of stereochemistry (see the figure, panel b)12,167. One carboxylic be noted that, in comparison, adults in industrialized
acid residue protonates the glycosidic oxygen whereas the other residue assists nations often have relatively stable microbiomes27,38. This
the deprotonation of a water nucleophile for direct displacement of the anomeric stability has been proposed to be driven by a typically
substituent12,167. All steps proceed via oxocarbenium ion-​like transition states. Although consistent (and often fibre-​poor) diet resulting in the
glycoside hydrolases largely follow the mechanisms described above, deviations have extinction of other­wise mutable bacterial populations
been reported, several of which are described in the text13. For interested readers,
accompanied by an overall loss of bacterial diversity27,39.
CAZypedia provides clear and useful descriptions of the mechanisms, their history and
other concepts related to carbohydrate-​active enzymes (CAZymes) and carbohydrates168.
As an adaptation to longer-​term dietary habits, sym­
bionts can also undergo genome-​level changes, an
a interesting example of this phenomenon being the
Catalytic Catalytic Catalytic acquisition of CAZymes involved in the degradation
acid/base acid/base acid/base
of porphyran and agarose (present in algal polysaccha­
O
O

O rides commonly consumed in East Asian cultures) by
O
O ROH
H
OH gut symbionts40–42. These CAZymes were most likely
O H O
O H
O acquired via transposon-​mediated horizontal gene trans­
O OH
R fer from marine bacteria that were probably consumed

O O with the algae40,41.
O O –
O O

Dietary fibres
Catalytic nucleophile Catalytic nucleophile Catalytic nucleophile Gut bacteria have evolved many methods for the degra­
dation of carbohydrates from the diet. These carbohy­
drates — commonly referred to as dietary fibre — are
b Catalytic acid Catalytic acid
generally plant-​derived, escape host degradation and are
O O instead acted on by the human gut microbiota32. Starch
O ROH O–
is one of the best studied fibres in terms of its role in the
H
O O gut microbiota and is the only polysaccharide for which
O
R Homo sapiens produces its own degradative enzymes7.
OH
O H However, depending on the preparation of the food and
H
the properties of the starch, between 2 and 50% of it can

O O HO O escape host degradation and will be passed on to the gut
microbiota43. Numerous different pathways for starch
Catalytic base Catalytic base degradation and uptake in various gut microorganisms

NATURe RevIeWS | Microbiology

0123456789();:
Reviews

Cross-​feeding
have been characterized (recently reviewed by Cerqueira was recently proposed for the GH99 family and supported
A phenomenon in which et al.43) but one of the best known is that of the starch uti­ by elegant mechanistic studies52. Briefly, a carboxylate in
microorganisms utilize lization system (Sus) in B. thetaiotaomicron43. Elucidation the active site deprotonates the mannose 2-​hydroxyl con­
metabolites and degradation of the eight-​gene polysaccharide utilization locus (PUL), comitantly with nucleophilic attack at the anomeric centre
products derived from other
microorganisms.
which itself was the first PUL elucidated, was initiated by by the alcohol, generating a short-​lived 1,2-​anhydro sugar.
Anderson and Salyers in 1989 (refs44,45) (Box 2). This intermediate is then hydrolysed by attack of water at
Outer membrane vesicles The seminal work on Sus provided a general model the anomeric centre with general base catalysis provided
Vesicles with distinct protein for PULs, which has been extended to others through­ by an active-​site glutamate residue52 (Fig. 2). Two surface
cargo that are derived from
out the Bacteroidetes phylum46. This method of degra­ endo-​α-1,6-​mannanases (GH76) perform further degra­
the outer membrane of
Gram-​negative bacteria
dation has been described as ‘selfish’ in some instances dation to produce oligosaccharides that are then trans­
and are released into the as it minimizes the distribution of nutrients to neigh­ ported into the periplasm for further metabolism47. In the
environment. bouring gut microorganisms47. In general, degradation periplasm, the degradation of the imported oligosaccha­
of polysaccharides is initiated by extracellular cleav­ rides is carried out by an array of glycoside hydrolases
age of the polysaccharide and is carried out by surface with varying specificity. Complete degradation of the
endo-​glycoside hydrolases. This produces large oli­ phosphorylated branches requires mannose-6 phos­
gosaccharides that are then rapidly transported to the phatases (BT2630 or BT3783) to remove the 6-​phosphate
periplasm where the oligosaccharide is broken down linkages, thereby allowing further degradation by BT3774
into simple monosaccharides and disaccharides. This (GH38), which is unable to accommodate the charged
process effectively excludes other bacteria from taking 6-​phosphate linkage in the –1 subsite.
in any released carbohydrates47. In other examples, the It must be noted that not all carbohydrate degradation
degradation products will not be rapidly transported in the human gut microbiota is selfish. In fact, complex
into the periplasm and can instead be shared with other carbohydrate cross-​feeding networks seem to be com­
bacteria as described below48–51. mon in the microbiota. In many instances, co-​culturing
The selfish model was first noted by Cuskin et al. for the of bacteria that initiate carbohydrate degradation (often
degradation of yeast α-​mannan by B. thetaiotaomicron47. referred to as keystone degraders) can enable growth of
Yeast-​containing fermented foods have been part of strains that would otherwise not be able to utilize the
human diets for centuries; thus, it is of little wonder that carbohydrate (secondary degraders)48–51. In numerous
gut bacteria such as B. thetaiotaomicron have evolved cases, it seems that secondary degraders just lack the sur­
ways to digest the yeast cell wall47. α-​Mannan degradation face endo-​glycoside hydrolases that are responsible for
is initiated by BT3862, a surface endo-​α-1,2-​mannosidase initial cleavage, as exogenous addition of these enzymes
(GH99) responsible for liberating Man-​α-1,3-​Man or integration of the required genes enables carbohydrate
disaccharides47 (Fig. 2). A novel substrate-​assisted catalytic utilization50,51,53. In a notable example of cooperation
mechanism proceeding through an epoxide intermediate through cross-​feeding, B. ovatus produces extracellu­
lar inulin-​degrading enzymes despite not utilizing the
degradation products (and, in fact, under certain cir­
Box 2 | Susc and SusD: defining polysaccharide utilization loci cumstances exhibiting a loss of fitness upon production
Polysaccharide utilization loci (PUL) are genomic loci that encode the carbohydrate-​
of these glycoside hydrolases)49. Rather, the production
active enzymes (CAZymes), glycan binding proteins, transporters, and sensors and of these enzymes seems to aid in growth of secondary
regulators required for binding, cleavage and import of carbohydrates169. Within degraders of inulin such as Bacteroides vulgatus. In turn,
the loci, SusC, an outer membrane TonB-​dependent transporter, and SusD, a surface B. vulgatus provides a competitive edge to B. ovatus
glycan-​binding protein, are of particular interest. Both SusC and SusD are critical to PUL through an as yet unclear mechanism49. Similar rela­
functionality, and removal of either leads to a strong reduction of PUL functionality. Thus, tionships have since been identified between numerous
the presence of these two genes is a canonical hallmark for Gram-​negative PULs. Recent different bacteria54. This network of interactions is fur­
structural characterization has shown that SusC and SusD associate in the membrane. ther expanded by long-​range carbohydrate degradation
In what has been termed the ‘pedal bin’ model (see the figure), SusD acts as a flexible mediated by outer membrane vesicles50,53. Bacteroides spp.
hinged lid over SusC in the absence of substrate, exposing binding sites in SusC170. Once
have been shown to package certain CAZymes into outer
substrate binds inside SusC, the SusD lid closes and substrate is allowed to pass through
SusC into the periplasm in a TonB-​dependent manner170. Recently, the definition of a PUL
membrane vesicles and release them into the extracel­
has been expanded to include similar gene clusters in Gram-​positive bacteria that lack lular milieu — enabling other members of the human
a SusC/D pair. These, along with many other aspects of carbohydrate degradation and gut microbiota to access a communal pool of utilizable
uptake, are discussed in greater detail in the recent review by Briggs et al.54. carbohydrates50,53.
The degradation of carbohydrates of higher complex­
Substrate
ity necessitates larger PULs containing a broader array
of enzyme classes. One of the most complex examples
concerns the degradation of rhamnogalacturonan II
SusD
(RG-​II), a common polysaccharide in pectin55. There
Outer membrane are a total of 21 distinct glycosidic linkages in RG-​II
SusC with numerous ester modifications, pyranose and fura­
nose sugars, as well as l-​sugars and d-​sugars55. B. theta­
iotaomicron carries out near complete degradation
Inner membrane of this carbohydrate (leaving a single glycosidic link­
age uncleaved) using numerous CAZymes contained
TonB across three PULs. Perhaps surprisingly, degradation

www.nature.com/nrmicro

0123456789();:
Reviews

α1,3 α1,3 α1,3


P Phosphate
α1,3 α1,3
α1,3
Man
α1,2
α1,2
α1 α1,3 GlcNAc
α1,3 α1,3
α1,2 6 P
α1,2 α1,3 α1
α1,6
α1,2 α1,2 P
α1,6 α1,2 α1,2 6 α1,3 α1,3
α1,2 α1,2
α1,6
α1,6 α1,3 α1,2
α1,2 α1,2
α1,6 α1,6 α1,2 α1,3 α1,6
Surface endo-α-1,6-
α1,3 α1,6 Surface endo-α-1,2- α1,2 α1,3 mannosidases α1,3
α1,6
β1,4 mannosidases α1,6
BT2623 GH76
BT3862 GH99 α1,3 α1,6 BT3792 GH76 α1,3
β1,4 α1,3
β1,4 α1,3
α1,3 α1
β1,4 P
6
α1,2 α1,2
α1,6 α1,2
α1,6
α1,6

Outer membrane

SusC
Periplasm +SusD
α1,3
Proposed GH99 mechanism
α1,3
α1,2 α1,2 α1,3
Enzyme Enzyme Enzyme α-1,2/3-Mannosidases (GH92)
H α-1,6-Endo-mannanases (GH76) α1,6
α1,6
O O– O O H O O– α-1,2-Mannosidase (GH38) α1
O
HO HO HO OH α-1,6-Mannosidases (GH76 and GH125) 6 P
O +H2O
HO HO OO
HO O α-1,3-Mannosidases (GH92) α1,2
Man Mannose-6-phosphatases
-ROH Man Man α1,6 α1,2

OR α1,3
H OH α1,6
O
O O H O O– O O H
H
Enzyme Enzyme Enzyme

Fig. 2 | Mechanism of α-mannan digestion by B. thetaiotaomicron. Overview of degradation carried out by Bacteroides
thetaiotaomicron first described by Cuskin et al.47. Briefly, several cell surface-​localized carbohydrate-​active enzymes
(CAZymes) initiate degradation of α-​mannan at the outer membrane. SusC/D-​like proteins then import oligosaccharides
into the periplasm. Following transportation into the periplasm, various enzymes carry out complete degradation. For
clarity, enzymes are represented as Pac-​Man shapes, and those drawn attached to the membrane are surface-​associated
lipoproteins. Insert: proposed chemical mechanism by which the glycoside hydrolase GH99 family carries out the first step
in α-​mannan degradation.

is accomplished with a single enzyme specific for each surveying the number of distinct PULs sequenced to
glycosidic linkage cleaved, highlighting the exquisite date59. However, there are additional complexities to this
regioselectivity and stereoselectivity of these enzymes55. ‘one PUL, one glycan’ paradigm. Increasingly complex
Homologues of one of the RG-​II PULs are present in carbohydrates often require additional co-​regulated
several bacteria and likely confer RG-​II metabolism55. gene clusters outside a central PUL, known as CAZyme
Indeed, it seems that the presence of homologous PULs clusters61, in order to be completely degraded62. In other
for many carbohydrates can be used as a relatively reli­ cases, different components of a polysaccharide will be
able indicator of glycan degradation capabilities48,56,57. targeted by different PULs48,57. In such instances, certain
Moreover, the presence of homologous PULs in differ­ organisms can contain only some of these PULs and it
ent bacteria likely provides a level of functional stability is likely that further carbohydrate degradation is only
and resilience to the human gut microbiota, as even if a achieved through cross-​feeding48.
certain glycan degrader is lost, another is able to carry
out the same role58. Host glycan degradation
The presence of a PUL also suggests that there is Mucin O-​glycans. Coating the lumen of the gut, at the
a glycan that is capable of being degraded59. Recently, interface between host and microorganism, is a layer of
Lapébie et al. extended this concept to estimate the total mucus. The primary constituents of this mucosal lin­
number of carbohydrate structures present in nature as ing are a family of proteins known as mucins, which are
being a few thousand59 (in contrast to the theoretical densely glycosylated with O-​glycans, and lesser amounts
1012 structures possible for hexasaccharides as originally of N-​glycans63. From the human perspective, the thick
calculated by Laine60). Lapébie et al. largely did this by mucus layer functions as a barrier against bacterial

NATURe RevIeWS | Microbiology

0123456789();:
Reviews

infection64. From the bacterial perspective, it functions glycopeptides from mucins for subsequent uptake and
as an attachment point to maintain their position in metabolism (Fig. 3b).
the flowing gastrointestinal tract65,66 and as a nutrient
source67–69, especially during times of limited dietary N-​Glycans. Addition of N-​linked glycans is one of the
carbohydrates70,71. Indeed, there is a poorly understood most common post-​translational modifications found
interplay between the microbiota and the host centred on extracellular and cell-​surface proteins86. The ubi­
on the mucus layer such that, in a somewhat paradoxi­ quity of N-​glycans on host epithelial surfaces likely
cal manner, mucus degradation by symbionts such provides a nutritional advantage for microorganisms
as Akkermansia muciniphila and B. thetaiotaomicron that can degrade and metabolize these glycans. Work by
induces host synthesis of mucus and thickens the Briliūtė et al. revealed the extensive enzymatic machin­
mucosal lining72–74. ery that Bacteroides spp. use for the degradation of
The complexity of mucosal carbohydrate structures, complex N-​glycans68. In response to being grown on an
with numerous different monosaccharide components, N-​glycoprotein as a sole carbon source, B. thetaiotao­
glycosidic linkages and modifications, likely is an impor­ micron upregulates 19 CAZymes from different families
tant barrier to bacterial penetration of the mucus71,75,76. across 6 loci. This extensive machinery provides access
Nonetheless, some bacteria do achieve this, typically to many structurally diverse complex N-​glycans68.
through a large repertoire of enzymes spread across As highlighted in Fig. 3c, the initial extracellular
many glycoside hydrolase families67, as well as acces­ degradation of N-​glycans involves an array of glycoside
sory enzymes such as sulfatases77 (Fig. 3a,b). By contrast, hydrolases, including endo-​β-​N-​acetylglucosaminidases
dietary polysaccharides such as starch that evolved to (ENGases), along with an extracellular sialidase and an
be easily degradable are less complex and require fewer endo-​glycoside hydrolase from the newly established
enzymes for coordinated degradation43,71. Even special­ GH163 family68. ENGases, primarily from the GH18 and
ized mucin degraders often target relatively few struc­ GH85 families, are responsible for the initial release of
tures, perhaps to limit the energetic costs of genome the glycan through cleavage within the chitobiose core
maintenance and enzyme expression71. Interestingly, (GlcNAc-​β1–4GlcNAc) of the core sugar sequence87.
the N-​glycans and O-​glycans of mucin share certain ENGases follow a substrate-​assisted mechanism (Fig. 3c),
structural motifs that lend themselves to being broken which departs from the canonical Koshland mecha­
down by the same enzymes, and to induction of the nism described in Box 1. Briefly, the 2-​acetamide acts
same PULs68. Work by Crouch et al. identified GH16 as the catalytic nucleophile to form the key glycosyl
endo-​β-​galactosidases in B. thetaiotaomicron that cleave oxazoline intermediate which is then hydrolysed with
the poly-​N-​acetyllactosamine (Galβ1–4GlcNAcβ1–3) overall net retention of stereochemistry88,89. ENGases
structures, which are common to O-​glycans, N-​glycans are often quite specific. For example, BT3987 from
and keratan sulfate78 (Fig. 3a). These GH16 family mem­ B. thetaiotaomicron cleaves within the chitobiose core
bers can accommodate many of the modifications and of oligomannose-​type N-​glycan but remains inactive
decorations present on these diverse glycan structures. against complex N-​glycans90.
It seems likely that only after initial cleavage of these Degradation of the core tetrasaccharide and other
extended decorations and transport across the outer N-​glycan-​derived oligosaccharides occurs upon trans­
membrane does a consortium of periplasmic enzymes port into the periplasm and cytoplasm. In the periplasm,
then remove these modifications78. an array of CAZymes, sulfatases and esterases further
Recently, there has been increased interest in a degrade the oligosaccharide into monosaccharides and
class of enzymes known as O-​glycoproteases. As the disaccharides, which can then be transported into the
name suggests, these proteases have a strict specific­ cytoplasm47,68. Recent efforts have shed light on cyto­
ity for O-​glycosylated proteins79 with requirements for plasmic degradation of N-​glycan-​derived disaccha­
both the substrate glycan structure and the amino acid rides. For example, in B. thetaiotaomicron, a GH130
sequence79–82. O-​Glycoproteases have varying specifici­ glycoside phosphorylase (BT1033) cleaves the Manβ-1,
ties, with some having higher activities towards mucin 4-GlcNAc disaccharide produced upon degradation of
domains (often referred to as mucinases), whereas others the N-​glycan core tetrasaccharide to generate α-​Man-1-
are active against mucins as well as less densely glyco­ phosphate (α-​Man1P) and GlcNAc91. This strategy,
sylated proteins79,80,82. Recent structural characteriza­ which seems widespread amongst Bacteroidetes, enables
tion by Pluvinage et al. has shown how the multidomain α-​Man1P to enter glycolysis while avoiding the expendi­
structure of the O-​glycoprotease ZmpB from Clostridium ture of ATP through use of a hexokinase91. An alternative
perfringens is specialized for mucin degradation83. In strategy, applied by Actinobacteria in various environ­
particular, the CBMs of ZmpB appear to be spaced such ments including the gut, utilizes a GH5 (subfamily 18)
that they would almost perfectly match the periodicity to hydrolyse the same linkage92,93.
of heavily glycosylated domains of mucin (which them­
selves have only recently been observed84) while placing Glycosaminoglycans. Epithelial cell-​derived glycosamino­
the catalytic domain near the more exposed regions of glycans represent a reliable source of complex carbohy­
the mucin substrate83. O-​Glycoproteases have also been drates for the human gut microbiota (Fig. 3d). PULHep from
identified in PULs and likely have a role in nutrient B. thetaiotaomicron encodes several genes required for the
acquisition85. In such cases, cleavage of mucin at the degradation of heparan sulfate, along with the structur­
cell surface is carried out by an O-​glycoprotease rather ally related heparin. PULHep contains four polysaccharide
than an endo-​g lycoside hydrolase, producing large lyases, a sulfamidase and a GH88 Δ4,5-​glucuronidase in

www.nature.com/nrmicro

0123456789();:
Reviews

a α1,2
c d
Gal Neu5Ac GlcN
β1,3 6S
α1,3 β1,4 α2,6 α2,6
β1,4 β1,3 α1 Man GalNAc GlcA
β1,4 β1,4
6S β1,3 Fuc GlcNAc IdoA
α2,3 β1,4 β1,4 β1,3 β1,4 β1,3 α1 α1,3 α1,6
β1,2
4,5-Uronic acid
β1,2
α2,3
Sialidase (GH33) β1,3
β1,4
Endo-galactosidase (GH16) α1 α1,3 α1,6 Sialidase (GH33) 6S 2S 6S 6S
α1,4 α1,4 α1,4 β1,4 α1,4 β1,4 α1,4 β1,4
6S β1,4
α1,3 Endo-glucosaminidases
α2,3 β1,3 β1,4 β1,6 β1,4 (GH18) NS NS
Endo-GlcNAcase α2,6
α1,2 β1,4 (GH163) β1,4 Polysaccharide lyase
β1,3
α1,3 β1,4 β1,3
α1,6 (PL12)
β1,4
6S β1,4 β1
β1,3 β1,4
Protein 6S 2S 6S
β1,4 α1,4 α1,4

SusC
+SusD Periplasm
α1,2 SusC
α1,3 β1,4 β1,3 +SusD Polysaccharide lyase
β1,3 6S β1,4 β1,3
(PL12 + PL13 + PL15)
Glucuronyl β1,4
Exo-hexosaminidases α1,3
α1,6
(GH20) hydrolase (GH88)
b α2,3 β1,3 Sulfatases 6S 2S 6S
α1 Sialidase (GH33) β1,4 β1,4 α1,4 α1,4
α1,3 α1,6
α2,3 β1,3 α1
Galactosidase (GH2)
β1,3 α1 β1,4 Fucosidase (GH29)
α2,6 Polysaccharide lyase mechanism
α2,3 β1,4 β1,3 α1 β1,4
A NG A NG
α1 β1,4
β1,3 Mucinase
β1,4 H
α2,3 H
β1,3 α1
O O –

O –
O O
Substrate-assisted mechanism (GH18, GH20)
O
Enzyme Enzyme Enzyme O O O
O
OR OR
O O H O O– O OH H
B– HB
+H2O H
O O O
OR O OH
-ROH –
NG
N O H HN O A
N O
H
α2,3 β1,3 α1 O –
O O
– – OH
O O H O O O O
α2,3
β1,3
β1,3 O
α1
β1,3 α1 Enzyme HB
Enzyme Enzyme
OR

e HN
O HN
O HN O
N –
O2C
O– O H N –
O 2C
O CO2– H N O–
O O
H O O OH
O H OR O
O
O H2O OH
OR CO2– ROH
O H2N
H2N
H2N

Fig. 3 | Host glycan degradation by bacteroidetes. a,b | Glycoside hydrolase GH16 family endo-​galactosidases (part a)
or mucinases (part b) can initiate mucin digestion by generating large oligosaccharides or glycopeptides for uptake and
further depolymerization. c | An array of carbohydrate-​active enzymes (CAZymes) from Bacteroides thetaiotaomicron
catalyse the break down of complex N-​glycans (top). Substrate-​assisted mechanism used by GH18 and GH20 families
(bottom). d | Degradation of the glycosaminoglycan heparan sulfate (top). Mechanism used by polysaccharide
lyases (bottom). e | Simplified mechanism utilized by GH88 and GH105 families in the processing of 4,5-​unsaturated
oligosaccharides produced by polysaccharide lyases. A, general acid catalytic residue; B, general base catalytic residue;
NG, neutralizing group.

addition to a co-​regulated O-​sulfatase to carry out the of uronic acids facilitates a stepwise E1cb mechanism
E1cb mechanism
A two-​step chemical reaction depolymerization of heparin and heparan sulfate94. involving deprotonation at C-5 and cleavage of the bond
in which an acidic proton is Degradation of heparin and heparan sulfate into oligosac­ from C-4 to the adjoining sugar via elimination, leading
abstracted by a base to form charides is initiated by a surface-​localized polysaccharide to the formation of a 4,5-​unsaturated oligosaccharide
a stabilized carbanion, followed lyase PL12 (BT4662) which exhibits preferential cleav­ product95 (Fig. 3d).
by elimination of a leaving
group as the carbanion lone
age at low sulfation regions46. Polysaccharide lyases have Following transport of the released oligosaccharides
pair moves to form a π-​bond a distinct mechanism compared with glycoside hydro­ into the periplasm, three other polysaccharide lyases
with the adjacent atoms. lases. The presence of a carboxylic acid at the C-5 position of varying specificity, in addition to several sulfatases,

NATURe RevIeWS | Microbiology

0123456789();:
Reviews

Xenobiotics
generate 4,5-​unsaturated disaccharides, which them­ xenobiotics and endobiotics, the human glycosyltrans­
Molecules that are not selves are substrates for a glucuronyl hydrolase (BT4658) ferase 1 (GT1) UDP-​glucuronyltransferase covalently
produced or expected to from the GH88 family94,96. BT4658 cleaves the linkage attaches a β-​glucuronic acid moiety to available OH, NH
be found within an organism between 4,5-​unsaturated uronic acid, and GlcNAc or SH (and even activated CH) functionalities on small
(for example, drugs).
(or GlcN), generating monosaccharide products that molecules via a single SN2 displacement17,100,101, as shown
Endobiotics are further metabolized, thus resulting in complete deg­ in Fig. 4. These glucuronides are then excreted into the
Molecules that are produced radation of target glycosaminoglycans by commensal gastrointestinal tract for disposal100. However, certain gut
or are expected to be bacteria. Enzymes of the GH88 family (and also GH105) bacteria can use these glucuronides as a carbon source
found within an organism
employ a non-​Koshland mechanism involving hydration by use of β-​glucuronidases (GUSs) (primarily of CAZy
(for example, hormones).
of the double bond and spontaneous decomposition of family GH2) to release the glucuronic acid, thereby reac­
the resultant hemiketal resulting in glycosidic bond tivating the compound. In numerous cases this reacti­
cleavage13 (Fig. 3e). B. thetaiotaomicron utilizes a simi­ vation has been shown to lead to toxic effects, with a
lar strategy for the metabolism of chondroitin sulfate, notable example being the severe diarrhoea brought on
dermatan sulfate and hyaluronic acid97–99. by cancer treatment using irinotecan102,103.
Structural characterization and activity profiling
Glucuronidation: linking CAZymes and drug of GUS enzymes in the gut microbiota have revealed
metabolism distinct active site structures that have clear effects on
It is becoming increasingly evident that the gut micro­ activity and substrate scope. In particular, the pres­
biome can have important roles in drug metabolism4. In ence or absence of two specific loops within the GUS
order to inactivate and increase the solubility of certain active site can result in enzymes that are specific for

R–X Excretion and/or


reabsorption
R–X
R–X

O–
O– H O–
UDP O–
O O– O
O X R O O
HO O O Microbial GUS HO
HO O HO X R OH
HO X R HO
HO HO OH
HO
O O OH +
HO
P R–X

O UMP Inactivated
with ↑ solubility Reactivated
with ↓ solubility
Where X=O, N, S or other suitable nucleophile

b OH
H H
O– O–
O O H
O O H O–
HO O O HO N
HO HO O O
N HO
HO HO O
N HO
N OH
O
Nicotine glucuronide Oestradiol-17-glucuronide
SN-38 glucuronide
OH O

Cl O N
F O
O O N
F N
H S
F N N N
H – O O O
F O –
O O OH
OH
O O
HOOH HO
OH

Regorafenib glucuronide Sulfinpyrazone glucuronide

Fig. 4 | glucuronidation of small molecules and reactivation by bacterial gUSs. a | In the liver, human
UDP-​glucuronyltransferase inactivates small molecules through glucuronidation. In the large intestine, microbial
β-​glucuronidases (GUSs) can reactivate these compounds. b | Selection of molecules known to be glucuronidated.
Note that SN-38 is the active metabolite of the anticancer drug irinotecan.

www.nature.com/nrmicro

0123456789();:
Reviews

either small nonpolar molecules102,104,105 or glucuronic aeruginosa, Acinetobacter nosocomialis M2 and other
acid-​containing glycosaminoglycans 104,106. Interest pathogens promote infection through degradation
now lies in the identification of the particular enzymes of glycoproteins involved in immune function81,119,120.
that reactivate troublesome conjugated drugs and Likewise, during infection, the GH18 ENGase from
the development of specific inhibitors, which could Streptococcus pyogenes cleaves N-​glycans from host IgG
be co-​administered with those drugs to selectively molecules, thereby decreasing the affinity of IgG for the
shut down their reactivation, thereby mitigating side Fc receptor and promoting survival of S. pyogenes121,122.
effects. The differing active site structures of these The degradation of N-​glycans in serum for use as an
GUS enzymes offers promise in the design of selective energy source has also been shown to fuel Bacteroides
inhibitors102,103,105,107. fragilis during infection123.

Disease and the microbiota Applications of CAZymes


Most pathogens cannot compete with resident microbi­ Bacterial glycosylation systems for biotherapeutic
ota for their carbohydrate food source, and so are effec­ production
tively excluded from the gut under normal conditions. Glycosyltransferases from the human gut microbiota
Consequently, disruptions to the gut ecosystem seem to are valuable tools in glycobiology. Compared with their
have an important role in establishment of pathogenic eukaryotic counterparts, bacterial glycosyltransferases
bacteria. For example, antibiotic treatment disrupts are often easier to express in Escherichia coli and do not
cross-​feeding networks between mucin degraders and have such strict substrate requirements. Furthermore,
non-​mucin degraders and allows the proliferation of as many bacteria synthesize polysaccharides that mimic
pathogenic bacteria such as Salmonella typhimurium and human glycosylation (to escape immunity)118, bacterial
Clostridioides difficile108. In such cases, pathogens that glycosyltransferases can be used for the synthesis of
lack the enzymes for host glycan degradation will scav­ glycoproteins with eukaryotic glycosylation patterns.
enge free monosaccharides released by the residents108,109. As an example, bacterial sialyltransferases have enabled
Introduction of new niches can also drive dysbiosis. For production of therapeutic glycoproteins with prolonged
instance, trehalose is a disaccharide (Glcα1–1αGlc) that half-​life124,125.
has only been introduced into the western diet on a large Bacterial oligosaccharyltransferases (OSTs) are a
scale in the past 20 years110,111. Paralleling its introduction class of enzyme with interesting biomedical applica­
has been an increased incidence of C. difficile infection tions. Physiologically, these enzymes are responsible
and the evolution of mechanisms for enhanced trehalose for the en bloc transfer of lipid-​linked oligosaccharides
utilization by epidemic strains110,111. to extracellular proteins in numerous pathogens 126.
Whereas most commensal bacteria are relegated to Intriguingly, the OST that generates N-​linked glycans in
the outer mucosal layers, during infection pathogens Campylobacter jejuni recognizes a motif similar to that
must often cross the mucosal barrier. Towards this end, of eukaryotic OSTs and has a similar fold127. However,
bacterial67,112 and viral113 pathogens express glycoside in contrast to their far more complex, multisubunit
hydrolases that break down mucosal glycans in the gut eukaryotic counterparts, bacterial OSTs are remarkably
and airways. Often assisting in this process are micro­ promiscuous and can transfer oligosaccharides with
bial adhesins, lectins and CBMs that endow bacteria minimal recognition requirements (often requiring
with the ability to attach to the mucosa, and thereby just an acetamide group at the C-2 position) to protein
localize the degradative CAZymes114. Microorganisms acceptors126. In nature, the requisite glycosyltransferases
can also take advantage of host-​expressed proteins with for synthesis of the lipid-​linked oligosaccharide donor
lectin activities115. In that regard, humans secrete trefoil often occur as gene clusters, which can be transferred
factor proteins, which bind a GlcNAc-​α1,4-​Gal disaccha­ into E. coli with the appropriate OST to enable synthesis
ride present on mucins115. This binding helps to oligo­ of glycoproteins carrying oligosaccharides that are nor­
merize mucin proteins, increasing the viscoelasticity mally found in lipopolysaccharides128. Such systems have
of the mucus. Helicobacter pylori has evolved a similar been engineered for the development and production of
Glc-​α1,4-​Gal disaccharide epitope, which is bound by glycoconjugate vaccines129,130. Through the introduction
trefoil factors and which likely, in turn, aids the binding of non-​natural gene clusters, these same OSTs can also
of H. pylori to gut mucins116. Many bacteria also express transfer non-​bacterial glycans of choice. Interestingly,
CAZymes that cleave this epitope117, thereby effectively this has enabled production of human-​like N-​linked
abrogating trefoil binding and likely weakening the and O-​linked glycans in E. coli131,132. Further work is
mucosal barrier115. needed to improve the yield and substrate scope of these
Many CAZymes are involved in evasion of the host enzymes (in many cases they only act on highly flexi­
immune response during infection. Perhaps the best ble regions of proteins)126 if they are to be viable means
known examples of this are the glycosyltransferases that of production of therapeutic glycoproteins in E. coli.
synthesize host glycan-​mimicking structures to help Indeed, it seems likely that mixing both eukaryotic and
the pathogen evade detection as non-​self118. In parti­ prokaryotic glycosyltransferases within glycosylation
cular, the presence of sialic acid on pathogens often pathways will provide an optimal route to production
helps trick the immune system into complacency118. of glycoprotein therapeutics131,133. For further details,
Enzymatic breakdown of molecules involved in the interested readers are directed to the in-​depth review
immune response is another strategy used by patho­ by Kightlinger et al. on recent advances in glycoprotein
gens. For example, O-​glycoproteases in Pseudomonas synthesis126.

NATURe RevIeWS | Microbiology

0123456789();:
Reviews

Antibody-​dependent
Synthesis of human milk oligosaccharides for and selectivity141–143 (Fig. 5c). Their approach largely relies
cellular toxicity microbiota manipulation on mutants of GH20 enzymes from B. bifidum, which
An immune response in which Given the key roles of the gut microbiota in human normally catalyse the breakdown of HMOs141–143. These
binding of antibodies to a cell health, it is of little surprise that there has been great enzymes can use the readily prepared oxazoline deriva­
surface results in the lysis of
interest in the development of ways to manipulate it. tives of GlcNAc and LNB as glycan donors144 and lactose
that cell by effector cells of the
immune system. Towards this end, approaches using dietary intervention, as an inexpensive glycan acceptor.
drugs and prebiotics have been developed. The intimate
link between carbohydrate metabolism and microbiome Glycan remodelling with bacterial enzymes
composition makes carbohydrate supplementation an The ability to generate homogeneous, well-​defined gly­
obvious choice3,134,135. coforms of antibodies and other therapeutic proteins
Bifidobacteria are naturally enriched in the infant is highly desirable for many pharmaceutical appli­
gut microbiota. Human breast milk contains a diverse cations as it enables evaluation of the impact of spe­
array of glycan structures collectively known as human cific glycan structures on biopharmaceutical efficacy,
milk oligosaccharides (HMOs)136. Humans lack the safety and function145. For example, removal of the
enzymes necessary for HMO degradation. Rather, core fucose or replacement of this core fucose (equiv­
HMOs enrich for beneficial microorganisms, primarily alent to 6-​deoxy-​l-​galactose) with 6-​azido-​l-​fucose
bifidobacteria, that are capable of their degradation136–138. or l-​galactose on N-​glycans present on monoclonal
Thus, HMOs have been suggested as an additive to antibody therapeutics leads to desirable increases
infant formula to enrich the microbiota of bottle-​fed in antibody-​dependent cellular toxicity146. Such results
infants. However, unlike many dietary polysaccha­ highlight the importance of glycan remodelling efforts.
rides, HMOs cannot be readily obtained from natu­ One approach for generating defined glycoforms
ral sources at an industrial scale. The most successful involves in vitro remodelling of the glycoprotein using
method for the commercial production of HMOs, such a wild-​type ENGase (GH18 or GH85) and its respec­
as 2′-​fucosyllactose (Fuc-​α1,2-​Gal-​β1,4-​Glc), has been tive glycosynthase mutants with activated substrates
through incorporation of glycosyltransferases (and other for selective installation of N-​glycan structures. First,
enzymes) from gut bacteria (commonly H. pylori) into deglycosylation of the glycoprotein is performed using
workhorse microorganisms for large-​scale production wild-​type ENGases, leaving a GlcNAc handle on the
by fermentation139. 2′-​Fucosyllactose produced in this target protein22. ENGase glycosynthase mutants can
manner is now incorporated into numerous infant milk then be used for the attachment of N-​glycan structures
formulas worldwide139. to this stub using activated oxazoline donors, which
Complementary methods to fermentation for HMO are readily generated from free oligosaccharides22,144
synthesis include in vitro approaches, which repurpose (Fig. 5b,d). Bacterial endoglycosidases from the GH18
glycoside hydrolases from gut bacteria for glycan synthe­ and GH85 families have been particularly useful in
sis rather than degradation. An excellent example of this glycan remodelling147 and have been used to generate
is the kilogram-​scale synthesis of lacto-​N-​biose (LNB; defined glycoforms of well-​known therapeutic anti­
Gal-​β1,3-​GlcNAc) from sucrose and free GlcNAc using bodies, including the blockbuster drugs Herceptin and
a glycoside phosphorylase from Bifidobacterium bifidum rituximab148–150. Currently, this approach is largely lim­
in a multienzyme reaction23. ited to the modification of a single glycoform on one site
More recent work has highlighted the potential for or multiple sites with the same glycoform149,150. However,
glycosynthase technology to be used for industrial-​scale recent work conducted by Wang and colleagues beauti­
synthesis of HMOs. The original glycosynthase concept fully demonstrated how the glycan structures at Fc and
was introduced by Withers and co-​workers in 1998 Fab domains could be introduced differentially by tak­
(ref.140). The basic principle relies on the mutation of ing advantage of the highly selective nature of ENGases
a catalytic residue of the glycoside hydrolase such that and of an α-1,6-​fucosidase from Lactobacillus casei151.
it cannot degrade its natural substrate (Fig. 5a,b). When In this way, the authors generated an engineered glyco­
this inactivated mutant is presented with an activated form of cetuximab with eightfold higher efficacy than
glycosyl donor of opposite anomeric configuration to the non-​engineered version. Similar approaches have
that of its natural substrate (and thereby mimicking the also been used to edit glycans on cell surfaces152. Another
covalent glycosyl enzyme intermediate of retaining gly­ approach towards selectivity is through introduction of
coside hydrolases), these are accepted as substrates in N-​linked GlcNAc and (pseudo-​eukaryotic) Glc handles
the glycosylation reaction and can form oligosaccharides into proteins using bacterial glycosyltransferases153–155.
and other glycoconjugates22. One of the key benefits of Use of these introduced handles along with ENGase
using glycosynthases over glycosyltransferases, the nat­ glycosynthase technology has opened the door to
ural enzymes for glycan synthesis, is that they do not site-​s elective and structure-​s elective installation of
require the use of expensive nucleotide sugar substrates. N-​glycans153–155. Clearly, this technology is in its early
Many variations of glycosynthases have been developed phases and an expanded toolkit of ENGases and other
over the years, but the basic principle remains the same enzymes is needed for biopharmaceutical remodelling
(for a comprehensive review see Danby and Withers22). to enable access to more synthetically complex glycan
Using glycosynthases, the Nidetzky group has devised structures and provide site selectivity.
synthetic routes for production of lacto-​N -​triose The human gut microbiome is an ideal place to
(GlcNAc-​β1,3-​Gal-​β1,4-​Glc) and lacto-​N-​tetraose search for endo-​glycoside hydrolases against host gly­
(Gal-​β1,3-​GlcNAc-​β1,3-​Gal-​β1,4-​Glc) with good yields cans to be used in glycan remodelling, as it contains

www.nature.com/nrmicro

0123456789();:
Reviews

a Catalytic Catalytic b Catalytic Catalytic


acid/base acid/base acid/base acid/base
O – O O O
F
O– OH O– OH
O H O O H O
O O R
O R O R
R H N+ HN
F O
O

CH3 CH3 CH3 CH3

Mutated catalytic Mutated catalytic Mutated oxazolinium Mutated oxazolinium


nucleophile nucleophile stabilizer stabilizer

c
OH HO OH OH HO OH
OH OH OH
HO
HO O O O O
O O O HO O O
HO O O
HO O
HN OH HO OH OH HO OH
O OH HN OH
OH O

Lacto-N-triose Lacto-N-tetraose

Wild-type ENGase Mutant ENGase

N O

e
OH OH OH
HO HO HO
O O O
OH OH
HO AcHN H AcHN HO
O O ROH O H2O O OH NADH
O NADH R AcHN O
HO NAD+ HO
NAD+
AcHN AcHN
O OH
R

Type O red blood cell

β1,3 β1
α1,2
GH109
Red blood cell

α1,3 β1,3 β1

GH36 galactosaminidase
α1,2
GalNAc
Type A red deacetylase
blood cell α1,3 β1,3 β1

α1,2
GalNAc GlcNAc Gal Galactosamine

Fig. 5 | Applications of enzymes from the gut microbiome. a,b | Glycosynthase concept initially put forward by Withers
and colleagues (part a) and adaptation for enzymes following a substrate-​assisted mechanism (part b). c | Human milk
oligosaccharides (HMOs) produced using bifidobacterial enzymes following the glycosynthase concept. d | General
scheme for N-​glycan remodelling using endo-​β-​N-​acetylglucosaminidases (ENGases). e | Two recent methods for use
of carbohydrate-​active enzymes (CAZymes) to generate type O blood from type A. Insert: overview of the mechanism
utilized by the glycoside hydrolase GH109 family for cleavage of the terminal GalNAc from the A antigen.

a vast reservoir of bacterial genetic diversity7. The ten­ of novel endo-​glycoside hydrolases extremely probable.
dency of gut bacteria to cleave and import whole oligo­ Indeed, recent work has highlighted the potential for
saccharides prior to degradation makes the discovery discovery of endo-​glycoside hydrolases from the human

NATURe RevIeWS | Microbiology

0123456789();:
Reviews

gut microbiome that are active on host glycans such as addition and reduction) to achieve net hydrolysis of the
mucins68,78,156. Of particular note has been the discovery terminal GalNAc24, as shown in Fig. 5e.
of enzymes that degrade the repeating LacNAc struc­ To identify even better suited enzymes, we recently
tures common in host glycans, as well as enzymes that screened the human gut microbiome for enzymes capa­
cleave the intact sialyl-​T antigen (a ubiquitous O-​glycan) ble of blood type degradation. The mucus layer of the
from proteins and cell surfaces78,156. human gut contains glycan structures including the A,
B and H antigens162. Thus, it seemed likely that bacte­
Universal blood production ria inhabiting the gut may produce enzymes capable of
Blood transfusion is an indispensable part of the blood antigen degradation67. This effort yielded a pair
health-​care system, saving many thousands of lives annu­ of highly active CAZymes — a carbohydrate esterase
ally, but must be performed with very careful attention to and a GH36 galactosaminidase from the bacterium
the ABO blood types of both donor and recipient157,158. Flavonifractor plautii — that work in tandem to remove
The basis of the ABO blood group system revolves the terminal N-​acetylgalactosamine of the A antigen25
around a set of related carbohydrates attached to glyco­ (Fig. 5e). This enzyme pair outcompetes any previously
proteins and lipids on the surface of red blood cells. The known A antigen cleaving enzyme, and potentially pro­
basic structure is that of the H antigen (which confers vides a more feasible method of production of universal
type O blood), with type B and A antigens being derived donor blood25. Results such as this further highlight the
from this structure by decoration with α-​galactose or wealth of valuable enzyme activities present within
α-​N-​acetylgalactosamine, respectively. The type O cells the human gut microbiome and portends the many yet
are non-​antigenic for the vast majority of people and can to be uncovered.
be donated to people with type A, B or AB as well as type
O blood, and are thus known as the universal donor type. Concluding remarks
As a consequence, type O blood is particularly important Knowledge of the diversity of CAZymes present in the
in emergency situations and is often in short supply159. human gut microbiota will advance our understanding
The use of glycosidases to remove the α-​galactose or of the intimate relationship between us and our micro­
α-​N-​acetylgalactosamine residues of type B and A blood bial selves and provide new insight into enzymes that
to produce universal donor type O blood was first pro­ can be exploited for various purposes. However, despite
posed by Goldstein et al. in 1982 and was shown to be a efforts thus far, we have only begun to scrape the surface
useful strategy for conversion of type B to type O in initial of CAZyme diversity within the human gut microbiota.
campaigns160. However, those studies used an inefficient Further research into how carbohydrates are partitioned
α-​galactosidase from green coffee beans that required within the gut microbiota will provide insights into how
unfavourable conditions and enormous amounts of dietary carbohydrates may affect our overall health.
enzyme for successful blood group conversion160. A sub­ Although there is an abundance of sequence information
stantial advance came in 2007 when Liu et al. discovered about gut bacteria, we still do not know the functions
the GH109 and GH110 families, which act on the A and of most of the genes present. Future efforts could use
B antigen, respectively24. These enzymes function at much ultra-​high-​throughput functional screening approaches
lower enzyme loadings and under more favourable con­ to catalogue such enzymatic activities 163–165. Such
ditions than the coffee bean α-​galactosidase24. Curiously, enzymes, and their engineered glycosynthetic mutants,
GH109s are proposed to follow a non-​conventional could prove useful for tailoring the glycosylation of bio­
reaction mechanism similar to that first demonstrated pharmaceuticals for improved efficacy and even advance
for enzymes of the GH4 family161. In this mechanism, our understanding of the fundamental roles of glycans.
the enzyme uses a tightly bound NAD+ cofactor to carry
out a sequence of chemical steps (oxidation, elimination, Published online xx xx xxxx

1. Sender, R., Fuchs, S. & Milo, R. Revised estimates for 8. Riley, L. W., Raphael, E. & Faerstein, E. Obesity in the bacterial polysaccharide monooxygenases.
the number of human and bacteria cells in the body. United States — dysbiosis from exposure to low-dose Microbiol. Mol. Biol. Rev. 81, e00015–e00017
PLoS Biol. 14, e1002533 (2016). antibiotics? Front. Public. Heal. 1, 69 (2013). (2017).
2. Kau, A. L., Ahern, P. P., Griffin, N. W., Goodman, A. L. 9. Koh, A., De Vadder, F., Kovatcheva-Datchary, P. & 16. Vaaje-Kolstad, G. et al. Characterization of the
& Gordon, J. I. Human nutrition, the gut microbiome Bäckhed, F. From dietary fiber to host physiology: chitinolytic machinery of Enterococcus faecalis
and the immune system. Nature 474, 327–336 short-chain fatty acids as key bacterial metabolites. V583 and high-resolution structure of its oxidative
(2011). Cell 165, 1332–1345 (2016). CBM33 enzyme. J. Mol. Biol. 416, 239–254
3. Martens, E. C., Kelly, A. G., Tauzin, A. S. & Brumer, H. 10. Price, K., Lewis, J., Wyatt, G. & Fenwick, G. Flatulence (2012).
The devil lies in the details: how variations in — causes, relation to diet and remedies. Nahrung 32, 17. Lairson, L. L., Henrissat, B., Davies, G. J. & Withers, S. G.
polysaccharide fine-structure impact the physiology 609–626 (1988). Glycosyltransferases: structures, functions, and
and evolution of gut microbes. J. Mol. Biol. 426, 11. Lombard, V., Ramulu, H. G., Drula, E., Coutinho, P. M. mechanisms. Annu. Rev. Biochem. 77, 521–555
3851–3865 (2014). & Henrissat, B. The carbohydrate-active enzymes (2008).
4. Zimmermann, M., Zimmermann-Kogadeeva, M., database (CAZy) in 2013. Nucleic Acids Res. 42, 18. Boraston, A. B., Bolam, D. N., Gilbert, H. J. &
Wegmann, R. & Goodman, A. L. Mapping human D490–D495 (2014). Davies, G. J. Carbohydrate-binding modules:
microbiome drug metabolism by gut bacteria and 12. Zechel, D. L. & Withers, S. G. Glycosidase mechanisms: fine-tuning polysaccharide recognition. Biochem. J.
their genes. Nature 570, 462–467 (2019). anatomy of a finely tuned catalyst. Acc. Chem. Res. 382, 769–781 (2004).
5. Johnson, K. V. A. & Foster, K. R. Why does the 33, 11–18 (2000). 19. Davies, G. J., Gloster, T. M. & Henrissat, B. Recent
microbiome affect behaviour? Nat. Rev. Microbiol. 13. Jongkees, S. A. K. & Withers, S. G. Unusual enzymatic structural insights into the expanding world of
16, 647–655 (2018). glycoside cleavage mechanisms. Acc. Chem. Res. 47, carbohydrate-active enzymes. Curr. Opin. Struct. Biol.
6. Proctor, L. What’s next for the human microbiome? 226–235 (2014). 15, 637–645 (2005).
Nature 569, 623–625 (2019). 14. Armendáriz-Ruiz, M., Rodríguez-González, J. A., 20. Koshland, D. E. Stereochemistry and the mechanism
7. El Kaoutari, A., Armougom, F., Gordon, J. I., Camacho-Ruíz, R. M. & Mateos-Díaz, J. C. in of enzymatic reactions. Biol. Rev. 28, 416–436
Raoult, D. & Henrissat, B. The abundance and Lipases and Phospholipases: Methods and Protocols (1953).
variety of carbohydrate-active enzymes in the human (ed. Sandoval, G.) 39–68 (Humana Press, 2018). 21. Berlemont, R. & Martiny, A. C. Glycoside hydrolases
gut microbiota. Nat. Rev. Microbiol. 11, 497–504 15. Agostoni, M., Hangasky, J. A. & Marletta, M. A. across environmental microbial communities.
(2013). Physiological and molecular understanding of PLoS Comput. Biol. 12, e1005300 (2016).

www.nature.com/nrmicro

0123456789();:
Reviews

22. Danby, P. M. & Withers, S. G. Advances in enzymatic 45. Anderson, K. L. & Salyers, A. A. Genetic evidence 67. Tailford, L. E., Crost, E. H., Kavanaugh, D. & Juge, N.
glycoside synthesis. ACS Chem. Biol. 11, 1784–1794 that outer membrane binding of starch is required Mucin glycan foraging in the human gut microbiome.
(2016). for starch utilization by Bacteroides thetaiotaomicron. Front. Genet. 6, 81 (2015).
23. Nishimoto, M. & Kitaoka, M. Practical preparation J. Bacteriol. 171, 3199–3204 (1989). 68. Briliūtė, J. et al. Complex N-glycan breakdown by
of lacto-N-biose I, a candidate for the Bifidus factor 46. Brown, H. A. & Koropatkin, N. M. Host glycan gut Bacteroides involves an extensive enzymatic
in human milk. Biosci. Biotechnol. Biochem. 71, utilization within the Bacteroidetes Sus-like paradigm. apparatus encoded by multiple co-regulated genetic
2101–2104 (2007). Glycobiology 31, 697–706 (2021). loci. Nat. Microbiol. 4, 1571–1581 (2019).
24. Liu, Q. P. Y. P. et al. Bacterial glycosidases for the 47. Cuskin, F. et al. Human gut Bacteroidetes can utilize This paper provides insights into how
production of universal red blood cells. Nat. Biotechnol. yeast mannan through a selfish mechanism. Nature B. thetaiotaomicron utilizes extensive enzymatic
25, 454–464 (2007). 517, 165–169 (2015). machinery to carry out the degradation of
25. Rahfeld, P. et al. An enzymatic pathway in the human 48. Luis, A. S. et al. Dietary pectic glycans are degraded structural variants of complex N-glycans.
gut microbiome that converts A to universal O type by coordinated enzyme pathways in human 69. Rahfeld, P. et al. Prospecting for microbial
blood. Nat. Microbiol. 4, 1475–1485 (2019). colonic Bacteroides. Nat. Microbiol. 3, 210–219 α-N-acetylgalactosaminidases yields a new
This paper highlights how functional metagenomics (2018). class of GH31 O-glycanase. J. Biol. Chem. 294,
can be applied for discovery of enzymes with 49. Rakoff-Nahoum, S., Foster, K. R. & Comstock, L. E. 16400–16415 (2019).
therapeutic potential from within the gut The evolution of cooperation within the gut 70. Martens, E. C., Chiang, H. C. & Gordon, J. I. Mucosal
microbiome. microbiota. Nature 533, 255–259 (2016). glycan foraging enhances fitness and transmission of a
26. Muegge, B. D. et al. Diet drives convergence in gut 50. Rakoff-Nahoum, S., Coyne, M. J. & Comstock, L. E. saccharolytic human gut bacterial symbiont. Cell Host
microbiome functions across mammalian phylogeny An ecological network of polysaccharide utilization Microbe 4, 447–457 (2008).
and within humans. Science 332, 970–974 (2017). among human intestinal symbionts. Curr. Biol. 24, 71. Pudlo, N. A. et al. Symbiotic human gut bacteria with
27. Sonnenburg, E. D. & Sonnenburg, J. L. The ancestral 40–49 (2014). variable metabolic priorities for host mucosal glycans.
and industrialized gut microbiota and implications 51. Cartmell, A. et al. A surface endogalactanase in MBio 6, e01282–15 (2015).
for human health. Nat. Rev. Microbiol. 17, 383–390 Bacteroides thetaiotaomicron confers keystone status 72. Derrien, M., Belzer, C. & de Vos, W. M. Akkermansia
(2019). for arabinogalactan degradation. Nat. Microbiol. 3, muciniphila and its role in regulating host functions.
28. Groussin, M. et al. Unraveling the processes shaping 1314–1326 (2018). Microb. Pathog. 106, 171–181 (2017).
mammalian gut microbiomes over evolutionary time. 52. Sobala, L. F. et al. An epoxide intermediate in 73. Hooper, L. V., Xu, J., Falk, P. G., Midtvedt, T. &
Nat. Commun. 8, 14319 (2017). glycosidase catalysis. ACS Cent. Sci. 6, 760–770 Gordon, J. I. A molecular sensor that allows a gut
29. Freter, R., Brickner, H., Botney, M., Cleven, D. (2020). commensal to control its nutrient foundation in a
& Aranki, A. Mechanisms that control bacterial This study provides key experimental evidence competitive ecosystem. Proc. Natl Acad. Sci. USA 96,
populations in continuous-flow culture models of for a substrate-assisted mechanism proceeding 9833–9838 (1999).
mouse large intestinal flora. Infect. Immun. 39, through a 1,2-epoxide intermediate by GH99 74. Bry, L., Falk, P. G., Midtvedt, T. & Gordon, J. A model
676–685 (1983). family members, one of the most recently of host–microbial interactions in an open mammalian
30. Wu, G. D. et al. Linking long-term dietary patterns elucidated glycoside hydrolase mechanisms. ecosystem. Science 273, 1380–1383 (1996).
with gut microbial enterotypes. Science 334, 53. Valguarnera, E., Scott, N. E., Azimzadeh, P. & 75. Benjdia, A. & Berteau, O. Sulfatases and radical SAM
105–109 (2011). Feldman, M. F. Surface exposure and packing of enzymes: emerging themes in glycosaminoglycan
31. Tap, J. et al. Gut microbiota richness promotes its lipoproteins into outer membrane vesicles are coupled metabolism and the human microbiota. Biochem. Soc.
stability upon increased dietary fibre intake in healthy processes in Bacteroides. mSphere 3, e00559-18 Trans. 44, 109–115 (2016).
adults. Environ. Microbiol. 17, 4954–4964 (2015). (2018). 76. Varki, A. Nothing in glycobiology makes sense,
32. Makki, K., Deehan, E. C., Walter, J. & Bäckhed, F. 54. Briggs, J. A., Grondin, J. M. & Brumer, H. Communal except in the light of evolution. Cell 126, 841–845
The impact of dietary fiber on gut microbiota in host living: glycan utilization by the human gut microbiota. (2006).
health and disease. Cell Host Microbe 23, 705–715 Environ. Microbiol. 23, 15–35 (2021). 77. Luis, A. S. et al. A single sulfatase is required to
(2018). 55. Ndeh, D. et al. Complex pectin metabolism by gut access colonic mucin by a gut bacterium. Nature 598,
33. Vangay, P. et al. US immigration westernizes the bacteria reveals novel catalytic functions. Nature 332–337 (2021).
human gut microbiome. Cell 175, 962–972.e10 544, 65–70 (2017). 78. Crouch, L. I. et al. Prominent members of the human
(2018). This tour de force offers insights into the gut microbiota express endo-acting O-glycanases to
34. Smits, S. A. et al. Seasonal cycling in the gut coordination of carbohydrate degradation by gut initiate mucin breakdown. Nat. Commun. 11, 4017
microbiome of the Hadza hunter-gatherers of bacteria, identifies several new CAZy families and (2020).
Tanzania. Science 357, 802–805 (2017). revises the structure of RG-II (one of the most This work first identifies surface-localized endolytic
35. David, L. A. et al. Diet rapidly and reproducibly alters complex dietary glycans known). glycoside hydrolases acting on O-glycans in
the human gut microbiome. Nature 505, 559–563 56. Tamura, K. et al. Molecular mechanism by which Bacteroides (as had been observed in other PULs
(2014). prominent human gut bacteroidetes utilize mixed- but not against O-glycans) and as such provided
36. De Filippis, F., Pellegrini, N., Laghi, L., Gobbetti, M. linkage β-glucans, major health-promoting cereal insights into degradation of mucosal glycans by gut
& Ercolini, D. Unusual sub-genus associations of polysaccharides. Cell Rep. 21, 417–430 (2017). bacteria.
faecal Prevotella and Bacteroides with specific dietary 57. Rogowski, A. et al. Glycan complexity dictates 79. Noach, I. et al. Recognition of protein-linked glycans
patterns. Microbiome 4, 57 (2016). microbial resource allocation in the large intestine. as a determinant of peptidase activity. Proc. Natl
37. Zaramela, L. S. et al. Gut bacteria responding to Nat. Commun. 6, 7481 (2015). Acad. Sci. USA 114, E679–E688 (2017).
dietary change encode sialidases that exhibit 58. Lozupone, C. A., Stombaugh, J. I., Gordon, J. I., This insightful work first illustrated the molecular
preference for red meat-associated carbohydrates. Jansson, J. K. & Knight, R. Diversity, stability and basis of glycoprotease activity.
Nat. Microbiol. 4, 2082–2089 (2019). resilience of the human gut microbiota. Nature 489, 80. Shon, D. J. et al. An enzymatic toolkit for selective
38. Leeming, E. R., Johnson, A. J., Spector, T. D. & 220–230 (2012). proteolysis, detection, and visualization of mucin-
Le Roy, C. I. Effect of diet on the gut microbiota: 59. Lapébie, P., Lombard, V., Drula, E., Terrapon, N. & domain glycoproteins. Proc. Natl Acad. Sci. USA 117,
rethinking intervention duration. Nutrients 11, 2862 Henrissat, B. Bacteroidetes use thousands of enzyme 21299–21307 (2020).
(2019). combinations to break down glycans. Nat. Commun. 81. Haurat, M. F. et al. The glycoprotease CpaA secreted
39. Sonnenburg, E. D. et al. Diet-induced extinctions 10, 2043 (2019). by medically relevant Acinetobacter species targets
in the gut microbiota compound over generations. 60. Laine, R. A. Invited commentary: A calculation of multiple O-linked host glycoproteins. mBio 11,
Nature 529, 212–215 (2016). all possible oligosaccharide isomers both branched e02033–20 (2020).
40. Hehemann, J.-H., Kelly, A. G., Pudlo, N. A., and linear yields 1.05 × 10 structures for a reducing 82. Malaker, S. A. et al. The mucin-selective protease StcE
Martens, E. C. & Boraston, A. B. Bacteria of the hexasaccharide: the isomer barrier to development enables molecular and functional analysis of human
human gut microbiome catabolize red seaweed of single-method saccharide sequencing or synthesis cancer-associated mucins. Proc. Natl Acad. Sci. USA
glycans with carbohydrate-active enzyme updates systems. Glycobiology 4, 759–767 (1994). 116, 7278–7287 (2019).
from extrinsic microbes. Proc. Natl Acad. Sci. USA 61. Terrapon, N. et al. PULDB: the expanded database of 83. Pluvinage, B. et al. Architecturally complex
109, 19786–19791 (2012). polysaccharide utilization loci. Nucleic Acids Res. 46, O-glycopeptidases are customized for mucin
This paper provides key insights into how new D677–D683 (2018). recognition and hydrolysis. Proc. Natl Acad. Sci. USA
capabilities for carbohydrate degradation can be 62. Despres, J. et al. Xylan degradation by the human gut 118, e2019220118 (2021).
obtained by gut bacteria via horizontal gene transfer. Bacteroides xylanisolvens XB1AT involves two distinct 84. Hughes, G. W. et al. The MUC5B mucin polymer is
41. Pluvinage, B. et al. Molecular basis of an agarose gene clusters that are linked at the transcriptional dominated by repeating structural motifs and its
metabolic pathway acquired by a human intestinal level. BMC Genomics 17, 326 (2016). topology is regulated by calcium and pH. Sci. Rep. 9,
symbiont. Nat. Commun. 9, 1043 (2018). 63. Corfield, A. P. The interaction of the gut microbiota 17350 (2019).
42. Hehemann, J. H. et al. Transfer of carbohydrate-active with the mucus barrier in health and disease in 85. Renzi, F. et al. Glycan-foraging systems reveal the
enzymes from marine bacteria to Japanese gut human. Microorganisms 6, 78 (2018). adaptation of Capnocytophaga canimorsus to the dog
microbiota. Nature 464, 908–912 (2010). 64. Martens, E. C., Neumann, M. & Desai, M. S. mouth. mBio 6, e02507–e02514 (2015).
43. Cerqueira, F. M., Photenhauer, A. L., Pollet, R. M., Interactions of commensal and pathogenic 86. Reily, C., Stewart, T. J., Renfrow, M. B. & Novak, J.
Brown, H. A. & Koropatkin, N. M. Starch digestion by microorganisms with the intestinal mucosal barrier. Glycosylation in health and disease. Nat. Rev. Nephrol.
gut bacteria: crowdsourcing for carbs. Trends Microbiol. Nat. Rev. Microbiol. 16, 457–470 (2018). 15, 346–366 (2019).
28, 95–108 (2020). 65. Ficko-Blean, E. & Boraston, A. B. Insights into the 87. Fairbanks, A. J. The ENGases: versatile biocatalysts
44. Anderson, K. L. & Salyers, A. A. Biochemical evidence recognition of the human glycome by microbial for the production of homogeneous N-linked
that starch breakdown by Bacteroides thetaiotaomicron carbohydrate-binding modules. Curr. Opin. Struct. Biol. glycopeptides and glycoproteins. Chem. Soc. Rev. 46,
involves outer membrane starch-binding sites and 22, 570–577 (2012). 5128–5146 (2017).
periplasmic starch-degrading enzymes. J. Bacteriol. 66. Sonnenburg, J. L., Angenent, L. T. & Gordon, J. I. 88. Knapp, S. et al. NAG-Thiazoline, an N-acetyl-β-
171, 3192–3198 (1989). Getting a grip on things: how do communities of hexosaminidase inhibitor that implicates acetamido
This seminal work describes the first characterization bacterial symbionts become established in our participation. J. Am. Chem. Soc. 118, 6804–6805
of Sus from B. thetaiotaomicron. intestine? Nat. Immunol. 5, 569–573 (2004). (1996).

NATURe RevIeWS | Microbiology

0123456789();:
Reviews

89. Vocadlo, D. J. & Withers, S. G. Detailed interactions: the Streptococcus pneumoniae 137. Charbonneau, M. R. et al. Sialylated milk
comparative analysis of the catalytic mechanisms paradigm. FEBS Lett. 592, 3865–3897 (2018). oligosaccharides promote microbiota-dependent
of β-N-acetylglucosaminidases from families 3 113. Vahey, M. D. & Fletcher, D. A. Influenza A virus surface growth in models of infant undernutrition. Cell 164,
and 20 of glycoside hydrolases. Biochemistry 44, proteins are organized to help penetrate host mucus. 859–871 (2016).
12809–12818 (2005). eLife 8, e43764 (2019). 138. Xiao, J. Z. et al. Distribution of in vitro fermentation
90. Trastoy, B. et al. Structural basis of mammalian 114. Etzold, S. & Juge, N. Structural insights into ability of lacto-N-biose I, a major building block of
high-mannose N-glycan processing by human gut bacterial recognition of intestinal mucins. Curr. Opin. human milk oligosaccharides, in bifidobacterial
Bacteroides. Nat. Commun. 11, 899 (2020). Struct. Biol. 28, 23–31 (2014). strains. Appl. Environ. Microbiol. 76, 54–59 (2010).
91. Nihira, T. et al. Discovery of β-1,4-d-mannosyl- 115. Järvå, M. A. et al. Trefoil factors share a lectin activity 139. Bych, K. et al. Production of HMOs using microbial
N-acetyl-d-glucosamine phosphorylase involved in that defines their role in mucus. Nat. Commun. 11, hosts — from cell engineering to large scale
the metabolism of N-glycans. J. Biol. Chem. 288, 2265 (2020). production. Curr. Opin. Biotechnol. 56, 130–137
27366–27374 (2013). 116. Reeves, E. P. et al. Helicobacter pylori (2019).
92. Higgins, M. A. et al. N-Glycan degradation pathways lipopolysaccharide interacts with TFF1 in a 140. Mackenzie, L. F., Wang, Q., Warren, R. A. J. &
in gut- and soil-dwelling Actinobacteria share common pH-dependent manner. Gastroenterology 135, Withers, S. G. Glycosynthases: mutant glycosidases
core genes. ACS Chem. Biol. 16, 701–711 (2021). 2043–2054.e2 (2008). for oligosaccharide synthesis. J. Am. Chem. Soc. 120,
93. Cordeiro, R. L. et al. N-Glycan utilization by 117. Ficko-Blean, E., Stubbs, K. A., Nemirovsky, O., 5583–5584 (1998).
Bifidobacterium gut symbionts involves a specialist Vocadlo, D. J. & Boraston, A. B. Structural 141. Schmölzer, K., Weingarten, M., Baldenius, K. &
β-mannosidase. J. Mol. Biol. 431, 732–747 (2019). and mechanistic insight into the basis of Nidetzky, B. Glycosynthase principle transformed
94. Cartmell, A. et al. How members of the human gut mucopolysaccharidosis IIIB. Proc. Natl Acad. Sci. USA into biocatalytic process technology: lacto-N-triose II
microbiota overcome the sulfation problem posed by 105, 6560–6565 (2008). production with engineered exo-hexosaminidase.
glycosaminoglycans. Proc. Natl Acad. Sci. USA 114, 118. Varki, A. Biological roles of glycans. Glycobiology 27, ACS Catal. 9, 5503–5514 (2019).
7037–7042 (2017). 3–49 (2017). 142. Schmölzer, K., Weingarten, M., Baldenius, K. &
This work illustrates how B. thetaiotaomicron 119. Bardoel, B. W. et al. Identification of an Nidetzky, B. Lacto-N-tetraose synthesis by wild-type
tackles the degradation of host glycosaminoglycans immunomodulating metalloprotease of Pseudomonas and glycosynthase variants of the β-N-hexosaminidase
with highly variable sulfation patterns, specifically aeruginosa (IMPa). Cell. Microbiol. 14, 902–913 from Bifidobacterium bifidum. Org. Biomol. Chem. 17,
heparin and heparan sulfate. (2012). 5661–5665 (2019).
95. Lombard, V. et al. A hierarchical classification of 120. Szabady, R. L., Lokuta, M. A., Walters, K. B., 143. Ruzic, L., Bolivar, J. M. & Nidetzky, B. Glycosynthase
polysaccharide lyases for glycogenomics. Biochem. J. Huttenlocher, A. & Welch, R. A. Modulation reaction meets the flow: continuous synthesis of
432, 437–444 (2010). of neutrophil function by a secreted mucinase of lacto-N-triose II by engineered β-hexosaminidase
96. Nakamichi, Y., Mikami, B., Murata, K. & Hashimoto, W. Escherichia coli O157∶H7. PLoS Pathog. 5, e1000320 immobilized on solid support. Biotechnol. Bioeng.
Crystal structure of a bacterial unsaturated glucuronyl (2009). 117, 1597–1602 (2020).
hydrolase with specificity for heparin. J. Biol. Chem. 121. Allhorn, M., Olin, A. I., Nimmerjahn, F. & Collin, M. 144. Noguchi, M., Tanaka, T., Gyakushi, H., Kobayashi, A.
289, 4787–4797 (2014). Human IgG/FcγR interactions are modulated by & Shoda, S. I. Efficient synthesis of sugar oxazolines
97. Martens, E. C. et al. Recognition and degradation streptococcal IgG glycan hydrolysis. PLoS ONE 3, from unprotected N-acetyl-2-amino sugars by using
of plant cell wall polysaccharides by two human gut e1413 (2008). chloroformamidinium reagent in water. J. Org. Chem.
symbionts. PLoS Biol. 9, e1001221 (2011). 122. Collin, M. et al. EndoS and SpeB from Streptococcus 74, 2210–2212 (2009).
98. Raghavan, V., Lowe, E. C., Townsend, G. E., Bolam, D. N. pyogenes inhibit immunoglobulin-mediated 145. Sjögren, J., Lood, R. & Nägeli, A. On enzymatic
& Groisman, E. A. Tuning transcription of nutrient opsonophagocytosis. Infect. Immun. 70, 6646–6651 remodeling of IgG glycosylation; unique tools with
utilization genes to catabolic rate promotes growth in a (2002). broad applications. Glycobiology 30, 254–267
gut bacterium. Mol. Microbiol. 93, 1010–1025 (2014). 123. Cao, Y., Rocha, E. R. & Smith, C. J. Efficient utilization (2020).
99. Ndeh, D. et al. Metabolism of multiple of complex N-linked glycans is a selective advantage 146. Li, C. et al. Site-selective chemoenzymatic modification
glycosaminoglycans by Bacteroides thetaiotaomicron for Bacteroides fragilis in extraintestinal infections. on the core fucose of an antibody enhances its Fcγ
is orchestrated by a versatile core genetic locus. Proc. Natl Acad. Sci. USA 111, 12901–12906 receptor affinity and ADCC activity. J. Am. Chem. Soc.
Nat. Commun. 11, 646 (2020). (2014). 143, 7828–7838 (2021).
100. Pellock, S. J. & Redinbo, M. R. Glucuronides in the 124. Lindhout, T. et al. Site-specific enzymatic polysialylation This recent example illustrates how modifications to
gut: sugar-driven symbioses between microbe and of therapeutic proteins using bacterial enzymes. the core fucose within N-glycans in the Fc domain
host. J. Biol. Chem. 292, 8569–8576 (2017). Proc. Natl Acad. Sci. USA 108, 7397–7402 can lead to desirable therapeutic properties.
101. Bolleddula, J. & Chowdhury, S. K. Carbon–carbon (2011). 147. Fairbanks, A. J. Chemoenzymatic synthesis of
bond cleavage and formation reactions in drug 125. Geissner, A. et al. 7-Fluorosialyl glycosides are glycoproteins. Curr. Opin. Chem. Biol. 53, 9–15
metabolism and the role of metabolic enzymes. hydrolysis resistant but readily assembled by (2019).
Drug Metab. Rev. 47, 534–557 (2015). sialyltransferases providing easy access to more 148. Parsons, T. B. et al. Optimal synthetic glycosylation
102. Wallace, B. D. et al. Alleviating cancer drug toxicity by metabolically stable glycoproteins. ACS Cent. Sci. 7, of a therapeutic antibody. Angew. Chem. Int. Ed. 55,
inhibiting a bacterial enzyme. Science 330, 831–835 345–354 (2021). 2361–2367 (2016).
(2010). 126. Kightlinger, W., Warfel, K. F., DeLisa, M. P. & 149. Liu, C.-P. et al. Glycoengineering of antibody
103. Kong, R. et al. Old drug new use — amoxapine and Jewett, M. C. Synthetic glycobiology: parts, systems, (Herceptin) through yeast expression and in vitro
its metabolites as potent bacterial β-glucuronidase and applications. ACS Synth. Biol. 9, 1534–1562 enzymatic glycosylation. Proc. Natl Acad. Sci. USA
inhibitors for alleviating cancer drug toxicity. (2020). 115, 720–725 (2018).
Clin. Cancer Res. 20, 3521–3530 (2014). 127. Napiórkowska, M., Boilevin, J., Darbre, T., 150. Huang, W., Giddens, J., Fan, S. Q., Toonstra, C. &
104. Pollet, R. M. et al. An atlas of β-glucuronidases Reymond, J.-L. & Locher, K. P. Structure of bacterial Wang, L. X. Chemoenzymatic glycoengineering of
in the human intestinal microbiome. Structure 25, oligosaccharyltransferase PglB bound to a reactive intact IgG antibodies for gain of functions. J. Am.
967–977.e5 (2017). LLO and an inhibitory peptide. Sci. Rep. 8, 16297 Chem. Soc. 134, 12308–12318 (2012).
105. Ervin, S. M. et al. Targeting regorafenib-induced (2018). 151. Giddens, J. P., Lomino, J. V., DiLillo, D. J., Ravetch, J. V.
toxicity through inhibition of gut microbial 128. Wacker, M. et al. N-linked glycosylation in & Wang, L. X. Site-selective chemoenzymatic
β-glucuronidases. ACS Chem. Biol. 14, 2737–2744 Campylobacter jejuni and its functional transfer into glycoengineering of Fab and Fc glycans of a
(2019). E. coli. Science 298, 1790–1793 (2002). therapeutic antibody. Proc. Natl Acad. Sci. USA 115,
106. Creekmore, B. C. et al. Mouse gut microbiome-encoded 129. Feldman, M. F. et al. Engineering N-linked protein 12023–12027 (2018).
β-glucuronidases identified using metagenome analysis glycosylation with diverse O antigen lipopolysaccharide 152. Tang, F. et al. Selective N-glycan editing on living
guided by protein structure. mSystems 4, e00452-19 structures in Escherichia coli. Proc. Natl Acad. Sci. USA cell surfaces to probe glycoconjugate function.
(2019). 102, 3016–3021 (2005). Nat. Chem. Biol. 16, 766–775 (2020).
107. Pellock, S. J. et al. Gut microbial β-glucuronidase 130. Stark, J. C. et al. On-demand biomanufacturing of 153. Lin, L. et al. Sequential glycosylation of proteins
inhibition via catalytic cycle interception. ACS Cent. Sci. protective conjugate vaccines. Sci. Adv. 7, eabe9444 with substrate-specific N-glycosyltransferases.
4, 868–879 (2018). (2021). ACS Cent. Sci. 6, 144–154 (2020).
108. Ng, K. M. et al. Microbiota-liberated host sugars 131. Valderrama-Rincon, J. D. et al. An engineered This work brings the field closest to selective
facilitate post-antibiotic expansion of enteric eukaryotic protein glycosylation pathway in introduction of specific N-glycan structures to
pathogens. Nature 502, 96–99 (2013). Escherichia coli. Nat. Chem. Biol. 8, 434–436 (2012). different sites within a protein — potentially
This paper provides an excellent illustration of 132. Natarajan, A. et al. Engineering orthogonal human enabling more thorough interrogation of the
how pathogens exploit antibiotic-induced dysbiosis O-linked glycoprotein biosynthesis in bacteria. roles of different glycoforms.
and intercept sugars left available by disrupted Nat. Chem. Biol. 16, 1062–1070 (2020). 154. Schwarz, F. et al. A combined method for producing
cross-feeding relationships for infection. 133. Du, T. et al. A bacterial expression platform for homogeneous glycoproteins with eukaryotic
109. Ganesh, B. P., Klopfleisch, R., Loh, G. & Blaut, M. production of therapeutic proteins containing N-glycosylation. Nat. Chem. Biol. 6, 264–266 (2010).
Commensal Akkermansia muciniphila exacerbates human-like O-linked glycans. Cell Chem. Biol. 26, 155. Xu, Y. et al. A novel enzymatic method for synthesis of
gut inflammation in Salmonella typhimurium-infected 203–212.e5 (2019). glycopeptides carrying natural eukaryotic N-glycans.
gnotobiotic mice. PLoS ONE 8, e74963 (2013). 134. Bindels, L. B., Delzenne, N. M., Cani, P. D. & Walter, J. Chem. Commun. 53, 9075–9077 (2017).
110. Collins, J. et al. Dietary trehalose enhances virulence Opinion: Towards a more comprehensive concept for 156. Wardman, J. F. et al. Discovery and development of
of epidemic Clostridium difficile. Nature 553, prebiotics. Nat. Rev. Gastroenterol. Hepatol. 12, promiscuous O-glycan hydrolases for removal of intact
291–294 (2018). 303–310 (2015). sialyl T-antigen. ACS Chem. Biol. 16, 2004–2015
111. Collins, J., Danhof, H. & Britton, R. A. The role 135. Delannoy-Bruno, O. et al. Evaluating microbiome- (2021).
of trehalose in the global spread of epidemic directed fibre snacks in gnotobiotic mice and humans. 157. Landsteiner, K. & Wiener, A. S. An agglutinable factor
Clostridium difficile. Gut Microbes 10, 204–209 Nature 595, 91–95 (2021). in human blood recognized by immune sera for rhesus
(2019). 136. Fushinobu, S. Unique sugar metabolic pathways of blood. Exp. Biol. Med. 43, 223–223 (1940).
112. Hobbs, J. K., Pluvinage, B. & Boraston, A. B. Bifidobacteria. Biosci. Biotechnol. Biochem. 74, 158. Daniels, G. & Reid, M. E. Blood groups: the past
Glycan-metabolizing enzymes in microbe–host 2374–2384 (2010). 50 years. Transfusion 50, 281–289 (2010).

www.nature.com/nrmicro

0123456789();:
Reviews

159. Selleng, K. et al. Emergency transfusion of patients hydrolase-containing clones as a resource for and Engineering Research Council of Canada (NSERC) during
with unknown blood type with blood group O rhesus D biocatalyst development. mSystems 4, e00082-19 the writing of this Review.
positive red blood cell concentrates: a prospective, (2019).
single-centre, observational study. Lancet Haematol. 166. Wolfenden, R., Lu, X. & Young, G. Spontaneous Author contributions
4, e218–e224 (2017). hydrolysis of glycosides. J. Am. Chem. Soc. 120, All authors contributed to all aspects of the article.
160. Goldstein, J., Siviglia, G., Hurst, R., Lenny, L. & 6814–6815 (1998).
Reich, L. Group B erythrocytes enzymatically 167. McCarter, J. D. & Stephen Withers, G. Mechanisms Competing interests
converted to group O survive normally in A, B, of enzymatic glycoside hydrolysis. Curr. Opin. Struct. P.R. and S.G.W. are co-founders of a company to develop
and O individuals. Science 215, 168–170 (1982). Biol. 4, 885–892 (1994). enzymes for conversion of blood type. The research underly-
161. Yip, V. L. Y. et al. An unusual mechanism of glycoside 168. Davies, G. et al. Ten years of CAZypedia: a living ing that is mentioned in this Review. J.F.W. and R.K.B. declare
hydrolysis involving redox and elimination steps encyclopedia of carbohydrate-active enzymes. no competing interests.
by a family 4 β-glycosidase from Thermotoga Glycobiology 28, 3–8 (2018).
Peer review information
maritima. J. Am. Chem. Soc. 126, 8354–8355 169. Grondin, J. M., Tamura, K., Déjean, G., Abbott, D. W.
Nature Reviews Microbiology thanks David Bolam, Bernard
(2004). & Brumer, H. Polysaccharide utilization loci:
Henrissat and Nicole Koropatkin for their contribution to the
162. Rossez, Y. et al. Almost all human gastric mucin fueling microbial communities. J. Bacteriol. 199,
peer review of this work.
O-glycans harbor blood group A, B or H antigens e00860–e00816 (2017).
and are potential binding sites for Helicobacter pylori. 170. Glenwright, A. J. et al. Structural basis for nutrient Publisher’s note
Glycobiology 22, 1193–1206 (2012). acquisition by dominant members of the human Springer Nature remains neutral with regard to jurisdictional
163. Tauzin, A. S. et al. Investigating host–microbiome gut microbiota. Nature 541, 407–411 (2017). claims in published maps and institutional affiliations.
interactions by droplet based microfluidics. Microbiome This paper reports the first crystal structure
8, 141 (2020). of SusCD complexes from B. thetaiotaomicron
164. Colin, P.-Y. et al. Ultrahigh-throughput discovery and provides evidence for a ‘pedal bin’ mechanism. Related links
of promiscuous enzymes by picodroplet functional CAZy database: http://www.cazy.org/
metagenomics. Nat. Commun. 6, 10008 (2015). Acknowledgements CAZypedia: https://www.cazypedia.org
165. Armstrong, Z. et al. High-throughput recovery and The authors were supported by funding from the Canadian
characterization of metagenome-derived glycoside Institutes of Health Research (CIHR) and the Natural Sciences © Springer Nature Limited 2022

NATURe RevIeWS | Microbiology

0123456789();:

You might also like