You are on page 1of 21

REVIEWS

Global view of human protein


glycosylation pathways and functions
Katrine T. Schjoldager   , Yoshiki Narimatsu   , Hiren J. Joshi    ✉ and Henrik Clausen    ✉
Abstract | Glycosylation is the most abundant and diverse form of post-translational modification
of proteins that is common to all eukaryotic cells. Enzymatic glycosylation of proteins involves
a complex metabolic network and different types of glycosylation pathways that orchestrate
enormous amplification of the proteome in producing diversity of proteoforms and its biological
functions. The tremendous structural diversity of glycans attached to proteins poses analytical
challenges that limit exploration of specific functions of glycosylation. Major advances in quantitative
transcriptomics, proteomics and nuclease-based gene editing are now opening new global ways to
explore protein glycosylation through analysing and targeting enzymes involved in glycosylation
processes. In silico models predicting cellular glycosylation capacities and glycosylation outcomes
are emerging, and refined maps of the glycosylation pathways facilitate genetic approaches to
address functions of the vast glycoproteome. These approaches apply commonly available cell
biology tools, and we predict that use of (single-cell) transcriptomics, genetic screens, genetic
engineering of cellular glycosylation capacities and custom design of glycoprotein therapeutics
are advancements that will ignite wider integration of glycosylation in general cell biology.

O-GlcNAcylation
Glycosylation of proteins is arguably the most diverse thereby instructing myriad functions6–8. The ensem-
The enzymatic process post-translational modification. Proteins are glycosylated ble of glycans found on glycoproteins, including
directed by the N-acetyl- by enzymes or through non-enzymatic glycation where glycosylphosphatidylinositol (GPI)-anchored proteins and
d-glucosamine (GlcNAc)
glucose (aldehyde form) reacts with lysine and arginine proteoglycans, alongside glycosphingolipids and free
glycosyltransferase (OGT) that
transfers GlcNAc to proteins
residues in proteins, and undergo further changes that oligosaccharides and polysaccharides constitutes the
(Ser and Thr residues) eventually lead to advanced glycation end products glycome of a cell, and the ensemble of glycoconjugates
occurring in the cytosol that serve important functions in ageing and disease, at the cell surface constitutes the glycocalyx (Fig. 1).
and nucleus of cells. especially diabetes1. The enzymatic protein glycosylation The glycome is diverse, involving different types
processes (herein referred to as glycosylation) mostly of glycoconjugates and oligosaccharides with varying
Isoenzymes
Enzymes that catalyse the
involve sequential concerted steps in the endoplasmic compositions, sequences and linkages of sugar moieties.
same reactions but differ reticulum (ER) and the Golgi system resulting in glyco- Nevertheless, despite this diversity, the glycoconjugates
in amino acid sequence and sylation of most (>85%) secretory proteins2,3. In addition, do share certain features, such as common structural
often have partially distinct the majority of nuclear and cytoplasmic proteins undergo scaffolds and terminal modifications9. The diversity in
(non-redundant) functions.
dynamic O-GlcNAcylation4, perhaps making glycosylation types of glycans and structures has expanded during
the most abundant post-translational modification (even evolution of eukaryotes likely in response to needs for
more abundant than phosphorylation)5. increased molecular cues and regulation10,11. Protein
Protein glycosylation is a complex, multistep pro- glycosylation pathways are nearly identical across mam-
cess that employs around 200 glycosyltransferase malian cells, although several glycan features were elim-
enzymes that determine which proteins are to become inated late in evolution by gene inactivation resulting in
Center for Glycomics, glycoproteins, the positions of glycans on those pro- xenoantigens12–14.
Department of Cellular and teins and the glycan structures assembled (Table 1). Studies of deficiencies in glycosylation enzymes
Molecular Medicine, Faculty of Furthermore, the initial attachment of glycans may be in animal models and human diseases have advanced
Health Sciences, University differentially regulated in cells by expression of distinct understanding of biological functions of protein glyco-
of Copenhagen, Copenhagen,
Denmark.
glycosyltransferase isoenzymes, and thus some proteins sylation and demonstrated that most glycosyltransferases
✉e-mail: joshi@sund.ku.dk; may or may not become glycoproteins depending on serve essential roles in mammalian physiology15–18.
hclau@sund.ku.dk their cell of origin and in response to functional needs. Glycosylation of proteins is thus integral to their func-
https://doi.org/10.1038/ Glycosylation greatly amplifies the proteome by pro- tion and should be considered in functional studies of
s41580-020-00294-x ducing diverse proteoforms with different properties, the proteome. However, our understanding of specific

Nature Reviews | Molecular Cell Biology


Reviews

Table 1 | initiation steps for human protein glycosylation pathways


Type Symbol linkage initiation glycosylation sites in target proteins
enzymes
Sequence motifs Specific
domain
N-Glycosylation LLO/GlcNAcβ–Asn OST complex N-X-S/T, X≠P None
β (STT3A/B)
N-X-C; N-G: N-X-V; X≠P
Asn

O-Glycosylation GalNAcα–Ser/Thr GALNT1–20 Weak isoform-specific None


α (Tyr) motifs51
Ser/Thr/Tyr GALNT11 C6-X-X-X-T-C1 LA
Fucα–Ser/Thr POFUT1 C -X-X-X-X-(S/T)-C
2 3
EGF
α POFUT2 C-X-X-(S/T)-C-X-X-G TSR
Ser/Thr

GlcNAcβ–Ser/Thr EOGT C5XX(G/P/S)(Y/F/W)(T/S) EGF


β GXXC6
Ser/Thr

Glycosylphosphatidylinositol Manα–Ser/Thr POMT1, POMT2 ND None


(GPI)-anchored α TMTC1–4 ND EC
glycoproteins Ser/Thr
A class of proteins that are
Unknown ND IPT
attached to the membrane Glcβ–Ser POGLUT1 C1-X-S-X-(A/P)-C2 EGF
lipid bilayer via a carboxy- β POGLUT2–3 C3-X-N-T-X-G-S-(F/Y)-X-C4
terminal glycolipid anchor Ser
consisting of phosphoethanola-
mine, an oligosaccharide core Xylβ–Ser XYLT1, XYLT2 a-a-a-a-G-S-G-a-(a/G)-a None
and phosphatidylinositol. β (a = D/E)
Ser
Proteoglycans
Proteins carrying one or more Galβ–Hyl COLGALT1–2 Collagen repeats Collagen
glycosaminoglycan chains β
attached covalently.
Hyl

Glycocalyx GlcNAcβ–Ser/Thr OGT None None


The cell coat comprising β
glycans and glycoconjugates
Ser/Thr
surrounding animal cells
found as an electron-dense Glcα–Tyr GYG Y194 in glycogenina NA
layer by electron microscopy.
α
It protects the cell from
Tyr
physical stress and mediates
a plethora of macromolecular C-Mannosylation Manα–Trp DPY19L1–4 W-X-X-W TSR
and cell–cell interactions. α
Trp
Xenoantigens
Antigens found in multiple Glypiation NA Protein–C(O) Transamidase Carboxy-terminal None
species that elicit antibodies EthNP–Man hydrophic segment
in a species without the (GPI anchor)
antigen after transplantation The minimal protein–glycan linkages for the distinct human protein glycosylation pathways are shown with the monosaccharide
of tissues and organs. A major linkage (including the anomeric α/β configuration of linkage) to amino acid residues in proteins (Figs 1 and 3), which include
xenoantigen in porcine to 11 types of O-glycosylation when the 2 O-Fuc, 3 O-Man and 2 O-Glc pathways are considered distinct. For references and details,
human transplantation is the
please refer to the main text. COLGALT, collagen O-Gal transferase; DPY19L, dpy-19 like C-Man transferase; EC, extracellular
cadherin immunoglobulin-like; EGF, epidermal growth factor-like repeat; EOGT, epidermal growth factor-domain specific
Galα1–3Galβ1–R glycan
O-GlcNAc transferase; EthNP, ethanolamine phosphate; Fuc, l-fucose; Gal, d-galactose; GalNAc, N-acetyl-d-galactosamine;
epitope (αGal). GALNT, polypeptide GalNAc transferase; Glc, d-glucose; GlcNAc, N-acetyl-d-glucosamine; GPI, glycosylphosphatidylinositol;
Hyl, hydroxylysine; IPT, immunoglobulin-like, plexin and transcription factor; LA, low-density lipoprotein receptor (LDLR) class A
Lectins repeats; LLO, lipid-linked oligosaccharide; Man, d-mannose; NA not applicable; ND, not determined; OGT, O-GlcNAc transferase;
Proteins that bind to glycans. OST, oligosaccharyltransferase; POFUT, protein O-fucosyltransferase; POGLUT, protein O-Glc transferase; POMT, protein O-Man
Major animal lectin families transferase; TMTC, transmembrane O-Man transferase targeting cadherin (also transmembrane and tetra-trico-peptide repeat
include Galectin, C-type, (TPR) repeat-containing protein); TSR, thrombospondin type 1 repeat; unknown, transmembrane O-Man transferase to be
P-type and I-type lectins. reported; X, any of the 20 amino acids; Xyl, d-xylose; XYLT, protein O-Xyl transferase. aThe unique glycosylation of glycogenin
(GYG) involves autoglucosylation of Tyr residue 194 (Glcα–Tyr) followed by formation of the glycogenin polymer, and is not
Lectins, mostly from plants,
further discussed in the text.
with well-characterized binding
specificities are frequently
used as tools in glycobiology. structure–function relationships and roles of specific glycan-specific antibodies and glycan-binding proteins
Lectins are often multivalent glycans on specific proteins is incomplete. (GBPs, such as lectins). It is therefore often perceived as
with binding affinities in the low The glycome is produced and regulated by the gly- a daunting task to uncover and dissect specific biolog-
micromolar range and binding
avidities approaching the
cosylation machinery in a single cell, yet analysis of ical functions of glycans and the underlying molecular
nanomolar range for larger glycans at the single-cell level is not possible with current mechanisms. Advances in next-generation sequencing
glycans with multiple epitopes. glycomics methods, which are limited to probing with and proteomics are beginning to provide single-cell

www.nature.com/nrm
Reviews

ol
ol
n n

2S 4S
n 6S

2S
LDLR 6S
3S
class A NOTCH NS n
repeats
3S Plasma
P membrane

GPI anchor Glyco- N-Linked O-GalNAc EGF TSR domain α-Dystro- Cadherin Plexin/ Proteo- Collagen
sphingolipid repeats glycan EC IPT domain glycan Hyl–Gal
(for example, NOTCH) domain O-Xyl

O-GlcNAc O-Fuc/O-Glc/ O-Man


O-GlcNAc/ C-Man

Nucleus and cytoplasm Domain specific

Ethanolamine phosphate Phosphate d-Glucose (Glc) l-Fucose (Fuc) N-Acetylneuraminic acid (NeuAc)

d-Glucuronic acid (GlcA) P Phosphoinositol d-Galactose (Gal) N-Acetyl-d-glucosamine (GlcNAc) D-Xylose (Xyl)

ol d-Ribitol (Rbo) d-Glucosamine (GlcN) d-Mannose (Man) N-Acetyl-d-galactosamine (GalNAc) Sulfation (S)

Fig. 1 | Main classes of glycoconjugates of the human cellular glycome. Some overlap in the later processing steps that involve elongation,
Depiction of the key components of the cellular glycome, highlighting types branching and capping of oligosaccharides is found among N-glycosylation
of glycosylation that are specific to distinct protein classes or protein and several types of O-glycosylation as well as in glycosphingolipid
domains. The glycans depicted are only illustrative examples of the glycan biosynthesis (Fig. 3). The background colour scheme is organized according
structures that can be synthesized by the different types of glycosylation to the colour of the first monosaccharide attached to the core protein,
pathways. N-glycans and most GalNAc-type O-glycans are widely found except for glycosylphosphatidylinositol (GPI)-anchored proteins and
on most proteins trafficking the cellular secretory pathway, whereas the glycolipids (shown in grey). The colouring scheme is useful for distinguishing
occurrence of domain-specific glycans is limited to specific protein the protein glycosylation pathways involved in the synthesis of different
domains. The enzymatic processes that orchestrate glycosylation of the types of glycans215 (see also Table 1). Glycan symbols are drawn according to
different types of glycoconjugates are partly distinct and partly overlapping. the Symbol Nomenclature for Glycans (SNFG) format246. Sugar repeat units
The initial attachment of the first monosaccharide (or oligosaccharide for are indicated by square brackets with ‘n’ to indicate a number of possible
N-glycosylation) to proteins represents the key initiation step that repeats. EC, extracellular cadherin; EGF, epidermal growth factor; Hyl,
determines which proteins and positions become glycosylated. These hydroxylysine; IPT, immunoglobulin-like, plexin, transcription factor; LDLR,
initiation steps are distinct for the different types of glycosylation pathways low-density lipoprotein receptor; NS, non-specific; TSR domain,
and, to large extent, direct the structures of glycans that are generated. thrombospondin type 1 repeat domain.

transcriptomes and proteomes, which has opened and the genes encoding the orchestrating enzymes.
the way for global analysis of the network of enzymes We also discuss the emerging genetic and systems bio­
that orchestrate protein glycosylation and the assess- logy approaches to the study of protein glycosylation and
ment of the glycosylation capacities of any given cell. dissection of biological functions. We are fully aware that
Accompanying these are the nuclease-based gene edit- the inherent space limitations necessitate some level of
ing technologies that — through precise manipulation generalization with omittance of numerous details (and
of glycosylation enzymes — provide virtually unlimited referencing available literature). Our aim here is to pro-
opportunities for engineering, exploration and custom vide a global view of how the human glycome is estab-
design of cellular glycosylation capacities. We can now lished and highlight our current understanding of its
probe glycosylation systematically through a genetic roles in physiology.
entry point, and we foresee that with additional efforts
we will be able to connect information on cellular gly- Basis of protein glycosylation
cosylation capacities with the actual outcome of the Glycans are not primary gene products and, in contrast to
glycome and roles of glycosylation in cells. proteins, their synthesis occurs without a template. The
Here, we provide an overview of the current know­ human genome contains around 700 genes encoding
ledge of the human protein glycosylation pathways enzymes, transporters and chaperones required for the

Nature Reviews | Molecular Cell Biology


Reviews

Dolichol
cellular glycosylation machinery, glycan modifications O-Gal transferase 1/2 (COLGALT1/2) and epidermal
(Dol). A polyisoprenol lipid that and their degradation (corresponding to 3–4% of the growth factor-domain specific O-GlcNAc transferase
serves as an acceptor for the genome)19–22. Over 200 of these genes encode glycosyl- (EOGT) — note that throughout the article we refer to
lipid-linked oligosaccharides transferases, and 173 of these work sequentially to estab- glycosyltransferases by their gene names and when refer-
in N-glycan biosynthesis.
lish the complex patterns of sugars found on glycoproteins ring specifically to genes we use italics) retained in the
Type II transmembrane and lipids20 (Table 1). Tremendous efforts by a large num- ER by C-terminal KDEL signals and use activated sugar
glycoproteins ber of laboratories allowed isolation, cloning, expression nucleotides for glycosylation38,39.
Single-pass transmembrane and characterization of these enzymes, thereby estab- Structural elaboration of glycans by sequential
glycoproteins with the amino
lishing insight into their substrate and kinetic pro­perties addition of monosaccharides to extend, branch and
terminus oriented towards the
cytosol and the carboxyl
and roles in glycosylation pathways (Supplementary cap growing oligosaccharides occurs largely in the
terminus facing the lumen Table 1) (for comprehensive reviews, see refs16,23–26). Golgi — one exception has been reported in the pro-
of the secretory pathway With complete genome sequences available, the reper- tein O-Man transferase (POMT)-directed synthesis of
or cell exterior. toire of glycosyltransferase genes is well characterized, O-Man glycans, with the first elongation step mediated
COPI-coated vesicles
and most known glycosidic linkages established between by POMGNT2 occurring in the ER40 (Fig. 3). Following
Coat protein complex I-coated sugar moieties are accounted for20. Nevertheless, new anterograde transport of glycoproteins to the surface,
vesicles that mediate intra- glycosyltransferase genes and even distinct glycosylation further glycosylation (in particular, sialylation) during
Golgi and Golgi-to-endoplasmic pathways are still being discovered27–30. recycling of membrane glycoproteins can occur41–43,
reticulum retrograde transport.
Protein glycosylation takes place in the secretory path- and more extensive modifications including change of
Multipass transmembrane
way (ER and Golgi), nucleus, cytoplasm and mito­chondria O-glycan core structures have been suggested44–47.
proteins of all eukaryotic cells (Fig. 2). Ten mono­saccha­rides — Glycosylation is orchestrated mainly by the kinetic
Proteins spanning the d-glucose (Glc), d-galactose (Gal), N-acetyl-d-glucosamine properties of glycosyltransferases and their compart-
membrane more than once. (GlcNAc), N-acetyl-d-galactosamine (GalNAc), l-fucose mentalization in Golgi stacks, with a distribution related
KDEL signals
(Fuc), d-glucuronic acid (GlcA), d-mannose (Man), to sequential biosynthetic steps19,20,22,48 (Fig. 2). Formation
A carboxy-terminal N-acetylneuraminic acid (Neu5Ac), d-xylose (Xyl) and of multimeric (homomeric and heteromeric) enzyme
Lys-Asp-Glu-Leu (KDEL) d-ribose (Rib) — derived from activated donor sugar complexes may contribute to the orchestration of these
retention sequence found nucleotides or dolichol (Dol)-linked donors are used to glycosylation steps49. Insight into the structures and
on endoplasmic reticulum
build the human glycome. Glycans are attached to pro- catalytic mechanisms of glycosyltransferases reveals
(ER)-resident proteins. The
KDEL receptors recognizing
teins in four different ways — N-linked to asparagine common structural scaffolds with distinct acceptor
this signal facilitate the (Asn), O-linked to the hydroxyl groups of serine (Ser), substrate specificities partly conferred by variable loop
retrograde movement of threonine (Thr) or tyrosine (Tyr), C-linked to trypto- regions extending from the core catalytic unit50,51. It is
ER-based proteins from the phan (Trp) and glypiation — and there are several dif- further proposed that evolutionary diversification of
Golgi and back to the ER
by coat protein complex I
ferent O-linked sugars including GalNAc, Fuc, GlcNAc, the functions of glycosyltransferases involves mutations
(COPI) vesicles. Man, Glc, Xyl and Gal (Table 1). Different types of pro- in the common core sugar nucleotide binding region
tein glycosylation are broadly defined by the sugar– and varying loop regions, which drive the divergence
Sialylation protein linkage, the initial monosaccharide linked to in donor sugars and acceptor substrate recognition,
Modification by the addition of
proteins and, for some types of O-glycosylation, by the respectively50–52. Glycosyltransferases utilizing activated
sialic acids, which are a large
family of glycans derived from
enzymes directing the first step in protein glycosylation donor nucleotide sugars have high specificity for the
the neuraminic acid (Neu) (Fig. 3). Many types of protein glycosylation (Table 1) start nucleotide (although they may have some flexibility for
monosaccharide with a in the ER, two start in the early Golgi (GalNAc-type and the donor monosaccharide) and, in general, form only
nine-carbon backbone. In Xyl-type) and O-GlcNAcylation takes place in the cyto- one type of glycosidic linkage structure50. The final glyco­
humans, N-acetylneuraminic
acid (Neu5Ac) is the most
plasm and nucleus (GalNAc-type O-glycosylation has sylation outcome is influenced by many other factors,
common sialic acid, often also been reported in the nucleus31, but this result may including the availability of substrates and sugar nucleo­
found in the non-reducing be an experimental artefact resulting from glycosylation tides, competing glycosylation reactions, co-factors
terminal of glycoconjugates. occurring during cellular fractionation). Most glyco- (such as Mn2+), intracellular transport, pH and actions
syltransferases are type II transmembrane glycoproteins of protein chaperones and glycosidases, as well as by
O-Glycan core structures
The initiating O-GalNAc
with ER and Golgi lumen-oriented catalytic domains general factors, for example stress, that may affect the
glycan can be extended to that make use of activated sugar nucleotides (UDP-Glc/ normal cellular state.
form four different common GlcNAc/Gal/GalNAc/Xyl/GlcA, GDP-Man/Fuc,
core structures. Core1, CMP-NeuAc and CDP-ribitol) as donor substrates, Human protein glycosylation pathways
Galβ1–3GalNAcα1–O-Ser/Thr;
Core2, GlcNAcβ1–6(Galβ1–3)
and have a short carboxy-terminal segment required The known glycome is generated through 16 distinct
GalNAcα1–O-Ser/Thr; Core3, for retrograde transport from the Golgi to the ER via glycosylation pathways — distinguished on the basis of
GlcNAcβ1–3GalNAcα1– COPI-coated vesicles32,33. Type II glycosyltransferases are the sugar–protein linkage, the initial monosaccharide
O-Ser/Thr; and Core4, prone to proteolytic cleavage in stem regions, unteth- linked to proteins and the unique initiating enzymes —
GlcNAcβ1–6(GlcNAcβ1–3)
ering their catalytic domains from the ER or Golgi which are directed by at least 173 distinct glycosyltrans-
GalNAcα1–O-Ser/Thr. The
core structures can be further
mem­branes34–36 and accounting for the presence of glyco­ ferases (Fig. 3; see also Supplementary Fig. 1). These gly-
elongated or branched. syltransferases in body fluids37. Some ER-resident cosylation pathways include, apart from 2 types of lipid
glycosyltransferases are multipass transmembrane proteins glycosylation, 14 distinct types of protein glycosylation,
and utilize Dol-linked donor substrates (Dol-P-Man, including N-glycosylation, 11 types of O-glycosylation,
Dol-P-Glc or the lipid-linked oligosaccharide precursor), C-mannosylation and generation of GPI-anchored pro-
whereas some glycosyltransferases are soluble ER-resident teins (Table 1). Protein glycosylation involves a series of
enzymes (protein O-fucosyltransferase 1/2 (POFUT1/2), sequential steps to build characteristic oligosaccharide
protein O-Glc transferase 1–3 (POGLUT1–3), collagen structures, including an initiation step determining

www.nature.com/nrm
Reviews

Oligosaccharyltransferase
proteins to be glycosylated, immediate core extension roles (for example, GALNT11 serves specifically the
(OST) complex steps with options for different core structures, elon- low-density lipoprotein receptor (LDLR)-related recep-
A membrane protein gation/branching steps that expand (and repeat) com- tor family (LRPs) and activates their ligand binding
complex in the endoplasmic mon structural motifs and capping steps that terminate properties)65. GalNAc-type O-glycosylation cross-talks
reticulum that transfers an
oligosaccharide from a dolichol
oligosaccharide chains (Fig. 3). with other post-translational modifications (examples
pyrophosphate-activated include FAM20C Ser phosphorylation66,67, VLK Tyr
donor to N-linked acceptor Initiation of protein glycosylation. The initiation step for phosphorylation68 and TPST1/2 Tyr sulfation69).
sequences on secreted each type of protein glycosylation is distinct and regu­ Fuc, Glc and GlcNAc types of O-glycosylation are initi-
proteins.
lated by one or more unique glycosyltransferase, or, for ated in the ER. The most prominent targets for these types
Translocon
N-glycosylation, an oligosaccharyltransferase (OST) complex of O-glycosylation are the NOTCH receptor epidermal
A protein complex that or, for GPI-anchored proteins, a GPI–transamidase growth factor (EGF)-like repeats (Fig. 1). NOTCH receptor
mediates translocation of complex that transfers the preassembled GPI anchor to O-glycosylation represents one of the most complex types
newly synthesized polypeptides the C-terminus of select proteins in the ER53,54 (Table 1). of glycan-mediated regulation of receptor functions (see
from the cytosol across the
endoplasmic reticulum
A total of 47 of the 173 glycosyltransferases direct initiation also Functions of glycosylation below)39,70–72. Initiation
membrane. steps of protein glycosylation. Initiation of N-glycosylation of Fuc-type O-glycosylation is directed by two POFUTs,
and likely POMT-directed O-mannosylation occur wherein POFUT1 serves EGF repeats and POFUT2
Oxidoreductase co-translationally. serves related thrombospondin type 1 repeats. Glc-type
An enzyme that catalyses thiol–
N-glycosylation is initiated in the ER by the oligo- O-glycosylation of EGF repeats in NOTCH is differen-
disulphide exchange reactions.
In vivo oxidoreductases are
saccharyltransferase (OST) complex assembled with tially regulated by the three POGLUTs: POGLUT1 has
important in the oxidative STT3A or STT3B catalytic subunits for co-translational wide specificity for many NOTCH EGF repeats, whereas
protein folding that takes place and post-translational glycosylation, respectively, and POGLUT2 and POGLUT3 have specificities for a single
in the endoplasmic reticulum. these subunits appear to provide some regulation of functionally important repeat (NOTCH1 EGF11 and
Well-known examples are
PDI and ERp57.
N-glycosites55,56. The OST–STT3A complex is asso- NOTCH3 EGF10) and glycosylate at a different position73
ciated with the ER peptide translocon54, whereas the (Table 1). GlcNAc-type O-glycosylation of EGF repeats
Lectin domains OST–STT3B complex includes MAGT1 or TUSC3 is regulated by EOGT74. All of these initiation enzymes
Carbohydrate-binding protein oxidoreductase subunits57. The OST–STT3B complex require folded repeat domains for activity and acceptor
domains.
appears to be the main source of released oligosac- sites have defined sequence motifs.
Sulfation
charides derived from deglycosylation of misfolded Man-type O-glycosylation initiates in the ER and
An enzymatic process that N-glycoproteins destined for proteasomal degradation involves at least three distinct types of initiation enzymes.
transfers a sulfo group to another and found widely in the cytosol56,58,59. A yeast-related O-mannosylation type is directed by the
molecule, for example a glycan, GalNAc-type O-glycosylation of Ser/Thr and pos- POMT1/2 heteromeric complex75. Interestingly, whereas
by modifying a hydroxyl group
on a monosaccharide by
sibly Tyr is initiated in the Golgi by up to 20 poly­ the yeast Man O-glycoproteome is diverse and similar
addition of a sulfo group. peptide GalNAc transferase (GALNT) isoenzymes to the GalNAc-type O-glycoproteome76, the human
Sulfotransferases catalyse with distinct and partly overlapping specificities POMT1/2 complex appears to selectively target the
the reaction using 3′-phospho- (of note, only 15 of those have so far been confirmed mucin-like region in α-dystroglycan and a very limited
5′-adenylyl sulfate (PAPS) as
to be active enzymes25,60), which leads to the genera- number of other proteins27. Two additional types of
a donor.
tion of the simple GalNAcα1–O-Ser/Thr monosac- animal O-mannosylation were recently discovered.
EGF-like repeats charide structure also known as the cancer-associated These are driven by four transmembrane O-Man trans-
Common motifs of 30–40 Tn antigen. GALNTs generally lack clear acceptor ferase targeting cadherins (TMTCs), dedicated primar-
amino acids found in the sequence motifs in target proteins, but they do exhibit ily to modifying the cadherin superfamily, and an as yet
extracellular domain of
transmembrane proteins or in
differences in substrate specificities and orchestrate unreported enzyme that selectively targets IPT domains
proteins known to be secreted. regulation of sites and patterns of O-glycans in pro- found in plexins and in receptor tyrosine kinases c-MET
The epidermal growth factor teins in cooperative ways25,51. Some GALNTs initiate and RON27 (Table 1).
(EGF)-like repeats include six GalNAc transfer directly to peptides, whereas others Mannosyl moieties can also be attached to Trp resi-
conserved cysteines forming
only transfer to prior GalNAc-glycosylated peptides dues (in the consensus WxxW motif; shown for throm-
three disulfide bonds.
(designated follow-up glycosylation). Follow-up glyco- bospondin type 1 repeats and type I cytokine receptors),
Thrombospondin type 1 sylation can occur within a short range (1–3 residues) known as C-mannosylation. This modification is driven
repeats or a long range (6–17 residues) from the initial glyco- by four dpy-19 like C-Man transferase (DPY19L) gly-
(TSRs) Common protein sylation site, and the latter is mediated by C-terminal cosyltransferases and occurs in the ER, presumably
motifs of 50–60 amino acids
(6 conserved cysteines forming
GalNAc-binding lectin domains , a unique prop- co-translationally77.
3 disulfide bonds) found on erty among metazoan glycosyltransferases 51. The Xyl-type O-glycosylation is characteristic of pro-
transmembrane proteins and GalNAc-type O-glycoproteome is extensive, with teoglycans and initiated in the Golgi by protein O-Xyl
proteins in the extracellular more than 3,000 human O-glycoproteins and over transferase 1 (XYLT1) and XYLT2 that both have a rela­
matrix.
15,000 identified O-glycosites61. Analysis of differen- tively defined sequence motif (Table 1). Xyl-type glyco-
Mucin
tial O-glycoproteomes of pairs of isogenic cells with sylation at select Ser residues primes the biosynthesis of
A large viscous heavily disabled GALNT genes confirms that the expressed glycosaminoglycan chains (GAGs). XYLT1 and XYLT2 are
O-glycosylated protein. repertoire of GALNTs in a given cell determines differentially expressed and have overlapping substrate
Mucins are the most abundant its O-glycoproteome, with individual isoenzymes specificities78. The diversity of proteoglycans is limited,
macromolecule in biofluids and
mucus, covering most epithelial
making distinct non-redundant contributions 62–64. but a recent sensitive glycoproteomics strategy almost
surfaces in the body. Some GALNTs, for example GALNT1 and GALNT2, doubled the number of known proteoglycans79.
have major contributions to the O-glycoproteome, Hydroxylysine (HYL)-Gal O-glycosylation is
whereas others serve specific proteins and functional limited to collagens (important components of the

Nature Reviews | Molecular Cell Biology


Reviews

Cell-surface Galectins
glycoprotein

Plasma membrane NEUs

Single-pass
Golgi resident
Endocytosis 7
Soluble enzyme
with KDEL

5 Secretion
Degradation
Multipass
transmembrane protein
Secretion
Retrograde
SPPL3/BACE1 transport ACP2/
ACP5
Adaptor proteins
COPI
CMP Lysosome
Sugar nucleotide SIATs
transporter CMP
CMP

UDP Sugar UMP TGN


GDP Sugar UDP Capping
GMP
4
CMP Sugar GMP
Sugar nucleotide
donors
GDP ol
Elongation and branching
CDP Lysosomal
enzyme
Dol-P-Man/Glc Clathrin
M6PR 6
Core extension or IGF2R
N-Acetylneuraminic GNPTAB/G
acid (NeuAc) Golgi
3
d-Xylose (Xyl) UMP
Phosphate
UMP
UDP Initiation
ol d-Ribitol (Rbo)

d-Glucuronic acid UDP


(GlcA) XYLT1/2 GALNTs
UDP UDP
d-Glucose (Glc) 2
d-Galactose (Gal) COPI
d-Mannose (Man)
GMP
l-Fucose (Fuc)
GDP
1 KDELR UMP
N-Acetyl-d-
glucosamine
(GlcNAc)
N-Acetyl-d- UDP COLGALT1/2
galactosamine EOGT POFUT1/2
POGLUTs
(GalNAc) UMP
POMTs,
TMTCs, COPII
ER
UDP UMP UDP UDP DPY19Ls UDP
GDP
Ribosome
UDP
OST
OGT UDP
OGT UDP
Nucleus Cytoplasm

www.nature.com/nrm
Reviews

◀ Fig. 2 | Subcellular organization of protein glycosylation. The initiation steps of an O-GlcNAc hydrolase (OGA; also known as MGEA5)
most types of protein glycosylation occur in the endoplasmic reticulum (ER) and involve serves to dynamically regulate on/off O-GlcNAc modi-
transfer of a first monosaccharide to an amino acid (to Ser, Thr, Tyr, Trp), or in the case of fications on proteins in concert with phosphorylations4.
N-glycosylation to a preformed oligosaccharide (to Asn). Two types of O-glycosylation OGT contains amino-terminal transmembrane and
(GalNAc-type and Xyl-type) are initiated in the Golgi. O-GlcNAcylation directed by
tetra-trico-peptide repeats (TPRs) that mediate protein–
O-GlcNAc transferase (OGT) occurs in the cytosol and nucleus. The glycosyltransferases
directing these steps are indicated by their gene name (or for N-glycosylation, the oligo- protein interactions and orchestrate access to substrates83.
saccharyltransferase (OST) enzyme complex). Further glycosylation steps (core extension, The residues modified by O-GlcNAcylation are not fur-
elongation and capping) occur throughout the Golgi and trans-Golgi network (TGN). ther glycosylated but pose particular analytic challenges,
ER-resident glycosyltransferases directing initiation of protein glycosylation are being labile and of low stoichiometry.
transmembrane proteins (type III) or are equipped with a Lys-Asp-Glu-Leu (KDEL) retrieval
signal recognized by the KDEL receptor (KDELR) associated with COPI vesicles (1). Further processing of protein glycosylation. The process-
Glycosyltransferases in the Golgi are type II transmembrane proteins with luminal catalytic ing of protein glycosylation involves sequential steps
domains and short cytoplasmic carboxy-terminal sequences (2). Glycosyltransferases adding further monosaccharides to growing oligosac-
use dolichol (Dol)-linked donor substrates (Dol-P-Man, Dol-P-Glc or the lipid-linked charide chains by glycosyltransferases, resulting in core
oligosaccharide precursor) or activated sugar nucleotides transported from the cytosol
extension, elongation and branching, and final capping
into the ER and/or Golgi lumen by members of the SLC35 family of solute carriers (3).
Although there are over 30 human members of the SLC35 family247, only 9 of these have of glycans (Fig. 3). The core extension refers to glycosyla-
been demonstrated to serve in sugar nucleotide transport and their specificities for tion steps and glycosyltransferases unique to particular
donors are not fully clarified. The cisternal maturation model dictates that Golgi-resident types of protein glycosylation. For N-glycosylation, the
glycosyltransferases are maintained and distributed across stacks by retrograde coat initially attached preformed N-glycan oligosaccharide
protein complex I (COPI) vesicular transport directed by motifs in short cytoplasmic tail is trimmed by α-mannosidases in the ER, and sequen-
sequences (4). Stem/transmembrane regions of glycosyltransferases can undergo tial addition of β-GlcNAc residues by MGATs gen-
proteolytic cleavage by, for example, signal peptide peptidase-like 3 (SPPL-3) or erates complex-type bi-antennary, tri-antennary and
β-secretase 1 (BACE-1) proteases in the secretory pathway releasing catalytic domains tetra-antennary N-glycan core structures. GalNAc-type
to the extracellular milieu (5). N-Glycoproteins acquiring mannose-6-phosphate (M6P) O-glycosylation involves four distinct O-glycan core
in the early Golgi by action of the GlcNAc-1-phosphotransferase complex (complex of
structures (Core1–Core4). POMT1/2-driven Man-type
GNPTA, GNPTB and GNPTG, which catalyses formation of GlcNAc1-P-Man linkages)
followed by the uncovering enzyme GlcNAc-1-phosphate hydrolase (NAGPA, which O-glycosylation involves three distinct core struc-
removes the GlcNAc residue leaving M6P) are recognized by the cation-dependent tures (Cores M1–M3). Interestingly, TMTC-initiated
(M6PR) and cation-independent (insulin-like growth factor 2 receptor (IGF2R)) M6P O-mannosylation does not appear to be processed
receptors, and transported in clathrin-coated vesicles to the lysosome where mannose is and appears as the Man monosaccharide. Xyl-type
dephosphorylated by lysosomal acid phosphatases (ACP2 and ACP5), and the receptors O-glycosylation involves a common tetrasaccharide
are recycled back to the Golgi (6). From the cell surface, glycosylated proteins can that is extended by either chondroitin sulfate or heparan
undergo endocytosis followed by recycling, degradation in lysosomes or retrograde sulfate polysaccharides.
transport to the TGN/Golgi (7). Neuraminidases (NEU1–4) may remove sialic acids The elongation and branching biosynthetic steps may
previously attached during the capping step of glycosylation (sialylation), and such be shared among different types of protein glycosylation,
desialylated glycoproteins may be recognized by different glycan-binding proteins,
and the involved glycosyltransferases therefore function
such as galectins, and upon internalization undergo resialylation by sialyltransferases
(SIATs) again in the TGN. CDP, cytosine diphosphate; CMP, cytosine monophosphate; in multiple glycosylation pathways (Fig. 3). Elongation
COLGALT, collagen O-Gal transferase; DPY19L, dpy-19 like C-Man transferase; EOGT, primarily involves N-acetyllactosamine (LacNAc
epidermal growth factor-domain specific O-GlcNAc transferase; GALNT, polypeptide type 2 chain Galβ1–4GlcNAc), often in the form of
GalNAc-transferase; GDP, guanosine diphosphate; GMP, guanosine monophosphate; repeating disaccharides (polyLacNAc) and branches
GNPTAB/G, GlcNAc-1-phosphotransferase; POFUT, protein O-fucosyltransferase; (Galβ1–4GlcNAcβ1–3(Galβ1–4GlcNAcβ1–6)Galβ1–
POGLUT, protein O-Glc transferase; POMT, protein O-Man transferase; TMTC, 4GlcNAc). The isomeric disaccharide type 1 LacNAc
transmembrane O-Man transferase targeting cadherins (also transmembrane and (Galβ1–3GlcNAc) or the N,N′-Diacetyllactosamine
tetra-trico-peptide repeat (TPR) repeat-containing protein); UDP, uridine diphosphate; (LacDiNAc, GalNAcβ1–4GlcNAc) disaccharide are
UMP, uridine monophosphate; XYLT, protein O-Xyl transferase. also found as terminal disaccharides on a common
scaffold of LacNAc glycans. Most of these elongation
extracellular matrix). HYL residues are generated by the and branching reactions are shared with glycolipids.
α-Dystroglycan
Dystroglycan is encoded by the activity of pro-collagen lysine hydroxylases (PLOD1–3), The capping step mainly involves terminal decoration
DAG1 gene and comprises two and these are subsequently glycosylated by COLGALT1/2 of the oligosaccharide chains with Fuc and sialic acid
non-covalently-bound subunits isoenzymes. These events take place in the ER before N-acetylneuraminic acid (Neu5Ac) and is directed
(α and β). The extracellular formation of the mature collagen triple helix, which is by the large sialyltransferase and fucosyltransferase
α-subunit with the O-Man
matri­glycan provides binding to
secreted80. In addition to catalysing lysine hydroxyla- families9,84,85 (Fig. 3).
laminin, and the transmembrane tion, PLOD3 also harbours galactosyltransferase and Processing steps may be specific for certain types
β-subunit provides binding to glucosyltransferase activity, at least in vitro, suggesting of protein glycosylation (pathway specific) or shared
dystrophin and the cytoskeleton. that this enzyme may also be involved in generating the among several types (pathway non-specific). Most
collagen-attached sugar chain81. pathway-specific enzymes do not have close paralogues,
IPT domains
(also known as TIG domains). Finally, O-GlcNAcylation is a highly abundant which infers that expression of these enzymes allows
Immunoglobulin–plexin– glycosylation affecting most cytosolic and nuclear predictions to be made about the cellular glycosylation
transcription (IPT) protein proteins that serves as a nutrient sensor and a master capacity and the glycan structures being produced (Fig. 3)
domains found on cell- switch in regulating signalling, transcription and cellu- (Supplementary Box 1). This concerns glycosyltrans-
surface transmembrane
receptors and intracellular
lar metabolism4,82. O-GlcNAcylation in the cytosol and ferases involved in initiation and most core extension
transcription factors with nucleus is directed by a single soluble O-GlcNAc trans- steps of the different types of protein glycosylation, with
an immunoglobulin-like fold. ferase (OGT) glycosyltransferase that in complex with a few notable exceptions: initiation of GalNAc-type

Nature Reviews | Molecular Cell Biology


Reviews

Specific Specific Non-specific


Elongation
and
Initiation Core extension branching Capping Sulfation

PGAP4 3S
P
PIGA R ST3GAL1–6 GAL3ST1
B3GALT1/2/5
PIGM 3S
PIGZ
PIGV PIGB R
R
Type 1 ST6GAL1/2
Lipid UGT8
glycosylation LacNAc R
B3GNT5 R 4S

B3GALNT1 R CHST8/9
ST6GALNAC1–6 GAL3ST3
UGCG A4GALT B4GALNT3/4 3S
B4GALT5/6 B3GALT4 LacDiNAc R R
R
B4GALNT1 6S
R
MGAT1 ST8SIA1–6 GAL3ST2 CHST1/3*
B4GALT1–4
(MGAT4D) MGAT4A–C 3S
Type 2
R GAL3ST4
LacNAc R
N-Glycosylation STT3A/B (OST) N MGAT3 B4GALNT2 A4GNT R
R 6S 6S
MGAT2
CHST5/
QC FUT8 CHST7/
B3GNT2–4 CHST4/
UGGT1 UGGT2 MGAT5 B3GNT7–9 R
R R CHST2/
C1GALT1 B3GAT1/2 CHST6
(C1GALT1C1) Core1 PolyLacNAc
ABO
Core2
Secretory GALNT1–20 S/T/Y Tn antigen GCNT1/3/4 R Lex Lea R
O-glycosylation B3GNT6 Core3 6S 6S
R R
Core4 GCNT2/7 R 3S CHST10*
FUT1/2 FUT3/5
MFNG/ FUT4–7/
POFUT1 S/T LFNG/ FUT9–11 R
RFNG

B3GLCT ER
POFUT2 S/T
DPM1 Dolichol

EOGT S/T
GDP
POMGNT1 Core M1 ALG6
ALG8
S/T ALG2 ALG11
POMT1/2 MGAT5B Core M2 RXYLT1 LARGE1/2
ALG13/14
S/T ol ol ALG3 ALG10
POMK ALG10B
B3GALNT2 FKTN/ B4GAT1 Core M3 DPAGT1 ALG9
FKRP Matriglycan ALG1
POMGNT2
TMTC1–4 S/T
? (IPT domains) ALG12

Secretory N-Linked LLO precursor biosynthesis


DPY19L1–4 W
C-mannosylation
2S DS
POGLUT1 XXYLT1
GXYLT1/2 CHPF/
Secretory CHPF2/ CHST15 DSE/DSEL
O-glycosylation POGLUT2/3 S CSGALNACT1/2 CHSY1/ 2S UST CS
CHSY3
B3GAT3 6S 4S CHST11–14
B3GALT6 3S CHST3* Sulfation
2S
XYLT1/2 S CHST10* HS 3S
B4GALT7 NDST1–4 GLCE R
NS 3S 2S 6S

COLGALT1/2 Hyl EXT1/2 NS 3S


6S
EXTL1/2
PLOD3 EXTL3 HS6ST1–3 HS3ST3A1/B1 HS2ST1 R
Nucleocytoplasmic OGT S/T
HS3ST1/2/4/5/6 6S 4S
O-glycosylation
2S 3S
Ethanolamine phosphate Phosphate d-Glucose (Glc) N-Acetyl-d-glucosamine (GlcNAc) R

Ceramide P Phosphoinositol d-Galactose (Gal) N-Acetyl-d-galactosamine (GalNAc) Linkage


2 3
d-Glucuronic acid (GlcA) l-Iduronic acid (IdoA) d-Mannose (Man) N-Acetylneuraminic acid (NeuAc)
R 4
ol d-Ribitol (Rbo) d-Glucosamine (GlcN) l-Fucose (Fuc) d-Xylose (Xyl)
8 6

www.nature.com/nrm
Reviews

◀ Fig. 3 | Human glycosylation pathways and enzymes. A global view of glycosylation Side-chain modifications of glycans. Sulfation is the most
pathways, the major structural elements of the glycans and the assigned (predicted) abundant and diverse glycan modification. Whereas 35
biosynthetic roles for glycosyltransferases as well as carbohydrate sulfotransferases Golgi sulfotransferases are involved in glycan sulfation,
(indicated are genes encoding these enzymes; please note that the enzymes that initiate only two sulfotransferases (TPST1/2) direct tyrosine pro-
the novel O-mannosylation specific for immunoglobulin-like, plexin, transcription factor
tein sulfation87. The majority of these sulfotransferases
(IPT) domains are currently not reported (question mark)). The 16 known glycosylation
pathways are organized into major biosynthetic steps specific for pathways (initiation
serve in decorating GAGs, and different sulfation pat-
and core extension) and those that are non-specific (elongation and branching, and terns produced on these large polysaccharides serve as
capping). For pathways involving isoenzymes, all isoforms are listed (with a dash or solidus distinct binding motifs for proteins and regulate wide
character). The background colours for the different protein glycosylation pathways essential biological roles (Fig. 3). Whereas the biosynthe-
mimic the colours of the initiating monosaccharide (lipid glycosylation pathways are sis and sulfation of GAGs is well understood, insight into
shown in grey), and these are maintained for the pathway-specific glycosylation steps. the specific structures of biologically active GAG motifs
Dashed lines indicate alternative pathways for chondroitin sulfate (CS) and dermatan and the sulfotransferase isoenzymes directing these is
sulfate (DS) or heparan sulfate (HS) biosynthesis on the common tetrasaccharide linker. still limited88,89. A different type of GAG, keratan sulfate,
The display is useful to peruse individual genes as well as the glycosylation processes is found on N-glycoproteins and O-glycoproteins and is
as an integrated system. Please note that the major structural variations characteristic
built on polyLacNAc repeats. Keratan sulfate synthesis
of the glycosylation pathways are illustrated, but the diagram is not intended to show all
structural permutations and variations found in cells (linkages only indicated for select is initiated by 6-O-sulfation of GlcNAc carried out by
structures). Supplementary Table 1 provides detailed information of the glycosyltrans- CHST6/2 and subsequent galactosylation by B4GALT4
ferase genes mapped. For information on transcriptional regulation of glycosylation and elongation by B3GNT7 with further 6-O-sulfation of
enzymes shown and their association with congenital diseases and genome-wide Gal involving CHST1/3 (ref.90). Some 14 sulfotransferases
association study traits, see Supplementary Figs 1 and 2 (note that these present the are predicted to direct sulfation of N-glycoproteins
biosynthetic pathways in a vertical orientation as compared with horizontal orientation and O-glycoproteins, but the specific roles of these
shown here and used before215). *Transferases that appear twice in the figure due to dual isoenzymes are only partly understood87 (Fig. 3).
pathway-specificity. COLGALT, collagen O-Gal transferase; DPY19L, dpy-19 like C-Man Phosphorylation of glycans regulates glycosylation
transferase; EOGT, epidermal growth factor-domain specific O-GlcNAc transferase; and serves as a gatekeeper for progression to elonga-
ER, endoplasmic reticulum; GALNT, polypeptide GalNAc-transferase; GDP, guanidine
tion steps. For example, the extracellular kinase POMK
diphosphate; Hyl, hydroxylysine; LacNAc, N-acetyllactosamine (Galβ1–4GlcNAc);
LAcDiNAc, N,N′-Diacetyllactosamine (GalNAcβ1–4GlcNAc); LLO, lipid-linked
phosphorylates the O-Man residue in α-dystroglycan
oligosaccharide; NS, non-specific; OGT, O-GlcNAc transferase; OST, oligosaccharyl­ to induce synthesis of the elaborate matriglycan — an
transferase; POFUT, protein O-fucosyltransferase; POGLUT, protein O-Glc transferase; extracellular matrix-binding motif on α-dystroglycan91,92
POMT, protein O-Man transferase; QC, quality control; R, variable underlying core (Fig. 3). FAM20B phosphorylates the Xyl residue in the
glycan; S, sulfation; TMTC, transmembrane O-Man transferase targeting cadherins forming tetrasaccharide linker, and this phosphorylation
(also transmembrane and tetra-trico-peptide repeat (TPR) repeat-containing protein); affects the third synthesis step directed by B3GALT6
XYLT, protein O-Xyl transferase. regulating GAG synthesis93,94.
Acetylation of sialic acids is an abundant modifica-
O-glycosylation is regulated by 20 GALNT isoenzymes; tion that regulates the interaction of sialoglycoproteins
tri-antennary branching of N-glycans is regulated by with cellular receptors such as the sialic acid-binding
MGAT4A and MGAT4B (note that role of MGAT4C is immunoglobulin-like lectins (Siglecs)95. Acetylation
unclear86); branching of GalNAc-type O-glycans to form also conveys resistance to sialic acid removal by most
Core2 or Core4 O-glycans is mediated by three isoen- sialidases7. Sialic acid acetylation occurs through 9-O
zymes (GCNT1, GCNT3 or GCNT4); extension of the or 7-O-acetylation of the activated Neu5Ac donor sugar
POFUT1-mediated O-Fuc glycan is regulated by three nucleotide by CASD1, and incorporation of acetylated
isoenzymes (MFNG, LFNG or RFNG); and the impor- Neu5Ac into glycans by sialyltransferases in the Golgi96.
tant steps determining chondroitin sulfate or heparan Sialate 9-O-acetylesterases serve as NeuAc deacetylases97.
sulfate GAG biosynthesis on the tetrasaccharide linker Of interest, certain viral receptors bind 9-O-acetyl
are governed by CSGALNACT1 or CSGALNACT2 and Neu5Ac (ref.98). Thus, acetylation of sialic acids in glycans
by EXTL2 or EXTL3, respectively. may function in regulating interactions with endogenous
Glycosyltransferases considered pathway non- receptors, while being exploited by pathogens.
specific include 17 enzymes that are involved in elon-
gation steps, including B3GNTs, B4GALTs, B3GALTs Context-specificity of glycosylation
and B4GALNTs, and 35 glycosyltransferases involved The glycosylation of proteins, in general, reflects the
in capping steps, including FUTs, ST3GALs, ST6GALs, glycosyltransferase repertoire and the glycosylation
ST6GALNACs and ST8SIAs as well as B3GATs, A4GNT capacities of the producing cell. However, the individ-
and ABO (Fig. 3). Arguably, these glycosylation pathway ual protein may not be efficiently glycosylated, leading
non-specific enzymes direct the greatest diversity in the to heterogeneities, and certain glycosylation features are
glycome, although they also produce common struc- targeted to specific proteins and, hence, not universally
tural scaffolds that may reduce this diversity in terms found. Moreover, the secretory pathway and glycosyl-
Glycosaminoglycan chains
(GAGs). Extended linear of distinct functional binding epitopes of the glycome9. ation machinery, in general, are influenced by numer-
polysaccharides comprising Moreover, most of these glycosyltransferases belong to ous cellular and environmental factors that affect the
repeating disaccharides. isoenzyme families with overlapping properties and glycosylation efficiency.
poorly understood non-redundant functions, which
Stoichiometry
The fraction of a glycosylation
at least partly hampers the ability to predict the glycan Heterogeneity in protein glycosylation. Different effi-
site in a glycoprotein that is structures produced by analysis of enzyme expression ciencies in the initiation of glycosylation at specific sites
occupied by a glycan. (Supplementary Box 1). in proteins (stoichiometry of glycan attachment) and

Nature Reviews | Molecular Cell Biology


Reviews

ABH
variable processing of glycans at sites in proteins (struc- proteins to select high-Man N-glycans105. Polysialylation
Carbohydrate antigens of the tures of the glycan) will result in macroheterogeneity (α2–8NeuAc) is another protein-specific modification
ABO blood group system. and microheterogeneity of glycosylation, respectively. found on select N-glycans of the neural cell adhesion
Glycosylation may be influenced by the overall pro- molecule and few other proteins that depends on inter-
Lipoproteins
Large complexes of lipids
tein structure and conformational constraints around actions with protein motifs106,107. Polysialylation may also
and proteins that transport glycosites. For example, the presence of high-Man be found on O-glycans of Neuropilin-2 (ref.108).
lipids in the blood. N-glycans at select sites in mature glycoproteins can
correlate with steric hindrance of the action of manno- Glycosylation features derived from the extracellular
Lipopolysaccharide sidases at these sites in the ER. Preferences for acquir- milieu. The glycome of a cell may to some degree also
A bacterial glycolipid endotoxin
and a major constituent
ing bi-antennary or multi-antennary glycans regardless depend on the glycosylation capacities of other cells
of the outer membrane of of the available glycosylation capacity are found; for through transfer of glycoconjugates and extracellular
Gram-negative bacteria. example, generation of the conserved IgG1 biantennary glycosylation reactions. For example, uptake of ABH
N-glycan (Asn297) in the Fc region shows that glycan– blood group glycolipids from plasma lipoproteins to
peptide backbone interactions and steric hindrance red blood cells109 and uptake of GPI-anchored glyco­
for processing enzymes result in inefficient branching, protein CD52 in sperm during maturation110 both serve to
galactosylation and sialylation99–102. Assessing hetero­ introduce new glycan structures. Glycosylation enzymes
geneity in glycosylation of specific proteins is inherently can also be secreted. Secreted ST6GAL1 can contri­
challenging in glycoproteomics and requires elabo- bute to extracellular sialylation of IgGs and remodel-
rate site-specific analysis with isolated glycoproteins, ling of cell-surface glycans111–114. Glycosyltransferases
but progress towards quantitative glycosite-specific and glycoproteins can also be transferred between
glycoproteomics workflows is being made103. cells via extracellular vesicles and non-membranous
nanoparticles (exomeres), having functional conse-
Protein-specific glycosylation features. Some gly- quences in recipient cells115. Finally, pathogenic bacte-
cosylation features are only observed in a subset of ria encode and can inject virulent glycosyltransferases
cellular proteins. The most notable example here is that O-glycosylate and N-glycosylate host proteins and
tagging of lysosomal hydrolases (some ~60 different interfere with cellular functions, as well as glycoside
N-glycoproteins) with mannose-6-phosphate (Man-6-P) hydrolases that reprogramme cell-surface glycans116–118.
by the GlcNAc-1-phosphotransferase (GNPTAB–
GNPTG) and the uncovering enzyme (UCE)23 (Fig. 2), Functions of glycosylation
which serves as a ligand for the Man-6-P receptors in the Glycosylation has roles in folding, quality control, sta-
trans-Golgi network to direct transport of these enzymes bility, transport and function of proteins7, and many
to the endo-lysosomal system104. Mechanistically, the of these functions serve the proteome globally. For
GlcNAc-1-phosphotransferase appears to recognize example, in the ER, the initial steps of N-glycosylation
conformational motifs in the diverse lysosomal enzyme guide protein folding and their quality control 119.

Fig. 4 | Protein glycosylation serves general roles and specific roles for regulates endocytosis and compartmentalization248. The large Siglec family ◀
protein functions. a | Specific roles of glycosylation in regulating protein promotes cell–cell interactions and regulates important functions in the
functions. Intracellularly, in the nucleus and cytosol, dynamic immune system by engaging numerous sialylated glycan epitopes. Selectins
O-GlcNAcylation regulates numerous cellular processes, including (P-selectin, L-selectin and E-selectin) specifically require both sialic acid and
transcription and signalling, and serves in nutrient sensing. In the secretory fucose, and in some cases also sulfate, for binding. Extrinsic glycan
pathway, endoplasmic reticulum (ER)-initiated types of glycosylation may interactions involve binding by bacterial lectins or adhesins mediating
serve in folding, stability and trafficking of proteins. Several types of bacteria–host interactions, including initial attachment and potential
glycosylation serve specific roles for select proteins driven by differentially invasion and colonization; for example, Helicobacter pylori encodes
regulated glycosylation at specific glycosites, for example, co-regulating numerous adhesins that recognize different histo-blood group-related
processing (for activation or inactivation) by proprotein convertases glycans in the human gastrointestinal tract mucosa249–251. Bacteria also
(GalNAc-type). Within the glycocalyx at the cell surface (zoom-in box), produce glycoconjugates like lipopolysaccharides and glycan structures
specific glycosites serve to (from left to right): regulate the stability and/or that mimic host glycans, allowing the bacteria to avoid recognition by host
activity of glycoproteins, by affecting their susceptibility to regulated immune cells (mimicry). b | Overview of the common scaffolds and capping
proteolytic processing (for example, G protein-coupled receptor (GPCR) motifs, and the combinations of these that help generate diversity of
amino-terminal cleavage to regulate their signalling or entire ectodomain glycans, which serve as the major ligands for GBPs (see Fig. 3 for enzymes
shedding of membrane proteins) (GalNAc-type); by providing contact involved). Core glycan structures (bottom left) are elongated with one of
points for cell adhesion (Poly-Sia and Man-type); by modulating the ligand three types of chains (type 1 N-acetyllactosamine (LacNAc, Galβ1–3GlcNAc),
specificity and/or affinity of receptors like low-density lipoprotein receptors type 2 LacNAc (Galβ1–4GlcNAc) or N,N′-Diacetyllactosamine (LacDiNAc,
(LDLRs) and NOTCH (GalNAc-type, Fuc-type and Glc-type); by modulation GalNAcβ1–4GlcNAc); labelled R1–R3, respectively). The core glycan
effector function of immunoglobulins (N-glycans); or by regulating receptor structures can be extended by chains from R1–R3 (except for O-Man cores,
dimerization and signalling (GalNAc-type). Glycans also serve in a myriad of which can only be extended by R2). Type 2 chains may be repeated to form
interactions with endogenous glycan-binding proteins (GBPs) (intrinsic, left) poly-LacNAc chains (indicated by square brackets, ‘n’ indicating the number
and microbial GBPs (extrinsic, right). Intrinsic glycan interactions serve in of repeat units). The terminal core structures may be variously fucosylated,
cell–cell adhesion and communication, and are mediated by, for example, sulfated and capped by sialic acids, as may residues along the poly-LacNAc
sialic acid-binding immunoglobulin-like lectins (Siglecs) binding to sialic chain, and the combinatorial action of relatively few transferases results in
acid capped glycans for immunomodulation; selectins binding to, for complex glycan structures with large diversity. To better indicate which
example, sialyl-Lewisa antigen for cell trafficking; and galectins binding to residues have been added in a synthesis step, the residues added
different β-Gal glycans for numerous functions, for example, cross-linking by previous steps are presented in grayscale. Pol II, polymerase II; X, any
cell-surface glycoproteins by Galectin-3 pentamers in a dynamic lattice that amino acid.

www.nature.com/nrm
Reviews

a Further, O-glycosylation via O-Fuc and O-Glc of folded


EGF-like and thrombospondin type 1 repeat domains
Cell Ligand Effector
binding function
stabilizes these after folding, enabling secretion71,120,121.
adhesion
Interestingly, other types of O-glycosylation do not
appear to be required for protein transport and secre-
Antibody tion. On the cell surface, glycoconjugates constitute the
Processing and Receptor glycocalyx that provides a barrier and a protective layer
Ectodomain dimerization
activity regulation
cleavage shielding the plasma membrane from physical stress
Signalling and shaping the cell surface. The glycocalyx serves to
mediate numerous intrinsic and extrinsic interactions.
Signalling
GPCR Signalling Excellent recent reviews cover these topics7–9, and here
we focus on non-global roles of glycosylation, high-
Processing by lighting examples where specific glycosyltransferases
pro-protein regulate specific functions at the level of glycosites or
convertases glycostructures (Fig. 4a).

R-X-X-R-T/S Functions determined at the glycosite level. The NOTCH


receptor illustrates how multiple types of O-glycosylation —
Cell–cell Intracellular Bacteria–host O-Fuc (directed by POFUT1), O-GlcNAc (directed by
interaction trafficking interaction EOGT)122 and O-Glc (directed by the POGLUT1–3
Golgi isoenzymes) — converge to modulate complex protein
Siglecs
Adhesins functions by decorating distinct glycosites in the many
Selectins
ER NOTCH EGF repeats39 (Table 1). With 20 isoenzymes,
Glycoprotein
Galectins folding GalNAc-type O-glycosylation by GALNTs provides the
Nucleus greatest opportunity for differential regulation of specific
DNA Pol II Pathogen mimicry sites in proteins, and GALNT isoenzymes serve highly
specific co-regulatory roles in fundamental processes,
Nutrient sensing including in the inhibition of protein processing by
Endocytosis and signalling
proprotein convertases35,123, in the inhibition (or, in some
cases, activation) of proteolytic cleavage by ADAM pro-
b teases with altered shedding of ectodomains of surface
Sialylation Fucosylation Sulfation proteins124, in the activation of β1-adrenergic receptor
n
by modulating its N-terminal cleavage125, in the modu-
n n lation of peptide hormone function and promoting their
6S stability126,127, and in promoting ligand binding by LDLR
6S
n and LRPs65 (Fig. 4a). These functions are subject to tight
OR n regulation by expression levels of specific GALNTs128,
which may be controlled by feedback loops as shown
n
R2 for GALNT3: GALNT3 uniquely co-regulates propro-
R1 R2
Sialylation tein convertase-mediated processing of FGF23 that
Sialylation Fucosylation
R1 R2 R3 S/T/Y Fucosylation serves to regulate phosphate homeostasis129, and in turn
Sulfation
the expression of GALNT3 is responsive to blood phos-
Elongation 3S phate levels130. Man-type O-glycosylation by TMTC1–4
n n
also provides for differential regulation of O-glycans on
3S
n cadherins and protocadherins that may regulate their
functions in cell adhesion131. Initiation of GAG synthesis
R1 R2 R3
R1 R2 R3 by XYLT1 and XYLT2 also likely directs different subsets
Type 1 Type 2 LacDiNAc R1 R2
LacNAc LacNAc of proteoglycans and corresponding functions78.
Fucosylation The dynamic and widespread O-GlcNAcylation in
Core extension Sulfation the nucleus and cytosol regulated by the OGT and OGA
N-Glycan pair of enzymes has a major role in nutrient sensing
S/T/Y through the hexosamine biosynthetic pathway and modu­
Core2 4S 3S
lating the cellular response to stress4. O-GlcNAcylation
at specific glycosites regulates functions of proteins,
S/T/Y S/T/Y R3 S/T/Y including their enzymatic activity, stability and locali-
N-X-S/T S/T Core1
Lipids zation, and the dynamic on/off nature of O-GlcNAc on
O-Man GalNAc-
(only R2) type Fucosylation glycosites enables O-GlcNAcylation to regulate tran-
Sulfation
scription and signalling events in crosstalk with protein
d-Glucose d-Mannose N-Acetyl-d-glucosamine N-Acetylneuraminic phosphorylation4. O-GlcNAcylation regulates transcrip-
(Glc) (Man) (GlcNAc) acid (NeuAc)
tion by modifying drivers of gene expression like Pol II,
d-Galactose l-Fucose N-Acetyl-d-galactosamine Sulfation (S)
(Gal) (Fuc) (GalNAc)
where C-terminal addition and removal of GlcNAc by
OGT and OGA during the transcription cycle regulates

Nature Reviews | Molecular Cell Biology


Reviews

Proprotein convertases
transcription activation and repression. Site-specific proteins resulting in increased affinity towards their
A family of seven secretory O-GlcNAcylation also regulates important processes target glycans147. Glycan binding motifs are generally
mammalian serine proteinases such as neuronal depolarization132 and intermediate fil- directed by multiple isoenzymes that are glycosylation
that post-translationally ament morphology and cell migration133. The OGT sub- pathway non-specific and highly regulated. However,
activate proproteins in the
secretory pathway by limited
strate selectivity is partly regulated through interactions these isoenzymes may have non-redundant activities
proteolysis after multiple basic between the TPR domains, adaptor proteins and target and selectively regulate glycosylation: in megakaryo-
residues with a general protein substrates83. cytes, the B4GALT1-driven LacNAc epitope regulates
recognition motif, (R/K)Xn(R/K). β1 integrin activity required for the formation of mature
A prototypical proprotein
Functions regulated at the glycan structure level. There is platelets148; terminal sialylation of LacNAc or LacDiNAc
convertase is furin.
abundant evidence that the structure of glycans on pro- O-glycans by ST6GAL1 was found to be important to
teins regulates diverse protein interactions and biological regulate B cell activation via lectin Siglec-2 (CD22)95,149;
functions. For N-glycan, for example, tetra-antennary and ST3GAL1-directed capping of Core1 O-glycans
branching by MGAT5 favours synthesis of polyLacNAc controls CD8+ T lymphocyte homeostasis by inhibiting
chains that promote multivalent interactions with lectin apoptosis and regulating memory cell formation150,151.
receptors, for example galectins134–136. Functionally,
MGAT5-mediated branching on α5β1 integrin serves to Changes of glycosylation in disease
enhance cell motility and growth137. By contrast, MGAT3 Glycosylation processes in cells are highly sensitive to
introduces a bisecting GlcNAc residue to the early stages the physiological state, and glycans are prevalent report-
of complex N-glycans that is not elongated and leads to ers of disease152,153. An overwhelming number of studies
inhibition of further N-glycan branching, and there- using lectins, antibodies and direct structural analyses of
fore is a major determining factor for the N-glycome glycan features have documented diverse changes in gly-
architecture138. In line with this, MGAT3-directed gly- cosylation in human diseases and especially cancers153.
cosylation of α5β1 integrin reduces its ability to bind to However, our insight into how these changes in gly-
its ligands in the extracellular matrix and interferes with cosylation occur, what the functional consequences,
cell migration139. As another example, FUT8-directed if any, are and the nature of specific mole­cular mech-
fucosylation of the N-glycan core modulates several anisms is still limited. Congenital disorders of glyco-
specific protein interactions, including EGF–EGFR sylation (CDGs) and genome-wide association studies
binding140, interactions between the T cell receptor and (GWAS) are beginning to enable us to deconvolve highly
the major histocompatibility complex II during T cell specific roles of glycosylation in common diseases (for
activation141, and IgG1 interactions with the FcγRIIIa a global overview of the glycosylation steps and path-
receptor99,142 (Fig. 4a). ways served by glycosyltransferase genes known to
Protein function can also be modulated by specific cause CDGs and/or implicated by GWAS traits, see
GalNAc-type O-glycan core extensions. GCNT1- Supplementary Fig. 2).
directed Core2 O-glycosylation promotes Galectin-
1-induced T cell death of immature T cell precursors and Lessons from congenital disorders of glycosylation.
activated T cells143. GCNT3-directed Core2 O-glycans The importance of glycosylation is clearly under-
are also required for surface expression of intestinal scored by decades of studies of rare monogenic CDGs,
cell differentiation markers144. The β3GlcNAc exten- where more than 60 of the 100 CDGs identified to
sion of O-Fuc on NOTCH regulates the interaction of date are caused by complete or partial loss of function
the receptor with its ligands, Delta and Jagged, and the three of a glycosyltransferase17,18 (Supplementary Table 1).
isoenzymes, MFNG, LFNG or RFNG, directing the CDGs may be caused by mutations in glycosyltrans-
O-Fuc elongation appear to target distinct EGF repeats ferase genes in all types of glycosylation pathways, but
and determine differential binding between these two currently most glycosyltransferase-related CDGs are
ligands71. Another example is assembly of the matri­ caused by deficiency in unique enzymes functioning in
glycan on α-dystroglycan. In the ER, POMGNT2 primes pathway-specific glycosylation steps where loss of func-
matriglycan synthesis by elongating select O-Man gly- tion results in global glycome changes (Supplementary
cans on α-dystroglycan145, and in the Golgi, LARGE1/2 Fig. 2a). These CDGs are generally characterized by
produce the functional glycosaminoglycan-like multisystemic disorders, with a wide spectrum and
matriglycan polymer28. severity of clinical manifestations predicted to arise
The common structural capping motifs on glycans from a myriad of impaired direct and indirect biologi­
found on several types of glycoproteins (Fig. 4b) comprise cal functions of glycans, and these are often difficult to
the major interactome for GBPs, including the endog- trace and dissect17,18.
enous lectin receptors galectins, Siglecs and selectins, CDGs were often identified by profiling N-glycans
and diverse microbial lectins and adhesins7,9,146 (Fig. 4). on an abundant serum glycoprotein (usually transfer-
Common for most mammalian lectins are shallow rin), but today CDGs are also discovered by genome
binding sites and low affinity interactions (micromolar sequencing that is uncovering CDGs with more subtle
to millimolar) for single glycan epitopes, allowing for phenotypes154. This has resulted in the discovery of novel
dynamic and reversible glycan–receptor associations. CDGs caused by deficiencies in isoenzymes with subtle
GBPs may also recognize more complex structures (clus- non-global glycosylation functions20,155, and these are
tered saccharides or discontinuous glycans) involving a pointing us to highly specific regulatory roles of protein
higher-order presentation of glycans that can be within glycosylation. The most illustrative examples so far are
a context of specific glycoconjugates and even specific CDGs caused by loss of function within the GALNT

www.nature.com/nrm
Reviews

isoenzymes, which have uncovered how site-specific Lessons from genome-wide association studies. Survey­
O-glycosylation fine-tunes highly specific protein func- ing glycosyltransferase genes implicated by GWAS for
tions and regulates major physiological processes such as diverse traits or predispositions interestingly revealed
blood phosphate homeostasis (GALNT3) and likely also that GWAS candidate genes represent almost a mirror of
kidney function (GALNT11)129,156,157 (Fig. 5). the glycosyltransferase genes so far identified as causing

Specific Healthy Cancer


Core elongation and branching
Initiation MGAT3↓ • Adhesion
• Migration ECM
Non-specific
Epithelial/
SLeX endothelial EMT
MGAT5↑ cell
α2-6-SA α2-6-SA
ST6GAL1↑ Metastasis
FUT8↑↓ ↑FUT6
• Introduction of bisecting GlcNAcs
• Changes in branching
• Changes in core Fuc SLea T cell
FUT3↑ NK cell Siglec 7/9
Macrophage
T Immune
GCNT1↓ TGFβ
Tn • Switch from 2-3 to 2-6 recognition
C1GALT1C1↓ sialic acid capping Activation Activation Siglec 15
COSMC↓ • Increased SLex and
SLea epitopes Immunosuppression
B3GALT5↓
↑GALNT3/T6 ↓B3GNT6 GCNT3↓
STn
ST6GALNAC1↑ Signalling

• Reduced Core2 branching Signalling


• Reduced Core1 elongation
• Reduced Core3/4 structures
• Accumulation of Tn and STn

Placental CS 2S

6S 4S
Apoptosis
NS 3S

6S Cell death Cell death


Changes in GAGs signalling signalling

d-Galactose (Gal) N-Acetyl-d-glucosamine (GlcNAc) d-Xylose (Xyl) T cell receptor, Death


Selectin Siglec receptor
NK cell activated
d-Mannose (Man) N-Acetyl-d-galactosamine (GalNAc)
receptor
l-Fucose (Fuc) N-Acetylneuraminic acid (NeuAc)
d-Glucuronic acid (GlcA) E-Cadherin Integrin CSF3R EGFR

Fig. 5 | common dysregulated glycosyltransferase genes in cancer. evasion from immune surveillance. This is mediated by the sialic acid-binding
Three major glycosylation pathways (N-glycosylation, GalNAc-type immunoglobulin-like lectin (Siglec) family of receptors, both inhibitory (for
O-glycosylation and O-Xyl glycosaminoglycans) that undergo characteristic example, Siglec 7/9) and activating (for example, Siglec 15), which recognize
changes in cancer (left) and examples of the biological effects these different sialic acid ligands and modulate the signalling responses to tumour
may exert in cancer (right). Glycosylation pathways are simplified ligands resulting in immunosuppression254. Changes in sialylation may be
accord­ing to the scheme in Fig. 3, and the key glycosyltransferase genes attributed to cancer-associated changes in glycosyltransferase genes in the
undergo­ing altered expression in cancer are indicated (arrows indicate GalNAc-type O-glycosylation pathway, most notably the ST6GALNAC1
up/downregulation of expression). Examples of specific effects of altered gene that induces expression of the STn epitope185–187. Signalling and
glycosylation in cancer are illustrated. Adhesion and migration: increased apoptosis: glycosylation can modulate the proximity of cell-surface
branching of N-glycans (MGAT5↑, MGAT3↓) and core fucosylation (FUT8) on receptors to each other, affecting the ability to directly interact or cluster,
adhesion receptors (for example, E-cadherin, integrins) modulates both cell– thereby affecting their signalling capability. For example, loss of a single
cell adhesion252,253 and, alongside increased α2-6-sialic acid (α2-6-SA) GalNAc-type O-glycosylation site (unknown polypeptide GalNAc
capping (ST6GAL1), cell–extracellular matrix (ECM) adhesion through transferase (GALNT) isoform) on one of the cytokine receptors of
integrin receptors253. Mechanisms behind the changes in adhesiveness can granulocytes, colony-stimulating factor 3 receptor (CSF3R), is suggested to
be related to the abundance of receptors, conformational changes or result in increased ligand independent dimerization and aberrant
presentation of binding epitopes to adhesive partners. These glycosylation signalling255. Similarly, increased N-linked branching and core fucosylation
changes are one of the drivers of epithelial–mesenchymal transition (EMT). (FUT8) on epidermal growth factor receptor (EGFR) increase receptor
Metastasis: alongside increased propensity for EMT, the metastatic potential dimerization and signalling promoting growth and progression of cancer181.
of cells is increased by the upregulation of expression of sialyl-Lewisx (SLex) By contrast, clustering of death receptors is reduced in cancers due to the
(FUT6 and ST6GAL1) and sialyl-Lewisa (SLea) (FUT3) glycan epitopes, which truncated GalNAc-type O-glycans and modified N-glycosylation, which
are recognized by selectins on epithelial and endothelial cells promoting leads to tumour cells escaping apoptotic fates256. CS, chondroitin sulfate;
tumour cell extravasation from vasculature and metastatic site homing196–198. GAG, glycosaminoglycan; NK cell, natural killer cell; NS, non-specific;
Immune recognition: altered sialylation patterns on tumour cells allow for S, sulfation; TGFβ, transforming growth factor-β.

Nature Reviews | Molecular Cell Biology


Reviews

Epithelial–mesenchymal
CDGs, and they concentrate around those found to In contrast to protein-coding somatic mutations,
transition exhibit high transcriptional regulation158 (Supplementary there is more evidence to support aberrant expression
The physiological process in Fig. 2a). This suggests the somewhat controversial con- of glycosyltransferases in cancer153,172–175; for example,
which epithelial cells transition clusion that dysregulation of glycosyltransferase iso­ as seen with a hotspot of non-coding mutations around
to a mesenchymal state by
loss of polarity and other
enzymes, rather than loss of function of these enzymes, the ST6GAL1 gene in B cell non-Hodgkin lymphomas176.
characteristics, and acquire is involved in more common disease conditions, such as Still, only a few studies have provided compelling insights
migratory and invasive coronary disease, osteoporosis, chronic kidney disease into the biosynthetic, structural and functional conse-
properties. This occurs during and schizophrenia identifiable by GWAS158. This predic- quences of misexpression of specific glycosyltransferases
embryonic development and
tion was first supported by studies of GALNT2 impli- in cancer, and we limit our discussion to these (of note,
during cancer progression.
cated in regulation of high-density lipoprotein (HDL) we excluded discussion of studies using RNA silencing to
Immune checkpoint and triglycerides important for cardiovascular health159. validate involvement of specific glycosyltransferase genes
An inhibitory pathway in the Individuals with disrupted GALNT2 show lower HDL as these generally require further validation due to ineffi-
immune system that is crucial levels155, which has been recapitulated in several animal ciencies in reduction of enzyme activities and off-target
for maintaining self-tolerance
and modulating the duration
models with loss of function of Galnt2 (ref.160). The GWAS effects). The mechanisms behind altered expression
and amplitude of physiological signal for GALNT2 is located close to a liver-specific may include epigenetic regulation169,177 or the action
immune responses. regulatory element that induces differential allele- of miRNAs178,179. Only a handful of key glycosyltrans-
specific transcription161,162, supporting the idea that ferases currently emerge as consistent players in cancer
Sialyl-Lewisx antigen liver-specific dysregulation of GALNT2 is the cause (Fig. 5). In the N-glycosylation pathway, tetra-antennary
A glycan determinant/epitope
and selectin ligand (NeuAcα2–
of the altered HDL metabolism. GALNT2 serves branching by MGAT5 (ref.135), bisecting GlcNAc by
3Galβ1–4(Fucα1–3) non-redundant O-glycosylation on ANGPTL3 and phos- MGAT3 (ref.180) and core fucosylation by FUT8 (ref.181)
GlcNAcβ1-R) found on pholipid transfer protein (PLTP), two proteins involved have important roles in modulating growth factor recep-
glycoproteins and glycolipids. in regulating HDL. GALNT3 is a GWAS candidate for tor activation and signalling, cell–cell and cell–matrix
bone mineral density163, which is consistent with CDG adhesion receptors182 and immune checkpoint inhibitors183.
loss of function causing hyperphosphataemia and familial A characteristic feature of most cancers is incom-
tumoral calcinosis with ectopic bone formations129,156,164. plete biosynthesis of GalNAc-type O-glycans and
GALNT11, a GWAS candidate for chronic kidney decline, accumulation of immature, short, truncated O-glycan
likely relates to its role in directing O-glycosylation of structures (including Tn and STn antigens) that are
the ligand binding regions of LDLR and LRPs, includ- normally not found on cells. Tn accumulation appears
ing LRP2 (also known as megalin) that serves as the to occur primarily through silencing of COSMC169,170,
major endocytic receptor in the proximal tubules of and this can provide the substrate for ST6GALNAC1 to
the kidney 65. GALNT11 O-glycosylation enhances produce STn184, which cannot be further glycosylated.
ligand affinity of LDLR65, and a mouse Galnt11–/– model Overexpression of the ST6GALNAC1 enzyme in cells
revealed an essential role of O-glycosylation in LRP2 alone may also override O-glycan extension and induce
regulation and kidney function157. STn expression185. These truncated O-glycans are recog-
nized by GBPs including macrophage galactose binding
Dysregulation of glycosyltransferases in cancer. Recent lectin (MGL) and Siglecs, and serve important roles
reviews detail the prevalent glycome changes that equip in modulating tumour immunity185–187. Expression of
cancer cells with distinct glycan features required dur- GALNTs is also commonly altered in cancer188–191; for
ing different stages of tumour growth and dissemination, example, de novo expression of GALNT6 may be directly
including immune evasion and metastasis153,165–167 (Fig. 5). involved in inducing characteristic features of colon can-
Given such common aberrations of glycosylation in can- cer cells including increased proliferation, compromised
cer, it may be surprising that somatic mutations in genes adhesion and loss of normal differentiation192.
controlling cellular glycosylation are extremely rare, and Yet another glycosylation alteration often associated
very few validated mutations in glycosyltransferases have with cancer is the change from Core2 to Core1 O-glycans
been reported in cancer. Mutations in COSMC encoding that can impact interactions with galectins and Siglecs.
a private chaperone required for C1GALT1 that directs This change may be driven by altered expression lev-
GalNAc-type O-glycan Core1 elongation were reported els of GCNT1 and ST3GAL1 and competition for the
in a few cervical cancers168,169, but studies in colorec- common unsialylated Core1 substrate193. Increased
tal and pancreatic cancers found hypermethylation of expression of ST6GAL1 in cancer may lead to a switch
COSMC rather than somatic mutations to be the major from α2-3 to α2-6-sialic acid capping of N-glycans and
reason for the O-glycan truncation that is a hallmark some O-glycans, which can contribute to cancer cell
of epithelial cancers (see also below)169,170. Relatively survival as α2-6-sialylation of tumour necrosis factor
rare heterozygous missense mutations in GALNT12 receptor 1 (TNFR1) inhibits TNF-induced internaliza-
were reported in colorectal cancers, and these were also tion of TNFR1 and prevents death receptor-mediated
shown to affect catalytic properties in recent structural apoptosis194,195. Furthermore, upregulation of FUTs
analysis171. An analysis of data from The Cancer Genome directs selectin-mediated vascular extravasation of
Atlas (TCGA) reveals that cancer genes are more often cancer cells and cancer cell metastasis196. One exam-
mutated (for example, KRAS is mutated in 8% of can- ple here is capping of LacNAc-terminated oligosac-
cers) than typical glycosyltransferases, which have pro- charides by sialyl-Lewisx antigen directed by FUT3 or
tein coding mutations in only around 1% of cancers, FUT6, which has key roles in mediating bone meta­
underlining the rarity of somatic mutations deleterious stasis through E-selectin binding in the vascular niche197.
to the glycosylation machinery. Furthermore, FUT3 together with B3GALT5 generate

www.nature.com/nrm
Reviews

Sialyl-Lewisa antigen
the isomeric sialyl-Lewisa antigen, which has been shown This void is likely a consequence of the great structural
A glycan determinant/epitope to induce hyperactivation of EGFR signalling and lead to complexity and functional (and mechanistic) diversity
and selectin ligand (NeuAcα2– pancreatitis, thereby promoting pancreatic cancer198. of glycans — with few universal and simple rules gov-
3Galβ1–3(Fucα1–4) O-GlcNAcylation and OGT are commonly dysregu- erning the process of glycosylation and the need for
GlcNAcβ1-R) found on
glycoproteins and glycolipids.
lated and have profound regulatory roles in cancer4,199,200. experts demanding analytic technologies to address
Enhanced O-GlcNAcylation is caused by the altered them — which overwhelm much of the scientific com-
Warburg effect cellular metabolism of cancer cells shifting to anaerobic munity and deter from considering glycosylation in their
The capacity for tumours glycolysis (the Warburg effect). This results in increased studies. As enhanced tools and knowledge in the field
to metabolize glucose
glucose flux through the hexosamine biosynthetic path- expand, the opacity of glycoscience is giving way and a
anaerobically when oxygen
is available, increase glucose
way with elevated levels of the UDP–GlcNAc donor plethora of opportunities emerge to explore functions of
uptake and produce lactate. sugar that enhances the activity of the OGT enzyme and protein glycosylation and to improve therapeutic protein
The enhanced flux of increases cellular O-GlcNAcylation. functionalities.
metabolites through the Also, GAGs and the proteoglycans they are attached
hexosamine biosynthetic
pathway may lead to
to show wide roles in cancer as they modulate signal- Genetic engineering of the glycome. Genetic approaches
increased O-GlcNAcylation. ling from growth factors and chemokines through to the study of the glycome and its many functions are
their receptors and affect adhesion between cells and appealing because they can be performed at single-cell
Paucimannose to the extracellular matrix88,89,201. It may be surprising resolution (in contrast to current structural approaches
An N-glycan structure
that the many glycosyltransferase and sulfotransferase that are not yet at the single-cell level) and they employ
consisting of two
N-acetyl-d-glucosamines
genes involved in their biosynthesis have not yet stood commonly available tools in cell biology today. Tools for
(GlcNAcs) and only one to out as having direct functions in cancer. Of note, one single-cell transcriptomics are available and proteom-
three mannose residues GAG epitope widely expressed on malignant cells — a ics are following, and we foresee that such data can be
(core Fuc optional). 4-O-sulfated form of chondroitin sulfate that normally used to develop in silico approaches to transform these
Haploid genetic screen
is restricted to the placenta and appears to be associated data into cellular glycosylation capacities (repertoire
A genetic screen performed with cell proliferation and invasion — does not so far of expressed glycosyltransferases) and enable prediction of
in haploid cells to ease the appear to be regulated by specific changes in expression the architecture of the glycome of a cell (Supplementary
search for recessive functions of the genes of the predicted biosynthetic pathway202,203. Box 1). Such in silico glycomics approaches would be
in mammalian cells.
Overall, cancer cells show prominent alterations to amenable to multi-omics workflows by allowing inte-
their glycome, with functional consequences for cancer gration of the glycome in proteomics and genomics.
pathogenesis. However, it appears that most of these Enabling analysis of glycome information by computa-
changes do not have a genetic basis, and more complex tional transformation of genetic and proteomic informa-
mechanisms seem to be at play in modulating glycosyl- tion will undoubtedly increase the number and diversity
ation in cancer. Perhaps the main cause of aberrancies of studies that address glycosylation and greatly advance
of glycosylation in cancer cells relates to general cellular our insight into regulation and functions of glycosyl-
perturbations associated with malignancy, including ation. We envision the development of an atlas of gly-
disorganization of the secretory pathway, Golgi frag- comes for human cells in the near future, much like the
mentation, ER stress, hypoxia and general metabolic emerging protein and gene expression atlas. Resources
perturbations204–206. Myriad associated effects, such as sprouting from genetic approaches to survey the gly-
ectopic localization of glycosyltransferases207,208, altered come have started to emerge, including collections of
pH homeostasis in the Golgi209 and metabolic availabil- validated, efficient gene targeting constructs for human
ity of donor sugars, impact the glycome in cancer cells. glycosyltransferase genes (GlycoCRISPR214), libraries
General disorganization of the secretory pathway is pre- of isogenic cells with combinatorial engineered glyco­
dicted to have a predominant role in defining the can- sylation capacities that display the glycome on cells
cer glycome. For example, the presence of paucimannose in a tractable manner for interrogation of functions
N-glycans on cell-surface glycoproteins in cancer is sug- (GAGOme203 and GlycoDisplay215) and libraries of
gested to be a result of inappropriate exposure to lyso- mammalian host cells for recombinant production
somal enzymes by as yet unknown ways210. As another of proteins with different glycans for direct testing of
example, relocation of GALNTs from the Golgi to the ER glycan functions216 (Box 1). Such tools offer simple and
was proposed to induce expression of Tn211, and forced direct ways to explore and exploit glycosylation.
expression of GALNT1 in the ER in a mouse liver cancer Genetic dissection of glycosylation pathways and
model strongly enhanced tumour growth through hyper- the biological functions of their products already have
glycosylation and activation of matrix metalloprotease wide support from studies in cell systems217 and model
MMP14 (ref.212). Finally, a recent CRISPR–Cas screen organisms15,16, and with the rapid progress in develop-
revealed that the zinc transporter SLC39A9 is essential ing efficient precise gene editing tools, more systematic
for efficient O-glycosylation as well as N-glycosylation, and global dissection of glycosylation pathways is now
and deficiency in this transporter can lead expression of possible218. A first example illustrating the power of a
the truncated cancer-associated O-glycan Tn213. whole-genome dissection approach to analysing glyco-
sylation uncovered most of the genes required for syn-
Glycome engineering and its prospects thesis of the matriglycan on α-dystroglycan (Fig. 3) with
Despite a rapidly increasing wealth of experimental the use of a haploid genetic screen in human cells219. More
support for diverse functions of protein glycosylation, generally applicable are genome-wide loss-of-function
current insight into specific structure–function rela- screens using CRISPR–Cas9 knockout, which have
tionships and the regulation of these is still fragmented7. identified not only glycosyltransferase genes but also

Nature Reviews | Molecular Cell Biology


Reviews

Box 1 | Next-generation cell-based glycan arrays and combinatorial gene knockout to systematically cover
known individual biosynthetic steps are being employed
Glycan arrays are essential tools for exploring the structural features required for and can take advantage of validated efficient tools. Select
binding to glycans, and they have been instrumental in decoding the informational screens may also be used with organotypic tissue models
content of complex glycans257. Glycan arrays require continued synthesis (or isolation) for dissection of functions of different types of glyco-
of glycans with limitations on size and complexity that can be synthesized, and they
sylation and distinct isoenzymes, as recently shown in
generally present glycans in high densities without the natural context of their protein
or lipid carriers258. to circumvent some of these limitations, it is possible to use a genetic human skin organoids64,225.
‘tree-pruning’ approach — glycotopiary — to engineer and extensively reprogramme More rational engineering of glycosylation capacities
the glycosylation capacities of a human cell line to construct large libraries of isogenic in cells offers opportunities to interrogate established
cell lines that each differ in expression of one or more glycosyltransferases and, hence, genetic and biosynthetic rules with the added potential
display the glycome with loss or gain of distinct glycosylation features215. the creation of surprises for discovery of novel glycosylation path-
of smaller sub-libraries of such engineered cells rationally designed to display specific ways, such as the TMTC-directed O-mannosylation
features of the glycome — for example, elaborated glycans only on select types of discovered by the observation that knockout of the
glycoconjugates (glycoconjugate sub-library), N-glycoproteins with different branching POMT1/2 genes eliminated only select O-mannosylation
(N-glycan sub-library) or glycans with different types of sialic acid capping (sialome capacities27. Libraries of engineered isogenic cells that
sub-library) — enables step-by-step dissection of the importance of these features for
each lack one or more glycosyltransferases can be used
biological interactions226. Cell-based glycan arrays can be interrogated by traditional
cell-binding assays such as flow cytometry, but also with any other types of assay to display parts of the human glycome in the natu-
applicable to cells. the cell-based arrays can be used to express proteins of interest ral environment of the cell surface and the context in
on the cell surface or, when possible, as secreted glycoproteins with different glycan which glycans are naturally found on different types
structures for studying the effects of specific glycan structures. For example, the of glycoconjugates203,215 (Box 1). Such cell-based glycan
cell-based array can be used readily to produce various distinct glycoforms of a libraries may be probed by any assay commonly used
glycoprotein therapeutic that can be tested in preclinical studies for identifying the with cells, and may provide a sustainable self-renewable
optimal design of glycans. importantly, using cell-based glycan arrays for probing resource to survey glycan interactions of GBPs, substi-
biological interactions provides knowledge of the genes required (and not required) tuting analyses with traditional glycan arrays compris-
for display of the specific glycan binding epitope and this result informs on the ing individual immobilized glycans. Whereas studies
biosynthetic basis, which ultimately enables prediction of the structural features of
with immobilized glycans provide information on the
the glycans necessary for the binding interaction studied. the extended information
provided by cell-based glycan arrays enables scientists to readily use genomics and structural requirement for interactions with glycans,
proteomics approaches to further study and dissect identified functions in other studies with the genetic engineered cell libraries pro-
models. Moreover, this information also provides design matrices for engineering other vide information on the biosynthetic and genetic basis
mammalian host cells for recombinant production of glycoprotein therapeutics with of the glycosylation required for the interacting glycans.
desirable and reproducible glycan patterns216,239. The structural features of interacting glycans are pre-
dicted using maps of glycosylation pathways226 (Fig. 3).
This extended information greatly facilitates further
led to the discovery of unexpected genes and functions studies into functions using genetic strategies commonly
required for glycosylation. Examples of the discover- available to non-experts.
ies made with such screens include the demonstration Overall, despite the complexity of the glycome, our
of N-glycan core fucosylation (mediated by FUT8) as knowledge is expanding, mainly owing to the application
a positive regulator of cell-surface expression of the of ever-more sophisticated analytical techniques. We
immune checkpoint receptor on T cells183, the require- predict an explosion in genetic dissection applications
ment of sialic acid biosynthesis (with a role of sialic acid including not only disruption of genes but also gene
transporter SLC35A1) for avian H5N1 influenza virus activation. Popularization of such tools and continued
replication220 and the role of tetra-antennary N-glycans progress in this space now hold promise for the devel-
(instigated by MGAT5) for CD8+ T cell-mediated glio- opment of an atlas of cellular glycosylation and glycomes
blastoma tumour-cell killing221. The genetic approach is of all human cells in the near future, which could then
particularly useful in dissecting the structural features be explored — with readily available genetic dissection
that guide interactions with GAGs and confer their func- tools — for gaining functional insight into the glycome.
tions due to difficulties in sequencing the GAG poly-
saccharide chains and structural determination of such Glycoengineering of therapeutics. Glycans are often
binding motifs203,222. One example here is the demon- regarded as ‘necessary evils’ in biologics production
stration of the role of specific GAG chains on heparan pipelines. The freedom to perform extensive engineer-
sulfate in the cellular uptake of Tau protein223. ing of glycosylation capacities in mammalian host cells
Despite the various fruitful insights, a limitation provides wide opportunities for the improvement and
of genetic screens is identifying glycosylation features optimization of current biologics production pipelines
covered by isoenzyme redundancies. For example, a and for the development of custom-designed glycopro-
screen for avian H5N1 influenza virus replication medi- tein therapeutics216. Engineering glycans on therapeu-
ators only identified the sialic acid transporter as being tic proteins has been pursued for decades by chemical,
Tau protein involved in the process, but not any of the sialyltrans- chemo-enzymatic and genetic approaches in differ-
Tubulin binding protein ferases and other genes expected to be involved in the ent cell types (Escherichia coli, yeast, plant, insect and
involved in neurodegenerative sialylation pathway224. In this case, focused screens with mammalian227–230), but engineering cells already tooled
diseases called tauopathies
where Tau aggregates into
combinatorial targeting of multiple isoenzymes covering for human glycosylation provide a simpler and more
intracellular neurofibrillary redundant steps are needed to identify the candidate iso- versatile starting point and opportunities for develop-
tangles. enzyme genes involved. Targeted screens with individual ing more complex and physiologically relevant designs

www.nature.com/nrm
Reviews

Native mass spectrometry


as well as novel designs offering improved pharmacoki- lysosomal storage disorder, Fabry disease, and found,
An analytical technique netic and pharmacodynamic properties. Availability of surprisingly, that glycodesigns without exposed man-
used to study non-denatured comprehensive cell libraries of isogenic cells with dif- nose, but capped with α2-3-linked sialic acid, showed
proteoforms and protein ferent glycosylation capacities that enable production improved circulation and biodistribution with efficient
complexes in the gas phase.
of glycoproteins with controlled diversity in glycan reduction of accumulated glycolipid species in a mouse
Glucocerebrosidase structures attached should greatly facilitate advances model239. Glycoengineering may also be used to improve
(GBA). A lysosomal enzyme that in this respect, and the increased understanding of the immunogenicity of recombinant produced vaccines by
hydrolyses glucosylceramide outcomes of genetic engineering are providing relia- introducing glycan structures known to target immune
(GlcCer) cerobroside into
ble design matrices for custom-designed glycoprotein regulatory lectin receptors240,241, and for example, to
glucose and free ceramide.
therapeutics216 (Box 1). better mimic high-mannose glycans often found on
Gaucher disease The primary objectives for glycoengineering are viral envelope glycoproteins and recognized by broadly
A rare genetic lysosomal desires to produce safe, consistent and efficient biolog- neutralizing monoclonal antibodies242.
storage disease caused by ics. Species differences in glycosylation and immunity In addition to approaches in cells, glycoengineering
mutations in the GBA gene
encoding glucocerebrosidase
to non-human glycans are posing challenges for use of of entire animals to remove immunogenic glycans for
(GBA). Damaging mutations non-human cell lines for production of biologics. The organ sourcing for human transplants has a long his-
in GBA cause progressive Chinese hamster ovary (CHO) cell line is currently tory, with approaches to remove the strongly immuno­
accumulation of the glycolipid the lead production host for human protein biologics genic αGal epitope widely expressed on non-primate
glucosylceramide (GlcCer)
because of its simple human-like glycosylation capaci- mammalian cells and, recently, the generation of a
substrate for the enzyme with
damaging effects on tissues
ties, but heterogeneity of the glycans found on recom- triple-knockout pig with elimination of three immuno-
and organs. Enzyme binant CHO-expressed glycoproteins still challenges genic glycoepitopes, αGal, N-glycolylneuraminic acid
replacement therapy is regulatory demands for consistency and imposes exten- (Neu5Gc) and terminal GalNAc243. Although increased
currently the treatment sive analytic efforts to produce pure glycoprotein species immunotolerance achieved by these approaches may not
of choice for this disease.
with the desired properties231. However, with engineered solve all issues with xenotransplantation, it still holds
α-Galactosidase homogeneous glycosylation to reduce proteoform com- great promise for more specific uses of animal tissues
(GLA). A lysosomal enzyme plexity, the downstream analysis can be largely simpli- in human medicine, such as for heart valve curation
that hydrolyses α-galactose fied and perhaps reduced to native mass spectrometry232, and as acellular dermal matrices for reconstructive sur-
from glycolipids and
which improves quality control of biologics. gery. Currently, porcine dermal matrices treated with an
glycoproteins.
The first glycoengineered biologic agent approved for effective α-galactosidase to remove αGal epitopes244 are
Fabry disease clinical application was the lysosomal glucocerebrosidase in wide clinical use (Strattice™)245.
A rare, genetic X-linked (GBA) used for treatment of the lysosomal storage disor-
lysosomal storage disease der, Gaucher disease. GBA was engineered to remove M6P Conclusions and perspective
caused by damaging
mutations in the GLA gene
from its N-glycans that mediates uptake by Man-6-P Most proteins are glycosylated through 1 or more of
encoding α-galactosidase A, receptor233, and instead expose Man residues to achieve 14 distinct glycosylation pathways that in humans involve
which catalyses the efficient mannose receptor-mediated uptake by macro­ at least 173 glycosyltransferases as well as many other
hydrolysis of glycolipid phages, which are the main cell type affected by GBA enzymes that modify glycans. Glycosylation produces
globotriacylceramide (Gb3).
storage defect, and efficient targeting to lysosomes233. a vastly larger number of glycan structures through
αGal epitope
Major progress in glycoengineering has been seen in combinatorial use of enzymes and of repeated com-
A Galα1–3Galβ1–4GlcNAc-R the area of immunotherapy for cancer142. Engineering mon scaffolds. As such, glycosylation of proteins clearly
glycan epitope (also known as therapeutic IgG antibodies with low core fucose on the provides the greatest and most diverse expansion of the
the Galili antigen) found on Fc N-glycan234,235 to increase antibody-dependent cell proteome. This complexity, compounded by analytical
glycoproteins and glycolipids
of non-primate mammals and
cytotoxicity is a major advancement and such antibod- limitations (sensitivity, site-specific glycan analysis of
new world monkeys, but ies are now used in cancer therapies142. Unnatural short glycoproteins and sequencing of GAGs), has for many
absent in humans, apes and trisaccharide N-glycans have been engineered by intro- years been challenging to capture and dissect func-
old world monkeys because duction of an endo-β-N-acetylglucosaminidase (endoT) tionally, and studies of the glycome have lagged behind
of an inactivated GGTA1 gene.
in the Golgi to serve a similar purpose236. A novel ele- studies of other omes. However, protein glycosylation
gant strategy using installation of inducible expression pathways are now sufficiently outlined to start applying
of glycosyltransferase genes (FUT8 and B4GALT1) systems biology approaches to explore regulation of the
has also shown the ability to fine-tune control of IgG glycome through quantitative transcriptomics and/or
N-glycosylation 237. Finally, extensive engineering proteomics data, opening new avenues in understanding
of N-glycosylation capacities demonstrates that multiple structure–function relationships of protein glycosylation.
homogeneous glycoforms can be produced238. Genetic approaches to studying assembly and func-
Relying only on endogenous glycosylation capac- tions of the glycome have a long history. The new gene
ities of mammalian cells severely limits the discovery editing tools are now allowing more comprehensive
potential for novel, improved glycan designs for thera- genetic dissection and genome-wide screens of func-
peutics. However, with engineering, there are now only tions of glycans, generation of cell-based glycan dis-
a few limitations in achievable designs and options for play platforms for interrogation of glycan interactions
testing these203,216. Stably engineered CHO and HEK293 and functions, and custom design of glycoproteins —
cells offer production hosts for a wide range of glyco- advances that will ignite wider integration of glyco-
forms for experimental screening and discovery of sylation in cell biology and systems biology. From a
new improved designs. Inspired by the above-noted translational standpoint, these advances provide oppor-
success for Gaucher disease233, we engineered variants tunities to discover and test improved drug and vac-
of α-galactosidase used for the treatment of another cine designs95,240. To fully benefit from these resources,

Nature Reviews | Molecular Cell Biology


Reviews

it is important to develop approaches to efficiently and pathways may provide sufficient foundation for devel-
reproducibly translate cellular glycosylation capacities oping global predictions of cellular glycosylation capac-
(transcriptome/proteome information) into the archi- ities and the resulting glycome at the single-cell level
tecture of the glycome generated (Supplementary Box 1). based on transcriptomics and proteomics data — an
We are now in an era where it is simpler — and per- era that could be designated in silico glycomics. We
haps more informative — to use genetic approaches to envision the development of a comprehensive atlas of
study protein glycosylation and functions, rather than cellular glycomes across the entire human body and
traditional analysis of the structures of glycans on pro- integration of protein glycosylation features in gene
teins, but considerable experimentation and validation and protein databases.
of these approaches are still needed. We posit that the
Published online xx xx xxxx
biosynthetic and genetic rules for human glycosylation

1. Fournet, M., Bonté, F. & Desmoulière, A. Glycation 23. Kornfeld, R. & Kornfeld, S. Assembly of asparagine- 41. Snider, M. D. & Rogers, O. C. Intracellular movement of
damage: a possible hub for major pathophysiological linked oligosaccharides. Annu. Rev. Biochem. 54, cell surface receptors after endocytosis: resialylation
disorders and aging. Aging Dis. 9, 880–900 (2018). 631–664 (1985). of asialo-transferrin receptor in human erythroleukemia
2. Steentoft, C. et al. Precision mapping of the human 24. Narimatsu, H. Human glycogene cloning: focus on cells. J. Cell Biol. 100, 826–834 (1985).
O-GalNAc glycoproteome through SimpleCell β3-glycosyltransferase and β4-glycosyltransferase 42. Duncan, J. R. & Kornfeld, S. Intracellular movement
technology. EMBO J. 32, 1478–1488 (2013). families. Curr. Opin. Struct. Biol. 16, 567–575 of two mannose 6-phosphate receptors: return to the
This paper presents a deep analysis of the human (2006). Golgi apparatus. J. Cell Biol. 106, 617–628 (1988).
GalNAc-type O-glycoproteome. 25. Bennett, E. P. et al. Control of mucin-type 43. Litvinov, S. V. & Hilkens, J. The epithelial sialomucin,
3. Zielinska, D. F., Gnad, F., Wiśniewski, J. R. & Mann, M. O-glycosylation: a classification of the polypeptide episialin, is sialylated during recycling. J. Biol. Chem.
Precision mapping of an in vivo N-glycoproteome GalNAc-transferase gene family. Glycobiology 22, 268, 21364–21371 (1993).
reveals rigid topological and sequence constraints. 736–756 (2012). 44. Razawi, H. et al. Evidence for Core 2 to Core 1 O-glycan
Cell 141, 897–907 (2010). 26. Tsuji, S., Datta, A. K. & Paulson, J. C. Systematic remodeling during the recycling of MUC1. Glycobiology
This paper presents a deep analysis of N-glycosites nomenclature for sialyltransferases. Glycobiology 6, 23, 935–45 (2013).
in the human proteome. v–vii (1996). 45. Gilmour, A. M. et al. A novel epidermal growth
4. Hart, G. W. Nutrient regulation of signaling and 27. Larsen, I. S. B. et al. Discovery of an O-mannosylation factor receptor-signaling platform and its targeted
transcription. J. Biol. Chem. 294, 2211–2231 (2019). pathway selectively serving cadherins and translation in pancreatic cancer. Cell Signal. 25,
5. Hart, G. W. et al. Glycosylation of Nuclear and protocadherins. Proc. Natl Acad. Sci. USA 114, 2587–2603 (2013).
Cytoplasmic Proteins is as Abundant and as Dynamic 11163–11168 (2017). 46. Haxho, F., Neufeld, R. J. & Szewczuk, M. R.
as Phosphorylation. in Glyco- and Cellbiology (eds 28. Yoshida-Moriguchi, T. & Campbell, K. P. Matriglycan: Neuraminidase-1: a novel therapeutic target
Wieland, F. & Reutter, W.) 91–103 (Springer, 1994). a novel polysaccharide that links dystroglycan to the in multistage tumorigenesis. Oncotarget 7,
6. Aebersold, R. et al. How many human proteoforms basement membrane. Glycobiology 25, 702–713 40860–40881 (2016).
are there? Nat. Chem. Biol. 14, 206–214 (2018). (2015). 47. Lillehoj, E. P. et al. NEU1 sialidase expressed in
7. Varki, A. Biological roles of glycans. Glycobiology 27, 29. Praissman, J. L. et al. The functional O-mannose human airway epithelia regulates epidermal growth
3–49 (2017). glycan on α-dystroglycan contains a phospho-ribitol factor receptor (EGFR) and MUC1 protein signaling.
8. Spiro, R. G. Protein glycosylation: nature, distribution, primed for matriglycan addition. eLife 5, e14473 J. Biol. Chem. 287, 8214–8231 (2012).
enzymatic formation, and disease implications of (2016). 48. Welch, L. G. & Munro, S. A tale of short tails, through
glycopeptide bonds. Glycobiology 12, 43R–56R 30. Hirata, T. et al. Identification of a Golgi GPI-N- thick and thin: investigating the sorting mechanisms
(2002). acetylgalactosamine transferase with tandem of Golgi enzymes. FEBS Lett. 593, 2452–2465 (2019).
9. Cummings, R. D. The repertoire of glycan determinants transmembrane regions in the catalytic domain. 49. Kellokumpu, S., Hassinen, A. & Glumoff, T.
in the human glycome. Mol. Biosyst. 5, 1087–1104 Nat. Commun. 9, 405 (2018). Glycosyltransferase complexes in eukaryotes:
(2009). 31. Cejas, R. B., Lorenz, V., Garay, Y. C. & Irazoqui, F. J. long-known, prevalent but still unrecognized.
10. Joshi, H. J. et al. SnapShot: O-glycosylation pathways Biosynthesis of O-N-acetylgalactosamine glycans Cell. Mol. Life Sci. 73, 305–325 (2016).
across kingdoms. Cell 172, 632–632.e2 (2018). in the human cell nucleus. J. Biol. Chem. 294, 50. Moremen, K. W. & Haltiwanger, R. S. Emerging
11. Springer, S. A. & Gagneux, P. Glycomics: revealing 2997–3011 (2019). structural insights into glycosyltransferase-mediated
the dynamic ecology and evolution of sugar molecules. 32. Tu, L., Chen, L. & Banfield, D. K. A conserved synthesis of glycans. Nat. Chem. Biol. 15, 853–864
J. Proteom. 135, 90–100 (2016). N-terminal arginine-motif in GOLPH3-family proteins (2019).
12. Chou, H. H. et al. A mutation in human CMP-sialic acid mediates binding to coatomer. Traffic 13, 1496–1507 51. de Las Rivas, M., Lira-Navarrete, E., Gerken, T. A. &
hydroxylase occurred after the Homo-Pan divergence. (2012). Hurtado-Guerrero, R. Polypeptide GalNAc-Ts: from
Proc. Natl Acad. Sci. USA 95, 11751–11756 (1998). 33. Liu, L., Doray, B. & Kornfeld, S. Recycling of Golgi redundancy to specificity. Curr. Opin. Struct. Biol. 56,
13. Larsen, R. D., Rivera-Marrero, C. A., Ernst, L. K., glycosyltransferases requires direct binding to 87–96 (2019).
Cummings, R. D. & Lowe, J. B. Frameshift and coatomer. Proc. Natl Acad. Sci. USA 115, 8984–8989 52. Taujale, R. et al. Deep evolutionary analysis reveals
nonsense mutations in a human genomic sequence (2018). the design principles of fold A glycosyltransferases.
homologous to a murine UDP-Gal:β-d-Gal(1,4)- 34. Kuhn, P.-H. et al. Secretome analysis identifies novel eLife 9, e54532 (2020).
d-GlcNAc α(1,3)-galactosyltransferase cDNA. signal peptide peptidase-like 3 (Sppl3) substrates and 53. Kinoshita, T. & Fujita, M. Biosynthesis of GPI-anchored
J. Biol. Chem. 265, 7055–7061 (1990). reveals a role of Sppl3 in multiple Golgi glycosylation proteins: special emphasis on GPI lipid remodeling.
14. Christiansen, D. et al. Humans lack iGb3 due to the pathways. Mol. Cell. Proteom. 14, 1584–1598 J. Lipid Res. 57, 6–24 (2016).
absence of functional iGb3-synthase: implications for (2015). 54. Wild, R. et al. Structure of the yeast
NKT cell development and transplantation. PLoS Biol. 35. Schjoldager, K. T.-B. G. et al. A systematic study of oligosaccharyltransferase complex gives insight into
6, e172 (2008). site-specific GalNAc-type O-glycosylation modulating eukaryotic N-glycosylation. Science 359, 545–550
15. Stanley, P. What have we learned from proprotein convertase processing. J. Biol. Chem. 286, (2018).
glycosyltransferase knockouts in mice? J. Mol. Biol. 40122–40132 (2011). 55. Ruiz-Canada, C., Kelleher, D. J. & Gilmore, R.
428, 3166–3182 (2016). 36. Shifley, E. T. & Cole, S. E. Lunatic fringe protein Cotranslational and posttranslational N-glycosylation
16. Lowe, J. B. & Marth, J. D. A genetic approach to processing by proprotein convertases may contribute of polypeptides by distinct mammalian OST isoforms.
mammalian glycan function. Annu. Rev. Biochem. 72, to the short protein half-life in the segmentation Cell 136, 272–283 (2009).
643–691 (2003). clock. Biochim. Biophys. Acta 1783, 2384–2390 56. Cherepanova, N. A. & Gilmore, R. Mammalian cells
17. Freeze, H. H., Chong, J. X., Bamshad, M. J. & (2008). lacking either the cotranslational or posttranslocational
Ng, B. G. Solving glycosylation disorders: fundamental 37. Paulson, J. C. & Colley, K. J. Glycosyltransferases. oligosaccharyltransferase complex display substrate-
approaches reveal complicated pathways. Structure, localization, and control of cell type-specific dependent defects in asparagine linked glycosylation.
Am. J. Hum. Genet. 94, 161–175 (2014). glycosylation. J. Biol. Chem. 264, 17615–17618 Sci. Rep. 6, 20946 (2016).
18. Jaeken, J. & Péanne, R. What is new in CDG? (1989). 57. Ramírez, A. S., Kowal, J. & Locher, K. P.
J. Inherit. Metab. Dis. 40, 569–586 (2017). 38. Liefhebber, J. M., Punt, S., Spaan, W. J. & Cryo–electron microscopy structures of human
19. Moremen, K. W., Tiemeyer, M. & Nairn, A. V. van Leeuwen, H. C. The human collagen β(1-O) oligosaccharyltransferase complexes OST-A and
Vertebrate protein glycosylation: diversity, synthesis galactosyltransferase, GLT25D1, is a soluble OST-B. Science 366, 1372–1375 (2019).
and function. Nat. Rev. Mol. Cell Biol. 13, 448–462 endoplasmic reticulum localized protein. BMC Cell 58. Harada, Y., Masahara-Negishi, Y. & Suzuki, T.
(2012). Biol. 11, 33 (2010). Cytosolic-free oligosaccharides are predominantly
20. Joshi, H. J. et al. Glycosyltransferase genes that cause 39. Harvey, B. M. & Haltiwanger, R. S. Regulation of generated by the degradation of dolichol-linked
monogenic congenital disorders of glycosylation are Notch function by O-glycosylation. Adv. Exp. Med. oligosaccharides in mammalian cells. Glycobiology
distinct from glycosyltransferase genes associated with Biol. 1066, 59–78 (2018). 25, 1196–1205 (2015).
complex diseases. Glycobiology 28, 284–294 (2018). 40. Ogawa, M. et al. GTDC2 modifies O-mannosylated 59. Lu, H. et al. Mammalian STT3A/B oligosaccharyl­
21. Hansen, L. et al. A mutation map for human glycoside α-dystroglycan in the endoplasmic reticulum to transferases segregate N-glycosylation at the
hydrolase genes. Glycobiology 30, 500–515 (2020). generate N-acetyl glucosamine epitopes reactive with translocon from lipid-linked oligosaccharide
22. Cummings, R. D. & Pierce, J. M. The challenge and CTD110.6 antibody. Biochem. Biophys. Res. Commun. hydrolysis. Proc. Natl Acad. Sci. USA 115,
promise of glycomics. Chem. Biol. 21, 1–15 (2014). 440, 88–93 (2013). 9557–9562 (2018).

www.nature.com/nrm
Reviews

60. Shan, A. et al. Polypeptide N-acetylgalactosaminyl­ 81. Scietti, L. et al. Molecular architecture of the acid in nervous system development, stability, disease,
transferase 18 non-catalytically regulates the ER multifunctional collagen lysyl hydroxylase and and regeneration. Physiol. Rev. 94, 461–518 (2014).
homeostasis and O-glycosylation. Biochim. Biophys. glycosyltransferase LH3. Nat. Commun. 9, 3163 107. Bhide, G. P., Prehna, G., Ramirez, B. E. & Colley, K. J.
Acta Gen. Subj. 1863, 870–882 (2019). (2018). The polybasic region of the polysialyltransferase
61. Joshi, H. J. et al. GlycoDomainViewer: a bioinformatics 82. Slawson, C. & Hart, G. W. O-GlcNAc signalling: ST8Sia-IV binds directly to the neural cell adhesion
tool for contextual exploration of glycoproteomes. implications for cancer cell biology. Nat. Rev. Cancer molecule, NCAM. Biochemistry 56, 1504–1517
Glycobiology 28, 131–136 (2018). 11, 678–684 (2011). (2017).
62. Schjoldager, K. T. et al. Deconstruction of 83. Lazarus, M. B., Nam, Y., Jiang, J., Sliz, P. & Walker, S. 108. Bhide, G. P., Fernandes, N. R. J. & Colley, K. J.
O-glycosylation–GalNAc-T isoforms direct distinct Structure of human O-GlcNAc transferase and its Sequence requirements for neuropilin-2 recognition
subsets of the O-glycoproteome. EMBO Rep. 16, complex with a peptide substrate. Nature 469, by ST8SiaIV and polysialylation of its O-glycans.
1713–1722 (2015). 564–567 (2011). J. Biol. Chem. 291, 9444–9457 (2016).
63. Narimatsu, Y. et al. Exploring regulation of protein 84. Tsuji, S., Datta, A. K. & Paulson, J. C. Systematic 109. Marcus, D. M. & Cass, L. E. Glycosphingolipids
O-glycosylation in isogenic human HEK293 cells by nomenclature for sialyltransferases. Glycobiology 6, with Lewis blood group activity: uptake by human
differential O-glycoproteomics. Mol. Cell. Proteom. v–vii (1996). erythrocytes. Science 164, 553–555 (1969).
18, 1396–1409 (2019). 85. Oriol, R., Mollicone, R., Cailleau, A., Balanzino, L. & 110. Parry, S. et al. The sperm agglutination antigen-1
64. Bagdonaite, I. et al. O-glycan initiation directs Breton, C. Divergent evolution of fucosyltransferase (SAGA-1) glycoforms of CD52 are O-glycosylated.
distinct biological pathways and controls epithelial genes from vertebrates, invertebrates, and bacteria. Glycobiology 17, 1120–1126 (2007).
differentiation. EMBO Rep. 21, e48885 (2020). Glycobiology 9, 323–334 (1999). 111. Wandall, H. H. et al. The origin and function of platelet
65. Wang, S. et al. Site-specific O-glycosylation of 86. Nagae, M., Yamaguchi, Y., Taniguchi, N. & Kizuka, Y. glycosyltransferases. Blood 120, 626–635 (2012).
members of the low-density lipoprotein receptor 3D structure and function of glycosyltransferases 112. Manhardt, C. T., Punch, P. R., Dougher, C. W. L. &
superfamily enhances ligand interactions. involved in N-glycan maturation. Int J. Mol. Sci. 21, Lau, J. T. Y. Extrinsic sialylation is dynamically
J. Biol. Chem. 293, 7408–7422 (2018). 437 (2020). regulated by systemic triggers in vivo. J. Biol. Chem.
This paper describes a role of site-specific 87. Honke, K. & Taniguchi, N. Sulfotransferases and 292, 13514–13520 (2017).
O-glycosylation in regulation of the affinity sulfated oligosaccharides. Med. Res. Rev. 22, 113. Jones, M. B. et al. B-cell-independent sialylation
of LDLR-related receptors. 637–654 (2002). of IgG. Proc. Natl Acad. Sci. USA 113, 7207–7212
66. Tagliabracci, V. S. et al. A single kinase generates the 88. Esko, J. D. & Selleck, S. B. Order out of chaos: (2016).
majority of the secreted phosphoproteome. Cell 161, assembly of ligand binding sites in heparan sulfate. 114. Irons, E. E. et al. B cells suppress medullary
1619–1632 (2015). Annu. Rev. Biochem. 71, 435–471 (2002). granulopoiesis by an extracellular glycosylation-
67. Tagliabracci, V. S. et al. Dynamic regulation of FGF23 89. Mikami, T. & Kitagawa, H. Biosynthesis and function dependent mechanism. eLife 8, e47328 (2019).
by Fam20C phosphorylation, GalNAc-T3 glycosylation, of chondroitin sulfate. Biochim. Biophys. Acta 1830, 115. Zhang, Q. et al. Transfer of functional cargo in
and furin proteolysis. Proc. Natl Acad. Sci. USA 111, 4719–4733 (2013). exomeres. Cell Rep. 27, 940–954.e6 (2019).
5520–5 (2014). 90. Kitayama, K., Hayashida, Y., Nishida, K. & Akama, T. O. 116. Lu, Q., Li, S. & Shao, F. Sweet talk: protein
This paper is the first demonstration of cross-talk Enzymes responsible for synthesis of corneal keratan glycosylation in bacterial interaction with the
between extracellular protein phosphorylation and sulfate glycosaminoglycans. J. Biol. Chem. 282, host. Trends Microbiol. 23, 630–641 (2015).
O-glycosylation. 30085–30096 (2007). 117. El Qaidi, S. et al. NleB/SseK effectors from Citrobacter
68. Bordoli, M. R. et al. A secreted tyrosine kinase acts in 91. Yoshida-Moriguchi, T. et al. SGK196 is a glycosylation- rodentium, Escherichia coli, and Salmonella enterica
the extracellular environment. Cell 158, 1033–1044 specific O-mannose kinase required for dystroglycan display distinct differences in host substrate specificity.
(2014). function. Science 341, 896–899 (2013). J. Biol. Chem. 292, 11423–11430 (2017).
69. Mehta, A. Y., Heimburg-Molinaro, J., Cummings, R. D. 92. Sheikh, M. O., Halmo, S. M. & Wells, L. Recent 118. Li, S. et al. Pathogen blocks host death receptor
& Goth, C. K. Emerging patterns of tyrosine sulfation advancements in understanding mammalian signalling by arginine GlcNAcylation of death domains.
and O-glycosylation cross-talk and co-localization. O-mannosylation. Glycobiology 27, 806–819 (2017). Nature 501, 242–246 (2013).
Curr. Opin. Struct. Biol. 62, 102–111 (2020). 93. Koike, T., Izumikawa, T., Tamura, J.-I. & Kitagawa, H. 119. Helenius, A. & Aebi, M. Roles of N-linked glycans in
70. Yu, H. & Takeuchi, H. Protein O-glucosylation: FAM20B is a kinase that phosphorylates xylose the endoplasmic reticulum. Annu. Rev. Biochem. 73,
another essential role of glucose in biology. in the glycosaminoglycan–protein linkage region. 1019–1049 (2004).
Curr. Opin. Struct. Biol. 56, 64–71 (2019). Biochem. J. 421, 157–162 (2009). 120. Vasudevan, D., Takeuchi, H., Johar, S. S., Majerus, E.
71. Holdener, B. C. & Haltiwanger, R. S. Protein 94. Wen, J. et al. Xylose phosphorylation functions & Haltiwanger, R. S. Peters plus syndrome mutations
O-fucosylation: structure and function. Curr. Opin. as a molecular switch to regulate proteoglycan disrupt a noncanonical ER quality-control mechanism.
Struct. Biol. 56, 78–86 (2019). biosynthesis. Proc. Natl Acad. Sci. USA 111, Curr. Biol. 25, 286–295 (2015).
72. Ogawa, M. & Okajima, T. Structure and function of 15723–15728 (2014). 121. Takeuchi, H. et al. O-Glycosylation modulates the
extracellular O-GlcNAc. Curr. Opin. Struct. Biol. 56, 95. Duan, S. & Paulson, J. C. Siglecs as immune cell stability of epidermal growth factor-like repeats and
72–77 (2019). checkpoints in disease. Annu. Rev. Immunol. 38, thereby regulates Notch trafficking. J. Biol. Chem.
73. Takeuchi, H. et al. Two novel protein 365–395 (2020). 292, 15964–15973 (2017).
O-glucosyltransferases that modify sites distinct from 96. Baumann, A.-M. T. et al. 9-O-Acetylation of sialic acids 122. Ogawa, M., Tashima, Y., Sakaguchi, Y., Takeuchi, H.
POGLUT1 and affect Notch trafficking and signaling. is catalysed by CASD1 via a covalent acetyl-enzyme & Okajima, T. Contribution of extracellular O-GlcNAc
Proc. Natl Acad. Sci. USA 115, E8395–E8402 intermediate. Nat. Commun. 6, 7673 (2015). to the stability of folded epidermal growth factor-like
(2018). 97. Orizio, F. et al. Human sialic acid acetyl esterase: domains and Notch1 trafficking. Biochem. Biophys.
This paper describes the identification and towards a better understanding of a puzzling Res. Commun. 526, 184–190 (2020).
differential functions of POGLUT isoenzymes in enzyme. Glycobiology 25, 992–1006 (2015). 123. Goth, C. K., Vakhrushev, S. Y., Joshi, H. J., Clausen, H.
glycosylation of NOTCH EGF-like repeats and their 98. Vlasak, R., Luytjes, W., Spaan, W. & Palese, P. & Schjoldager, K. T. Fine-tuning limited proteolysis:
regulation of NOTCH functions. Human and bovine coronaviruses recognize sialic a major role for regulated site-specific O-glycosylation.
74. Sakaidani, Y. et al. O-linked-N-acetylglucosamine acid-containing receptors similar to those of influenza Trends Biochem. Sci. 43, 269–284 (2018).
modification of mammalian Notch receptors by C viruses. Proc. Natl Acad. Sci. USA 85, 4526–4529 124. Goth, C. K. et al. A systematic study of modulation of
an atypical O-GlcNAc transferase Eogt1. Biochem. (1988). ADAM-mediated ectodomain shedding by site-specific
Biophys. Res. Commun. 419, 14–19 (2012). 99. Barb, A. W. & Prestegard, J. H. NMR analysis O-glycosylation. Proc. Natl Acad. Sci. USA 112,
75. Manya, H. et al. Demonstration of mammalian demonstrates immunoglobulin G N-glycans are 14623–14628 (2015).
protein O-mannosyltransferase activity: coexpression accessible and dynamic. Nat. Chem. Biol. 7, 147–153 125. Goth, C. K. et al. Site-specific O-glycosylation by
of POMT1 and POMT2 required for enzymatic (2011). polypeptide N-acetylgalactosaminyltransferase 2
activity. Proc. Natl Acad. Sci. USA 101, 500–505 100. Krapp, S., Mimura, Y., Jefferis, R., Huber, R. & (GalNAc-transferase T2) co-regulates β1-adrenergic
(2004). Sondermann, P. Structural analysis of human IgG-Fc receptor N-terminal cleavage. J. Biol. Chem. 292,
76. Neubert, P. et al. Mapping the O-mannose glycoforms reveals a correlation between glycosylation 4714–4726 (2017).
glycoproteome in Saccharomyces cerevisiae. and structural integrity. J. Mol. Biol. 325, 979–989 126. Hansen, L. H. et al. Discovery of O-glycans on
Mol. Cell. Proteom. 15, 1323–1337 (2016). (2003). atrial natriuretic peptide (ANP) that affect both its
77. Shcherbakova, A., Tiemann, B., Buettner, F. F. R. & 101. Bowden, T. A. et al. Chemical and structural analysis proteolytic degradation and potency at its cognate
Bakker, H. Distinct C-mannosylation of netrin receptor of an antibody folding intermediate trapped during receptor. J. Biol. Chem. 294, 12567–12578
thrombospondin type 1 repeats by mammalian glycan biosynthesis. J. Am. Chem. Soc. 134, (2019).
DPY19L1 and DPY19L3. Proc. Natl Acad. Sci. USA 17554–17563 (2012). 127. Madsen, T. D. et al. An atlas of O-linked glycosylation
114, 2574–2579 (2017). 102. Patel, K. R., Roberts, J. T. & Barb, A. W. Multiple on peptide hormones reveals diverse biological roles.
This paper describes the DPY19L isoenzymes variables at the leukocyte cell surface impact Fcγ Nat. Commun. 11, 4033 (2020).
directing C-mannosylation and identifies distinct receptor-dependent mechanisms. Front. Immunol. 10, 128. Hintze, J. et al. Probing the contribution of
differences in their substrate preferences. 223 (2019). individual polypeptide GalNAc-transferase isoforms
78. Roch, C., Kuhn, J., Kleesiek, K. & Götting, C. 103. Ye, Z., Mao, Y., Clausen, H. & Vakhrushev, S. Y. to the O-glycoproteome by inducible expression in
Differences in gene expression of human Glyco-DIA: a method for quantitative isogenic cell lines. J. Biol. Chem. 293, 19064–19077
xylosyltransferases and determination of O-glycoproteomics with in silico-boosted glycopeptide (2018).
acceptor specificities for various proteoglycans. libraries. Nat. Methods 16, 902–910 (2019). 129. Kato, K. et al. Polypeptide GalNAc-transferase T3
Biochem. Biophys. Res. Commun. 391, 685–691 104. Kornfeld, S. Trafficking of lysosomal enzymes in normal and familial tumoral calcinosis. Secretion of fibroblast
(2010). and disease states. J. Clin. Invest. 77, 1–6 (1986). growth factor 23 requires O-glycosylation. J. Biol. Chem.
79. Noborn, F. et al. Identification of chondroitin sulfate 105. Reitman, M. L. & Kornfeld, S. Lysosomal enzyme 281, 18370–18377 (2006).
linkage region glycopeptides reveals prohormones as targeting. N-Acetylglucosaminylphosphotransferase 130. Takashi, Y. et al. Activation of unliganded FGF
a novel class of proteoglycans. Mol. Cell. Proteom. 14, selectively phosphorylates native lysosomal enzymes. receptor by extracellular phosphate potentiates
41–49 (2015). J. Biol. Chem. 256, 11977–11980 (1981). proteolytic protection of FGF23 by its O-glycosylation.
80. Hennet, T. Collagen glycosylation. Curr. Opin. Struct. 106. Schnaar, R. L., Gerardy-Schahn, R. & Hildebrandt, H. Proc. Natl Acad. Sci. USA 116, 11418–11427
Biol. 56, 131–138 (2019). Sialic acids in the brain: gangliosides and polysialic (2019).

Nature Reviews | Molecular Cell Biology


Reviews

131. Larsen, I. S. B., Narimatsu, Y., Clausen, H., Joshi, H. J. 154. Ng, B. G. & Freeze, H. H. Perspectives on glycosylation 179. Kurcon, T. et al. miRNA proxy approach reveals hidden
& Halim, A. Multiple distinct O-mannosylation and its congenital disorders. Trends Genet. 34, functions of glycosylation. Proc. Natl Acad. Sci. USA
pathways in eukaryotes. Curr. Opin. Struct. Biol. 56, 466–476 (2018). 112, 7327–7332 (2015).
171–178 (2019). 155. Zilmer, M. et al. Novel congenital disorder of O-linked 180. Bhattacharyya, R., Bhaumik, M., Raju, T. S. & Stanley, P.
132. Rexach, J. E. et al. Dynamic O-GlcNAc modification glycosylation caused by loss of function of GALNT2. Truncated, inactive N-acetylglucosaminyltransferase III
regulates CREB-mediated gene expression and Brain 143, 1114–1126 (2020). (GlcNAc-TIII) induces neurological and other traits
memory formation. Nat. Chem. Biol. 8, 253–261 156. Topaz, O. et al. Mutations in GALNT3, encoding a absent in mice that lack GlcNAc-TIII. J. Biol. Chem.
(2012). protein involved in O-linked glycosylation, cause 277, 26300–26309 (2002).
133. Tarbet, H. J. et al. Site-specific glycosylation regulates familial tumoral calcinosis. Nat. Genet. 36, 579–581 181. Matsumoto, K. et al. N-Glycan fucosylation of
the form and function of the intermediate filament (2004). epidermal growth factor receptor modulates receptor
cytoskeleton. eLife 7, e31807 (2018). 157. Tian, E. et al. Galnt11 regulates kidney function by activity and sensitivity to epidermal growth factor
134. Dennis, J. W. & Brewer, C. F. Density-dependent glycosylating the endocytosis receptor megalin to receptor tyrosine kinase inhibitor. Cancer Sci. 99,
lectin-glycan interactions as a paradigm for modulate ligand binding. Proc. Natl Acad. Sci. USA 1611–1617 (2008).
conditional regulation by posttranslational 116, 25196–25202 (2019). 182. Agrawal, P. et al. A systems biology approach
modifications. Mol. Cell. Proteom. 12, 913–20 158. Hansen, L. et al. A glycogene mutation map for identifies FUT8 as a driver of melanoma metastasis.
(2013). discovery of diseases of glycosylation. Glycobiology Cancer Cell 31, 804–819.e7 (2017).
135. Granovsky, M. et al. Suppression of tumor growth 25, 211–224 (2015). 183. Okada, M. et al. Blockage of core fucosylation
and metastasis in Mgat5-deficient mice. Nat. Med. 6, 159. Willer, C. J. et al. Newly identified loci that influence reduces cell-surface expression of PD-1 and promotes
306–312 (2000). lipid concentrations and risk of coronary artery anti-tumor immune responses of T cells. Cell Rep. 20,
136. Martínez Allo, V. C. et al. Suppression of disease. Nat. Genet. 40, 161–169 (2008). 1017–1028 (2017).
age-related salivary gland autoimmunity by 160. Khetarpal, S. A. et al. Loss of function of GALNT2 184. Marcos, N. T. et al. Role of the human ST6GalNAc-I
glycosylation-dependent galectin-1-driven immune lowers high-density lipoproteins in humans, nonhuman and ST6GalNAc-II in the synthesis of the cancer-
inhibitory circuits. Proc. Natl Acad. Sci. USA 117, primates, and rodents. Cell Metab. 24, 234–245 associated Sialyl-Tn antigen. Cancer Res. 64,
6630–6639 (2020). (2016). 7050–7057 (2004).
137. Demetriou, M., Nabi, I. R., Coppolino, M., Dedhar, S. This paper describes validation of the first 185. Sewell, R. et al. The ST6GalNAc-I sialyltransferase
& Dennis, J. W. Reduced contact-inhibition and glycosyltransferase gene implicated as a GWAS localizes throughout the Golgi and is responsible
substratum adhesion in epithelial cells expressing candidate. for the synthesis of the tumor-associated sialyl-Tn
GlcNAc-transferase V. J. Cell Biol. 130, 383–392 161. Roman, T. S. et al. Multiple hepatic regulatory variants O-glycan in human breast cancer. J. Biol. Chem. 281,
(1995). at the GALNT2 GWAS locus associated with high-density 3586–3594 (2006).
138. Nakano, M. et al. Bisecting GlcNAc is a general lipoprotein cholesterol. Am. J. Hum. Genet. 97, 186. Tarp, M. A. & Clausen, H. Mucin-type O-glycosylation
suppressor of terminal modification of N-glycan. 801–815 (2015). and its potential use in drug and vaccine development.
Mol. Cell. Proteom. 18, 2044–2057 (2019). 162. Cavalli, M., Pan, G., Nord, H. & Wadelius, C. Looking Biochim. Biophys. Acta 1780, 546–563 (2008).
139. Isaji, T. et al. Introduction of bisecting GlcNAc beyond GWAS: allele-specific transcription factor 187. Kudelka, M. R., Ju, T., Heimburg-Molinaro, J. &
into integrin α5β1 reduces ligand binding and binding drives the association of GALNT2 to HDL-C Cummings, R. D. Simple sugars to complex disease —
down-regulates cell adhesion and cell migration. plasma levels. Lipids Health Dis. 15, 18 (2016). mucin-type O-glycans in cancer. Adv. Cancer Res. 126,
J. Biol. Chem. 279, 19747–19754 (2004). 163. Duncan, E. L. et al. Genome-wide association study 53–135 (2015).
140. Wang, X. et al. Core fucosylation regulates epidermal using extreme truncate selection identifies novel 188. Sutherlin, M. E. et al. Expression of three UDP-
growth factor receptor-mediated intracellular genes affecting bone mineral density and fracture risk. N-acetyl-α-d-galactosamine:polypeptide GalNAc
signaling. J. Biol. Chem. 281, 2572–2577 (2006). PLoS Genet. 7, e1001372 (2011). N-acetylgalactosaminyltransferases in adenocarcinoma
141. Liang, W. et al. Core fucosylation of the T cell receptor 164. de Las Rivas, M. et al. Molecular basis for fibroblast cell lines. Cancer Res. 57, 4744–4748 (1997).
is required for T cell activation. Front. Immunol. 9, 78 growth factor 23 O-glycosylation by GalNAc-T3. 189. Freire, T. et al. UDP-N-acetyl-d-galactosamine:
(2018). Nat. Chem. Biol. 16, 351–360 (2020). polypeptide N-acetylgalactosaminyltransferase 6
142. Shields, R. L. et al. Lack of fucose on human IgG1 165. Taniguchi, N. & Kizuka, Y. Glycans and cancer: role (ppGalNAc-T6) mRNA as a potential new marker for
N-linked oligosaccharide improves binding to human of N-glycans in cancer biomarker, progression and detection of bone marrow-disseminated breast cancer
FcγRIII and antibody-dependent cellular toxicity. metastasis, and therapeutics. Adv. Cancer Res. 126, cells. Int. J. Cancer 119, 1383–1388 (2006).
J. Biol. Chem. 277, 26733–26740 (2002). 11–51 (2015). 190. Song, K.-H. et al. GALNT14 promotes lung-specific
This paper identifies core fucose on the N-glycans 166. Schultz, M. J., Swindall, A. F. & Bellis, S. L. Regulation breast cancer metastasis by modulating self-renewal
of IgG1 in regulation of antibody effector functions. of the metastatic cell phenotype by sialylated glycans. and interaction with the lung microenvironment.
143. Nguyen, J. T. et al. CD45 modulates galectin-1- Cancer Metastasis Rev. 31, 501–518 (2012). Nat. Commun. 7, 13796 (2016).
induced T cell death: regulation by expression of Core 2 167. Christiansen, M. N. et al. Cell surface protein 191. Kitada, S. et al. Polypeptide N-acetylgalactosaminyl
O-glycans. J. Immunol. 167, 5697–5707 (2001). glycosylation in cancer. Proteomics 14, 525–546 transferase 3 independently predicts high-grade
144. Lee, S. H. et al. Core2 O-glycan structure is essential (2014). tumours and poor prognosis in patients with renal
for the cell surface expression of sucrase isomaltase 168. Ju, T. et al. Human tumor antigens Tn and sialyl cell carcinomas. Br. J. Cancer 109, 472–481 (2013).
and dipeptidyl peptidase-IV during intestinal cell Tn arise from mutations in Cosmc. Cancer Res. 68, 192. Lavrsen, K. et al. De novo expression of human
differentiation. J. Biol. Chem. 285, 37683–37692 1636–46 (2008). polypeptide N-acetylgalactosaminyltransferase 6
(2010). 169. Sun, X., Ju, T. & Cummings, R. D. Differential (GalNAc-T6) in colon adenocarcinoma inhibits the
145. Halmo, S. M. et al. Protein O-linked mannose expression of Cosmc, T-synthase and mucins in differentiation of colonic epithelium. J. Biol. Chem.
β-1,4-N-acetylglucosaminyl-transferase 2 (POMGNT2) Tn-positive colorectal cancers. BMC Cancer 18, 827 293, 1298–1314 (2018).
is a gatekeeper enzyme for functional glycosylation (2018). 193. Dalziel, M. et al. The relative activities of the C2GnT1
of α-dystroglycan. J. Biol. Chem. 292, 2101–2109 170. Radhakrishnan, P. et al. Immature truncated and ST3Gal-I glycosyltransferases determine O-glycan
(2017). O-glycophenotype of cancer directly induces oncogenic structure and expression of a tumor-associated
This paper describes the peptide substrate features. Proc. Natl Acad. Sci. USA 111, E4066–E4075 epitope on MUC1. J. Biol. Chem. 276, 11007–11015
selectivity of the ER-located POMGNT2 that (2014). (2001).
determines the O-Man glycosites that proceed 171. Fernandez, A. J. et al. The structure of the colorectal 194. Swindall, A. F. & Bellis, S. L. Sialylation of the Fas
towards biosynthesis of the elaborated matriglycan. cancer-associated enzyme GalNAc-T12 reveals how death receptor by ST6Gal-I provides protection
146. Varki, A. Glycan-based interactions involving nonconserved residues dictate its function. Proc. Natl against Fas-mediated apoptosis in colon carcinoma
vertebrate sialic-acid-recognizing proteins. Acad. Sci. USA 116, 20404–20410 (2019). cells. J. Biol. Chem. 286, 22982–22990 (2011).
Nature 446, 1023–1029 (2007). 172. Büll, C., Stoel, M. A., den Brok, M. H. & Adema, G. J. 195. Holdbrooks, A. T., Britain, C. M. & Bellis, S. L.
147. Cohen, M. & Varki, A. Modulation of glycan Sialic acids sweeten a tumor’s life. Cancer Res. 74, ST6Gal-I sialyltransferase promotes tumor necrosis
recognition by clustered saccharide patches. 3199–3204 (2014). factor (TNF)-mediated cancer cell survival via
Int. Rev. Cell Mol. Biol. 308, 75–125 (2014). 173. Dall’Olio, F. & Trinchera, M. Epigenetic bases of sialylation of the TNF receptor 1 (TNFR1) death
148. Giannini, S. et al. β4GALT1 controls β1 integrin aberrant glycosylation in cancer. Int. J. Mol. Sci. 18, receptor. J. Biol. Chem. 293, 1610–1622 (2018).
function to govern thrombopoiesis and hematopoietic 998 (2017). 196. Barthel, S. R. et al. α1,3 Fucosyltransferases are
stem cell homeostasis. Nat. Commun. 11, 356 (2020). 174. Ashkani, J. & Naidoo, K. J. Glycosyltransferase gene master regulators of prostate cancer cell trafficking.
149. Hennet, T., Chui, D., Paulson, J. C. & Marth, J. D. expression profiles classify cancer types and propose Proc. Natl Acad. Sci. USA 106, 19491–19496
Immune regulation by the ST6Gal sialyltransferase. prognostic subtypes. Sci. Rep. 6, 26451 (2016). (2009).
Proc. Natl Acad. Sci. USA 95, 4504–4509 (1998). 175. Dusoswa, S. A. et al. Glioblastomas exploit truncated 197. Esposito, M. et al. Bone vascular niche E-selectin
150. Comelli, E. M. et al. Activation of murine CD4+ and O-linked glycans for local and distant immune induces mesenchymal-epithelial transition and Wnt
CD8+ T lymphocytes leads to dramatic remodeling modulation via the macrophage galactose-type activation in cancer cells to promote bone metastasis.
of N-linked glycans. J. Immunol. 177, 2431–2440 lectin. Proc. Natl Acad. Sci. USA 117, 3693–3703 Nat. Cell Biol. 21, 627–639 (2019).
(2006). (2020). 198. Engle, D. D. et al. The glycan CA19-9 promotes
151. Priatel, J. J. et al. The ST3Gal-I sialyltransferase 176. ICGC/TCGA Pan-Cancer Analysis of Whole Genomes pancreatitis and pancreatic cancer in mice. Science
controls CD8+ T lymphocyte homeostasis by Consortium. Pan-cancer analysis of whole genomes. 364, 1156–1162 (2019).
modulating O-glycan biosynthesis. Immunity 12, Nature 578, 82–93 (2020). 199. Guo, H. et al. O-Linked N-acetylglucosamine
273–283 (2000). 177. Zeng, J. et al. Promoters of human Cosmc and (O-GlcNAc) expression levels epigenetically regulate
152. Ohtsubo, K. & Marth, J. D. Glycosylation in T-synthase genes are similar in structure, yet colon cancer tumorigenesis by affecting the cancer
cellular mechanisms of health and disease. Cell 126, different in epigenetic regulation. J. Biol. Chem. 290, stem cell compartment via modulating expression
855–867 (2006). 19018–19033 (2015). of transcriptional factor MYBL1. J. Biol. Chem. 292,
153. Mereiter, S., Balmaña, M., Campos, D., Gomes, J. & 178. Agrawal, P. et al. Mapping posttranscriptional 4123–4137 (2017).
Reis, C. A. Glycosylation in the era of cancer-targeted regulation of the human glycome uncovers microRNA 200. Hanover, J. A., Chen, W. & Bond, M. R. O-GlcNAc
therapy: where are we heading? Cancer Cell 36, 6–16 defining the glycocode. Proc. Natl Acad. Sci. USA 111, in cancer: an oncometabolism-fueled vicious cycle.
(2019). 4338–4343 (2014). J. Bioenerg. Biomembr. 50, 155–173 (2018).

www.nature.com/nrm
Reviews

201. Theocharis, A. D. & Karamanos, N. K. Proteoglycans engineered human organotypic skin model. Dev. Cell 250. Mahdavi, J. Helicobacter pylori SabA adhesin in
remodeling in cancer: underlying molecular 54, 669–684 (2020). persistent infection and chronic inflammation. Science
mechanisms. Matrix Biol. 75–76, 220–259 226. Büll, C., Joshi, H. J., Clausen, H. & Narimatsu, Y. 297, 573–578 (2002).
(2019). Cell-based glycan arrays — a practical guide to 251. Rossez, Y. et al. The LacdiNAc-specific adhesin LabA
202. Salanti, A. et al. Targeting human cancer by a dissect the human glycome. STAR Protoc. 1, 100017 mediates adhesion of Helicobacter pylori to human
glycosaminoglycan binding malaria protein. (2020). gastric mucosa. J. Infect. Dis. 210, 1286–1295
Cancer Cell 28, 500–514 (2015). 227. Naegeli, A. & Aebi, M. Current approaches to (2014).
This paper identifies a broadly expressed engineering N-linked protein glycosylation in bacteria. 252. Nagae, M. et al. Structure and mechanism of
cancer-associated 4-O-sulfated chondroitin sulfate Methods Mol. Biol. 1321, 3–16 (2015). cancer-associated N-acetylglucosaminyltransferase-V.
epitope using the malarial receptor VAR2CSA. 228. Hudak, J. E. & Bertozzi, C. R. Glycotherapy: Nat. Commun. 9, 3380 (2018).
203. Chen, Y.-H. et al. The GAGOme: a cell-based library new advances inspire a reemergence of glycans 253. Pinho, S. S. & Reis, C. A. Glycosylation in cancer:
of displayed glycosaminoglycans. Nat. Methods 15, in medicine. Chem. Biol. 21, 16–37 (2014). mechanisms and clinical implications. Nat. Rev. Cancer
881–888 (2018). 229. Van Landuyt, L., Lonigro, C., Meuris, L. & 15, 540–555 (2015).
This paper describes the development of a Callewaert, N. Customized protein glycosylation to 254. Läubli, H. & Varki, A. Sialic acid-binding
cell-based display of glycosaminoglycans. improve biopharmaceutical function and targeting. immunoglobulin-like lectins (Siglecs) detect
204. Petrosyan, A. Unlocking Golgi: why does morphology Curr. Opin. Biotechnol. 60, 17–28 (2019). self-associated molecular patterns to regulate
matter? Biochemistry 84, 1490–1501 (2019). 230. Montero-Morales, L. & Steinkellner, H. Advanced immune responses. Cell. Mol. Life Sci. 77, 593–605
205. Kulkarni-Gosavi, P., Makhoul, C. & Gleeson, P. A. plant-based glycan engineering. Front. Bioeng. (2020).
Form and function of the Golgi apparatus: scaffolds, Biotechnol. 6, 81 (2018). 255. Maxson, J. E. et al. Ligand-independence of the
cytoskeleton and signalling. FEBS Lett. 593, 231. Walsh, G. Post-translational modifications of colony stimulating factor 3 receptor (CSF3R) T618I
2289–2305 (2019). protein biopharmaceuticals. Drug Discov. Today 15, mutation results from loss of O-linked glycosylation
206. Petrosyan, A. Onco-Golgi: is fragmentation a gate 773–780 (2010). and increased receptor dimerization. J. Biol. Chem.
to cancer progression? Biochem. Mol. Biol. J. 232. Čaval, T., Tian, W., Yang, Z., Clausen, H. & 289, 5820–5827.(2014).
https://doi.org/10.21767/2471-8084.100006 Heck, A. J. R. Direct quality control of glycoengineered 256. Jiang, Y. et al. O-glycans on death receptors in
(2015). erythropoietin variants. Nat. Commun. 9, 3342 cells modulate their sensitivity to TRAIL-induced
207. Chia, J., Goh, G. & Bard, F. Short O-GalNAc glycans: (2018). apoptosis through affecting on their stability and
regulation and role in tumor development and 233. Grabowski, G. A. et al. Enzyme therapy in type 1 oligomerization. FASEB J. 34, 11786–11801
clinical perspectives. Biochim. Biophys. Acta 1860, Gaucher disease: comparative efficacy of mannose- (2020).
1623–1639 (2016). terminated glucocerebrosidase from natural and 257. Rillahan, C. D. & Paulson, J. C. Glycan microarrays
208. Gill, D. J., Clausen, H. & Bard, F. Location, location, recombinant sources. Ann. Intern. Med. 122, 33–39 for decoding the glycome. Annu. Rev. Biochem. 80,
location: new insights into O-GalNAc protein (1995). 797–823 (2011).
glycosylation. Trends Cell Biol. 21, 149–158 234. Umaña, P., Jean-Mairet, J., Moudry, R., Amstutz, H. 258. Blixt, O. et al. Printed covalent glycan array for
(2011). & Bailey, J. E. Engineered glycoforms of an ligand profiling of diverse glycan binding proteins.
209. Axelsson, M. A. B. et al. Neutralization of antineuroblastoma IgG1 with optimized Proc. Natl Acad. Sci. USA 101, 17033–17038
pH in the Golgi apparatus causes redistribution antibody-dependent cellular cytotoxic activity. (2004).
of glycosyltransferases and changes in the Nat. Biotechnol. 17, 176–180 (1999).
O-glycosylation of mucins. Glycobiology 11, 633–644 235. Yamane-Ohnuki, N. et al. Establishment of FUT8 Acknowledgements
(2001). knockout Chinese hamster ovary cells: an ideal host The authors are grateful to R. Schnaar and B. Henrissat for
210. Chatterjee, S. et al. Protein paucimannosylation is an cell line for producing completely defucosylated discussions and critical comments on the manuscript. They
enriched N-glycosylation signature of human cancers. antibodies with enhanced antibody-dependent thank H. Wandall, A. Halim, C. Büll, Y. Zhang and L. Hansen
Proteomics 19, e1900010 (2019). cellular cytotoxicity. Biotechnol. Bioeng. 87, 614–622 for help with the manuscript, and all members of the
211. Chia, J., Tay, F. & Bard, F. The GalNAc-T Activation (2004). Copenhagen Center for Glycomics for discussions. Supported
(GALA) pathway: drivers and markers. PLoS ONE 14, 236. Meuris, L. et al. GlycoDelete engineering of mammalian by the Lundbeck Foundation, the Novo Nordisk Foundation
e0214118 (2019). cells simplifies N-glycosylation of recombinant proteins. and the Danish National Research Foundation (DNRF107).
212. Nguyen, A. T. et al. Organelle specific O-glycosylation Nat. Biotechnol. 32, 485–489 (2014).
drives MMP14 activation, tumor growth, and 237. Chang, M. M. et al. Small-molecule control of
Author contributions
metastasis. Cancer Cell 32, 639–653.e6 (2017). antibody N-glycosylation in engineered mammalian
The authors contributed equally to all aspects of the article.
213. Yamaji, T. et al. A CRISPR screen using subtilase cells. Nat. Chem. Biol. 15, 730–736 (2019).
cytotoxin identifies SLC39A9 as a glycan-regulating 238. Schulz, M. A. et al. Glycoengineering design options
factor. iScience 15, 407–420 (2019). for IgG1 in CHO cells using precise gene editing. Competing interests
214. Narimatsu, Y. et al. A validated gRNA library for Glycobiology 28, 542–549 (2018). The University of Copenhagen has filed patent applications
CRISPR/Cas9 targeting of the human glycosyltransferase 239. Tian, W. et al. The glycosylation design space for related to engineering cellular glycosylation capacities and
genome. Glycobiology 28, 295–305 (2018). recombinant lysosomal replacement enzymes cell-based glycan arrays. H.C. and Y.N. are named inventors
215. Narimatsu, Y. et al. An atlas of human glycosylation produced in CHO cells. Nat. Commun. 10, 1785 on these applications. GlycoDisplay Aps has license rights to
pathways enables display of the human glycome by (2019). these patent applications, and H.C. and Y.N. hold shares in
gene engineered cells. Mol. Cell 75, 394–407.e5 240. RodrÍguez, E., Schetters, S. T. T. & van Kooyk, Y. The GlycoDisplay Aps. The remaining authors declare no competing
(2019). tumour glyco-code as a novel immune checkpoint for interests.
This paper describes the development and utility immunotherapy. Nat. Rev. Immunol. 18, 204–211
of a cell-based display of the human glycome. (2018). Peer review information
216. Yang, Z. et al. Engineered CHO cells for production of 241. Mathiesen, C. B. K. et al. Genetically engineered cell Nature Reviews Molecular Cell Biology thanks S. Flitsch, C. Reis
diverse, homogeneous glycoproteins. Nat. Biotechnol. factories produce glycoengineered vaccines that target and the other, anonymous, reviewer(s) for their contribution to
33, 842–844 (2015). antigen-presenting cells and reduce antigen-specific the peer review of this work.
217. Patnaik, S. K. & Stanley, P. Lectin-resistant CHO T-cell reactivity. J. Allergy Clin. Immunol. 142,
glycosylation mutants. Methods Enzymol. 416, 1983–1987 (2018). Publisher’s note
159–182 (2006). 242. Byrne, G. et al. CRISPR/Cas9 gene editing for the Springer Nature remains neutral with regard to jurisdictional
218. Steentoft, C. et al. Precision genome editing: creation of an MGAT1-deficient CHO cell line to claims in published maps and institutional affiliations.
a small revolution for glycobiology. Glycobiology 24, control HIV-1 vaccine glycosylation. PLoS Biol. 16,
663–680 (2014). e2005817 (2018).
219. Jae, L. T. et al. Deciphering the glycosylome of 243. Zhang, R. et al. Reducing immunoreactivity of porcine Supplementary information
dystroglycanopathies using haploid screens for bioprosthetic heart valves by genetically-deleting Supplementary information is available for this paper at
lassa virus entry. Science 340, 479–483 (2013). three major glycan antigens, GGTA1/β4GalNT2/ https://doi.org/10.1038/s41580-020-00294-x.
220. Han, J. et al. Genome-wide CRISPR/Cas9 screen CMAH. Acta Biomater. 72, 196–205 (2018).
identifies host factors essential for influenza virus 244. Liu, Q. P. et al. Identification of a GH110 subfamily
replication. Cell Rep. 23, 596–607 (2018). of α1,3-galactosidases: novel enzymes for removal of
Related links
Carbohydrate-Active enZYmes database: http://www.cazy.org
221. Ye, L. et al. In vivo CRISPR screening in CD8 T cells the α3Gal xenotransplantation antigen. J. Biol. Chem.
Copenhagen Center for Glycomics, University of
with AAV–Sleeping Beauty hybrid vectors identifies 283, 8545–8554 (2008).
Copenhagen: https://glycomics.ku.dk
membrane targets for improving immunotherapy 245. Ibrahim, A. M. S. et al. Acellular dermal matrices in
essentials of Glycobiology, 3rd edition: https://www.ncbi.
for glioblastoma. Nat. Biotechnol. 37, 1302–1313 breast surgery: a comprehensive review. Ann. Plast.
nlm.nih.gov/books/NBK310274/
(2019). Surg. 70, 732–738 (2013).
exPADsy glycomics: https://www.expasy.org/glycomics
222. Miller, R. L. et al. Shotgun ion mobility mass 246. Neelamegham, S. et al. Updates to the Symbol
GlycoDomainviewer: https://glycodomain.glycomics.ku.dk
spectrometry sequencing of heparan sulfate Nomenclature for Glycans guidelines. Glycobiology
GlyGen: https://www.glygen.org
saccharides. Nat. Commun. 11, 1481 (2020). 29, 620–624 (2019).
GlyTouCan: https://glytoucan.org
223. Stopschinski, B. E. et al. Specific glycosaminoglycan 247. Parker, J. L. & Newstead, S. Gateway to the
Handbook of Glycosyltransferases and Related Genes.
chain length and sulfation patterns are required for Golgi: molecular mechanisms of nucleotide sugar
editors: Taniguchi, N., Honke, K., Fukuda, M., Narimatsu, H.,
cell uptake of tau versus α-synuclein and β-amyloid transporters. Curr. Opin. Struct. Biol. 57, 127–134
Yamaguchi, Y., Angata, T. (eds.) springer Nature, 2014.
aggregates. J. Biol. Chem. 293, 10826–10840 (2019).
society for Glycobiology: http://glycobiology.org
(2018). 248. Nabi, I. R., Shankar, J. & Dennis, J. W. The galectin
international Glycoconjugate Organization: https://
224. Tian, S. et al. Genome-wide CRISPR screens for lattice at a glance. J. Cell Sci. 128, 2213–2219
intl-glyco.org
Shiga toxins and ricin reveal Golgi proteins critical (2015).
NetOGlyc 4.0: http://www.cbs.dtu.dk/services/NetOGlyc/
for glycosylation. PLoS Biol. 16, e2006951 (2018). 249. Ilver, D. Helicobacter pylori adhesin binding
225. Dabelsteen, S. et al. Essential functions of glycans fucosylated histo-blood group antigens revealed
in human epithelia dissected by a CRISPR–Cas9- by retagging. Science 279, 373–377 (1998). © Springer Nature Limited 2020

Nature Reviews | Molecular Cell Biology

You might also like