You are on page 1of 9

Carbohydrate Research 403 (2015) 60–68

Contents lists available at ScienceDirect

Carbohydrate Research
journal homepage: www.elsevier.com/locate/carres

Minireview

Re-visiting the structure of heparin


Benito Casu ⇑, Annamaria Naggi, Giangiacomo Torri
G. Ronzoni Institute for Chemical and Biochemical Research, via G. Colombo, 81 20133 Milan, Italy

a r t i c l e i n f o a b s t r a c t

Article history: The sulfated polysaccharide heparin has been used as a life-saving anticoagulant in clinics well before its
Received 13 June 2014 detailed structure was known. This mini-review is a survey of the evolution in the discovery of the pri-
Accepted 22 June 2014 mary and secondary structure of heparin. Highlights in this history include elucidation and synthesis of
Available online 3 July 2014
the specific sequence that binds to antithrombin, the development of low-molecular-weight heparins
currently used as antithrombotic drugs, and the most promising start of chemo-enzymatic synthesis.
Keywords: Special emphasis is given to peculiar conformational properties contributing to interaction with proteins
Heparin structure
that modulate different biological properties.
Active site for antithrombin
Low-molecular-weight heparins
Ó 2014 Elsevier Ltd. All rights reserved.
Heparin oligosaccharides
Protein binding
Conformations

1. Introduction 2. From ‘mysterious heparin’ to Wolfrom’s structure

Heparin is a well-known sulfated polysaccharide widely used as Heparin and its anticoagulant activity were ‘discovered’ in Tor-
an anticoagulant and antithrombotic drug and with perspective onto about 100 years ago by McLean and Howell, who were actu-
uses in other therapeutic fields. This paper is an overview of ally looking for a pro-coagulant substance extracted from tissues
advances in the knowledge of structure and structure-activity rela- and thought to be a phospholipid. It had taken some time to the
tionships of heparin as evolved over the past few decades, with scientific community to realize that heparin is in fact a sulfated
glimpses on its early history. To meet with the space constraint polysaccharide belonging to the class of glycosaminoglycans
of mini-reviews, the authors referred to previous extended reviews (GAG), constituted by alternating disaccharide sequences of an
and books,1–7 to some landmark papers and to most recent articles uronic acid and a hexosamine. It is noteworthy that, while still
of groups active in the field for recognizing contributions that being a ‘mysterious’ entity, that is, before the exact nature of the
could not be specifically mentioned in the present context. Such component carbohydrate residues and location of sulfate groups
an exercise implied some arbitrary choices: this overview follows along the polysaccharide chains were established, heparin started
the strategy of reporting ‘case studies’ involving the research to be successfully used as an anticoagulant in clinics, arising enthu-
groups of the authors. It was also meant as a tribute to Carbohy- siasm especially as a life-saving drug that permitted open-heart
drate Research for its steadily contributing, since its early issues, surgery.8
to the progresses in heparin research. Reviews on the history of heparin have been published by Rodé-
n9 and Barrowcliffe.10 Hereinafter some milestones of the early his-
Abbreviations: GAG, glycosaminoglycan; GlcA, D-glucuronic acid; GlcA2SO3, tory are mentioned. In 1935, Jorpes established that the uronic acid
D-glucuronic acid 2-sulfate; GlcN, D-glucosamine; GlcNSO36SO3, D-glucosamine and aminosugar ratio of heparin was 1:1 as in chondroitin sulfates.
N,6-sulfated; IdoA, L-iduronic acid; IdoA2SO3, L-iduronic acid 2-sulfate; TSD, Since the sulfur content of heparin corresponded to about 2.5 sul-
trisulfated disaccharide; AT, antithrombin; HA, high affinity for AT; LA, low-affinity fate groups per disaccharide unit and on the assumption of a reg-
for AT; ATBR, antithrombin-binding region; LMWH, low-molecular-weight heparin;
ular structure of its polysaccharide backbone, heparin was
GlcNSO33,6SO3, 3,6-O-sulfated D-glucosamine; ManNSO36SO3, D-mannosamine
6-sulfate; 1,6-anhydro-GlcNSO3, 1,6-anhydro-D-glucosamine N-sulfate; 1,6-anhydro- erroneously thought to be an oversulfated chondroitin sulfate. 
ManNSO3, 1,6-anhydro-D-mannosamine N-sulfate; ULMWH, ultra-low-molecular- Some years later Wolfrom, who thought that D-glucuronic acid
weight heparin; FGFs, fibroblast growth factors; FGF1, fibroblast growth factor-1; (GlcA) was the only uronic acid component of heparin, proposed
FGF2, fibroblast growth factor-2; FGFR, fibroblast growth factor receptor; 3OST-1 and
3OST-3, 3-O-sulfotranferases 1 and 3; K5, E. coli K5 polysaccharide; HQSCcos,
statistical correlation two-dimensional NMR spectroscopy.  
Curiously, a major crisis in the use of heparin in clinics was caused in the late
⇑ Corresponding author. 2007 and early 2008 by contamination with an unnatural oversulfated chondroitin
E-mail address: casu@ronzoni.it (B. Casu). sulfate (see Section 11).

http://dx.doi.org/10.1016/j.carres.2014.06.023
0008-6215/Ó 2014 Elsevier Ltd. All rights reserved.
B. Casu et al. / Carbohydrate Research 403 (2015) 60–68 61

the heparin structure to be represented by repeating units of alter- 4. Discovery and synthesis of the binding site for antithrombin
nating 1,4-linked GlcA2SO3 and GlcNSO36SO3 residues, where the Mechanism of anticoagulation
configuration of glycosidic bonds was proposed to be a for both GlcA
and GlcN on the basis of optical rotation data.11 The configuration of Soon after Rosenberg’s milestone finding that the anticoagulant
GlcA in heparin was subsequently demonstrated to be b.12 activity of heparin was mainly based on its ability to bind anti-
thrombin (AT), thus accelerating by several orders of magnitude
3. First re-visitation: L-iduronic acid enters into the picture the AT-mediated inhibition of coagulation factors, in 1976 Rosen-
berg’s, Lindahl’s and Andersson’s groups independently discovered
Wolfrom’s structure for heparin remained undisputed for sev- that heparin was heterogeneous in terms of its interaction with AT.
eral years. Although in the meantime L-iduronic acid (IdoA) had In fact, they separated chains with high affinity (HA) from those
been detected in some heparins, it was generally thought that with low affinity (LA) for AT, and found that the anticoagulant
these findings were reflecting residual contents of IdoA-containing activity was largely associated with the HA chains, these latter con-
GAGs such as dermatan sulfate, which is difficult to totally remove stituting only about one third of those of the unfractionated poly-
from heparin preparations. Undoubtedly, acceptance of Wolfrom’s saccharide (UFH). That also was a landmark finding and a
structure has been also a tribute to his outstanding reputation as a surprising one, since the compositional differences between HA
carbohydrate chemist. However, such a consent was mainly due to and LA chains were subtle and not easily detectable. Rosenberg
serious problems facing everybody who attempted to tackle the observed that nitrous acid-generated fragments obtained from
heparin structure using methods available at the time for obtaining HA heparin contained significantly more unsulfated IdoA than
di- and oligosaccharide fragments to reconstruct the structure of the LA species. Similarly the Ronzoni and Choay groups noticed
the polysaccharide. In the late 1960s and early 1970s the applica- marked differences in the NMR spectra of HA and LA oligosaccha-
tion of milder methods of acid hydrolysis and the advent of non- rides. However, the structure of the antithrombin-binding region
destructive analytical techniques such as NMR spectroscopy rap- (ATBR) remained elusive for some more time, until Lindahl’s group
idly changed the picture. In 1962, Cifonelli and Dorfman found established that the ATBR was a pentasaccharide (structure shown
IdoA to be a major uronic acid component of heparin,13 and in in Fig. 1B). This pentasaccharide contains the typical 3-O-sulfated
1968 preliminary NMR studies by Perlin indicated that the struc- GlcN, optionally also 6-O-sulfated, GlcNSO33,6SO3 residue.25 Note-
ture of heparin was not as simple as depicted by Wolfrom’s for- worthy, the unsulfated IdoA residue preceding this pentasaccha-
mula, and that IdoA residues substantially contributed to the ride sequence, systematically found in HA species from porcine
structure of the polysaccharide.14 Analysis of higher resolution mucosal heparin, did not significantly contribute to the affinity
(220 MHz) spectra15 and chemical studies16 together with inde- for AT. Identification of sulfate groups essential for high-affinity
pendent structural studies by Lindahl on nitrous-acid generated binding to AT (as depicted in Fig. 1B) was completed by the com-
fragments17 finally provided clear evidence that IdoA (mostly as bined application of classical methods for sulfated oligosaccharide
IdoA2SO3) was the prominent uronic acid of heparin. In the mean- analysis; this latter work, which appeared in n. 100 of Carbohydrate
time, Wolfrom had admitted that his earlier results had led to an Research,26 remains an important reference in the field. All sulfate
incorrect conclusion.18 As reviewed in Refs. 1,10, the hydrolytic pro- groups essential and/or important for high-affinity binding to AT
cedures previously applied were leading to the loss of IdoA-contain- (as depicted in Fig. 1B) were identified.27 The 3-O-sulfate group
ing fragments; as a consequence, Wolfrom’s analysis was based on taken as a marker of the ATBR was also characterized by a specific
the minor GlcA-containing ones. In a study exploiting for the first signal in the 13C NMR spectra of HA heparin oligosaccharides
time NMR spectroscopic analysis of enzymatic digests (provided by obtained with different methods.28 In variants of ATBR found in
Dietrich), Perlin also established the a-L-configuration of the other heparin sources, especially from bovine lung, the 3-O-sul-
IdoA2SO3 residues of heparin.19 The optical rotation of heparin was fated GlcN residue is prevalently N-sulfated and the IdoA residue
rationalized by studies with model synthetic iduronates.20 A com- preceding the active pentasaccharide is IdoA2SO3.29
plete analysis of 1H and 13C NMR spectra of heparin, highlighting Even before the structure of the ATBR was fully elucidated, the
its major components, was published in 1979.21 These findings led Choay group started an ambitious program of chemical synthesis
to establish the structure of the major disaccharide repeating units of the active pentasaccharide, reaching its goal in 1983.30 (For
of heparin as shown in (Fig. 1A). The trisulfated disaccharide (TSD) detailed coverage of discovery and synthesis of ATBR see
repeating sequences –IdoA2SO3–GlcNSO3,6SO3—were found to be Refs. 31–33) In a collaboration between Sanofi and Organon, the
especially prominent (up to 90%) in preparations from beef lung, pentasaccharide was later developed as an antithrombotic drug
an organ that for some time was the principal source of heparin, (fondaparinux, ArixtraÒ).33
and somewhat less represented (about 75%) in heparins from porcine Regarding the structure of heparin with respect to thrombin
mucosa, which largely replaced beef lung as an animal tissue for inhibition it was shown, using chemical synthesis, that a hexadeca-
preparation of clinical heparins. The complement to 100% of uronic saccharide displaying an ATBR at the reducing end is required.33
acids was GlcA, usually associated also with N-acetylated (instead The basis for the specificity of AT-mediated inhibition of thrombin
of N-sulfated) glucosamine residues. Biosynthetic studies, especially was later established by crystallographic studies using a synthetic
by the Uppsala group (reviewed in Ref. 22) definitively proved that hexadecasaccharide.34
GlcA- and GlcNAc-containing sequences were actual constituents of
the heparin chains, their presence being the result of incomplete 5. Minor sequences. Updated view of the heparin structure
modification of the biosynthetic precursor chains constituted by
repeating –GlcA–GlcNAc–sequences. These studies, together with Minor sequences, contributing to the structural microhetero-
structural analysis of extensively purified heparins (see Section 11) geneity of heparin, include internal GlcA2SO3 residues, first discov-
definitively established the concept that heparin was structurally ered by Conrad in liver HS (see Ref. 3) and identified in heparin
microheterogeneous. Heparins from different tissues and animal spe- lyase I-generated fragments of porcine mucosal heparin.35,36 N-
cies were found to have different contents of IdoA2SO3 and IdoA, and unsubstituted GlcN residues are among the very minor sequences
to be heterogeneous also in terms of size, being composed of chains of heparin (see Section 11). Some of the heparin chains terminate,
of different length, their mean MW ranging from 10 to 20 KDa at their reducing end, with a ‘linkage region’ (LR) reminiscent of
depending also on the method of preparation.23,24 the sequence –GlcA–Gal–Gal–Xyl–Ser linking the carbohydrate
62 B. Casu et al. / Carbohydrate Research 403 (2015) 60–68

(A)

(B)

Figure 1. (A) Major, trisulfated disaccharide (TSD) repeating units of heparin. (B) Hexasaccharidic heparin sequence containing the pentasaccharidic antithrombin-binding
region (ATBR). Typical residues of the ATBR are the 3-O-sulfated glucosamine GlcNSO33,6SO3 preceded by a nonsulfated GlcA. Sulfate groups essential for high-affinity
binding to AT (Refs. 27,32) are highlighted with ovals. In the highly sulfated heparins, such as bovine lung heparin, the 3-O-sulfated GlcN residue is prevalently N-sulfated,
and the IdoA residue preceding the active pentasaccharide is IdoA2SO3.29 In the synthetic pentasaccharide fondaparinux the 6-O-sulfated glucosamino residue is N-sulfated.33

chains to the original GAG proteoglycan; most often, the LR is par- ment of low-molecular-weight heparins (LMWHs) as antithrom-
tially eroded during purification processes of heparin (Refs. 37, and botic agents with a lower anticoagulant activity. Although the
Refs. therein). original expectation that LMWHs would involve lower risks of
The established heterogeneity of heparin chains in terms of size, bleeding than UFH turned out to be incorrect, these mini-heparins
sequence of the backbone residues and sulfation patterns, also have met with increasing success, mainly because of their better
depending on animal source and ways of purification (Ref. 38) pharmacokinetic properties and easy control of therapeutic doses.
makes it difficult to depict the heparin structure even in statistical Originally obtained by size-fractionation of heparin, the most com-
terms. A complete picture of heparin should consist of a list of its mon LMWHs were obtained by partial depolymerization of UFH.
numerous component chains, with indication of their relative con- For a recent review, see Ref. 46. For discussions on relationship
tents. Methods have been developed for sequencing heparin and between anticoagulant and antithrombotic activity of heparin see
heparan sulfate oligosaccharides39,40 and for mapping heparin Ref. 44; for development of LMWHs and their clinical use, see
chains by superimposing the structures of fragments obtained by Ref. 10.
graded enzymatic cleavage41 and great progresses have been made LMWHs obtained with different depolymerization procedures,
toward profiling all component species (for a review, see Ref. 42). and also those obtained with the same procedures but under
However, the isolation, sequencing and quantification of all the different reaction or purification conditions, have different compo-
heparin chains remain a formidable task. A way of depicting an sitional characteristics, as clearly shown, that is, by their oligosac-
average heparin chain (used by Linhardt’s group)43 is adopted here charide mapping47 and NMR spectra48,49 and are considered
(Fig. 2A) for representing an updated picture of an ATBR-containing different therapeutic entities.50 In fact, different LMWHs not only
heparin chain. The structure of the undegraded LR is reported in may have different end groups generated by the specific depoly-
Figure 2B. merization procedures, but may differ from each other also in com-
positional terms, including their content of intact ATBR sequences.
6. Low-molecular-weight-heparins As a typical example illustrated in Fig. 3, chains of LMWHs pre-
pared by cleavage of heparin with heparin lyase I terminate at their
The biological action of the active pentasaccharide stems on the nonreducing end with unsaturated uronic acid residues as in the
concept that it represents the essential heparin sequence that LMWHs prepared by chemical b-elimination. On the other hand,
strongly binds to AT and dramatically increases the rate of AT- the two LMWHs greatly differ at their ‘reducing’ terminals, where
mediated inhibition of factor Xa while only marginally inhibiting the enzymatically-generated chains prevalently terminate with a
thrombin. In fact, besides containing the sequence of the ATBR, GlcNSO36SO3 residue, while a substantial proportion of the end
in order to inhibit also thrombin (factor IIa) heparin chains should residues of the chemically-obtained ones are either ManNSO36SO3,
be at least 18 residues long (for comprehensive reviews, see Refs. 1,6-anhydro-GlcNSO3 or 1,6-anhydro-ManNSO3 (reviewed in Ref.
32,33,44). The molecular basis for the interactions relevant to the 46). As an additional example of structural modification induced
anticoagulant and antithrombotic activity of heparin species is in LMWHs by the specific method used for their preparation, enox-
illustrated by the 3D structures (compared in Ref. 44) of AT, hepa- aparin contains significantly more GlcA2SO3 residues than heparin
rin-activated AT and complexes of AT with thrombin and other and the other LMWHs.49,51
coagulation factors as determined by X-ray crystallography A number of reducing end residues of the enzymatically-
(mainly by Huntington’s group) or by NMR methods (by Mulloy’s derived LMWHs are constituted by the 3-O-sulfated residues
group). More detailed information on the mechanism of activation GlcNSO33,6SO3 typical of the ATBR. In fact, heparinase 1 cleaves
of AT is given in Section 9.2. the ATBR sequence,52,53 resulting in a lower anti-Xa activity with
The first report on the size-dependence of the anti-thrombin respect to chains of similar size containing intact ATBRs, such as
and anti-factor Xa activities of heparin45 triggered the develop- those obtainable under controlled chemical b-elimination.51 The
B. Casu et al. / Carbohydrate Research 403 (2015) 60–68 63

(A)

(B)

Figure 2. (A) Simplified formula of a chain of porcine intestinal mucosal heparin containing an ATBR sequence, thus with high affinity for antithrombin. I = IdoA; G = GlcA in
the ATBR; G0 = GlcA toward the reducing end (RE) of the chain. The structure is from Ref. 43. (B) Intact linkage region (LR); see Ref. 38 and Refs. therein. GLR = GlcA in the
linkage region, Gal1 and Gal 2 are two D-galactose residues.

(A)

(B)

(C)

Figure 3. Simplified structures of the LMWHs tinzaparin (A), enoxaparin (B) and dalteparin (C), showing prevalent terminal residues at the reducing and reducing ends.
Internal sequences are indicated as in non-depolymerized heparin (From Ref. 91).

compositional and structural characteristics of the recently intro- cal exploitation of the lower-MW fractions of LMWH. Studies
duced ‘generic’ (bioequivalent) LMWHs are required to closely have been made, mainly using controlled depolymerization of hep-
match those of the originators.54 arin with recombinant heparin-lyases, to obtain LMW and ultra-
low-molecular-weight heparins (ULMWHs) richer in ATBR than
7. Ultra-low-molecular-weight heparins current LMWHs.55 ULMWHs essentially consisting of the pentasac-
charidic ATBR were successfully obtained by a chemo-enzymatic
The success of the first ‘active’ synthetic pentasaccharide fonda- approach.56 Also successful was the attempt to prepare ULMWHs
parinux33 aroused interest in exploring the possible pharmaceuti- by adjusting the reaction conditions for the b-elimination reaction
64 B. Casu et al. / Carbohydrate Research 403 (2015) 60–68

used in the preparation of enoxaparin, to obtain a product consis- properties are often undesirable because of possible bleeding
tently richer in ATBR and with higher affinity for AT than enoxap- effects), heparins, required for modulating most of the non-AT-
arin,57 from which an octadecasaccharide containing three ATBRs mediated activities should be non-anticoagulant. The anticoagu-
in a single chain was isolated.58 lant activity of heparin species can be substantially decreased
In parallel with similar efforts with heparan sulfates, a number either by removing their chains with high affinity for AT, or by
of heparin oligosaccharides have been isolated over the years, modifying some of the ATBR groups or units that are essential for
mainly as fragments of heparin lyases- or nitrous acid-depolymer- high-affinity binding to AT. The first approach seems still not eco-
ized UHFs and LMWHs. A list of about one hundred heparin oligo- nomically feasible. As evident from inspection of the structure of
saccharides (from tetrasaccharide to tetradecasaccharide) the pentasaccharide sequence of the ATBS (Fig. 1B), extensive mod-
identified up to the year 2001 (mainly by Linhardt–Toida’s, Lin- ification of the sulfation pattern of heparin (as easily performed by
dahl’s, Sugahara’s and Gallagher–Turnbull’s groups) is reported in solvolytic desulfation or with controlled alkaline reaction)68–70
Ref. 4. More recently, the development of methods for heparin/ would remove some of the essential sulfate groups and decrease
HS sequencing39–41 has permitted to add many more oligosaccha- the anticoagulant properties. Also other partial or extensive modi-
rides to that list. Linhardt’s group isolated and fully characterized fications of heparin68–70 may involve loss of anticoagulant proper-
in heparin lyase I digests of porcine mucosal heparin up to 22 oli- ties, but the biological properties of most of these derivatives still
gosaccharides with size ranging from disaccharide to hexadecasac- do not seem to be systematically investigated. Removal of the
charide; notably, quite a few of these oligosaccharides terminate ATBR marker, that is, the 3-O-sulfate of the central GlcNSO33,6SO3,
with a reducing GlcNSO33,6SO3 residue.53 A number of oligosac- A⁄) residue of the pentasaccharide would result in an especially
charides containing the intact ATBR sequence were isolated from drastic decrease of the anticoagulant properties.32 A way of ‘inacti-
the LMWH enoxaparin.51 (See also Section 9). vating’ the ATBR consists in glycol-splitting, by periodate oxida-
Some attempts to preserve and increase the content of ATBR- tion, all nonsulfated uronic acid residues of heparin, including
containing chains as compared with that of current LMWHs have the GlcA residue in the ATBR proved to be essential for high-affin-
met with success. These achievements were made both by using ity binging to AT.71 Among the observed non-AT-related effects of
heparin-lyases III that do not cleave the active pentasaccharide heparins,67 only two extensively investigated, that is, interaction
sequence43,55 or by finding alkaline conditions for the b-elimina- with fibroblast growth factors and heparanase, are shortly
tion reaction that largely preserve the ATBR sequence.57 reviewed here. (For other non-AT-dependent structure–activity
studies using libraries of heparin derivatives and fragments stud-
8. More than anticoagulation ied, u.a., as inhibitors of chemokines and b-secretase, see Refs.
72,73, respectively.)
8.1. Targeting heparin-binding proteins other than
antithrombin 8.2. Interactions with growth factors

Besides to AT, heparin binds to a large number of proteins and Fibroblast growth factors (FGFs) utilize cell surface HS as a
modulates their biological functions.3,59–62 In fact, ‘heparin-bind- co-receptor in the assembly of signaling complexes with FGF-
ing proteins’ are so defined because binding to heparin is currently receptors on the plasma membrane. The best known members
used for their separation from other proteins, even if their actual of their family, which control angiogenesis and wound healing,
physiological binding polysaccharide is heparan sulfate (HS), are FGF1 and FGF2. The molecular mechanism for their activa-
which is constituted by very much the same sequences of heparin tion involves complexation of the FGF with a HS chain, which
but in very different proportions and arrangements.62 The biosyn- favors dimerization of the cytokine and also acts as template
thesis of HS is similar to that of heparin,22 but its post-polymer for formation of a ternary complex involving FGF receptors.
modification proceeds less homogeneously and is interrupted at (Extensively reviewed in Ref. 74). First indication on structural
earlier stages than for heparin, leaving blocks of nonsulfated requirements for heparin sequences that bind to FGF1 and
sequences –GlcA–GlcNAc–(NA regions, occasionally 6-O-sulfated) FGF2, especially on the requirement of 6-O-sulfate groups in
along with N,O-sulfated sequences (NS blocks) including also the the case of FGF1 were obtained using selectively desulfated
trisulfated disaccharides (TSD) units found in heparin. Several NA heparins as models.75
and NS blocks are variously interrupted by mixed sequences.62 More detailed knowledge on the heparin/HS FGF-binding
Although the heparin-like sequences are often involved in HS-pro- sequences, such as identification of the minimal ones required
tein interactions, the intrinsic structural diversity and organ and for binding, i.g., the –IdoA2SO3–GlcNSO3,6SO3–IdoA2SO3– motif
tissue specificities of HSs suggest that also specific NA and/or for FGF1 and –IdoA2SO3–GlcNSO3–IdoA2SO3– for FGF2, was subse-
mixed sequences may contribute to binding and biological proper- quently obtained by Lindahl’s and other groups using libraries of
ties of this intriguing polysaccharide.63 However, unravelling spec- oligosaccharides (reviewed in Ref. 4). Also synthetic oligosaccha-
ificities is complicated by current finding that some variability in rides (such a tetrasaccharide76 and a hexasaccharide77) were used
the sulfation pattern of a given heparin/HS sequence is compatible for studying the sequence and size requirements for heparin bind-
with efficient binding to the same protein.60 ing to FGF2. Structural requirements for formation of ternary hep-
Perception of the biological importance of HS and biochemical arin/FGF/FGFR-receptors are still not well defined. For FGF1, a
studies on this GAG have been initially slow, both because of its heparin octasaccharide is the shortest oligomer capable of dimeriz-
complexity and the low amounts obtainable from animal cells ing the growth factor and assembling the active ternary com-
and tissues. For this reason, heparin and heparin fragments have plex.78,79 Diversification of the structural determinants of FGFs–
been often used as a surrogate of HS sequences for obtaining pre- heparin interactions and implications for binding specificity were
liminary structure–activity information.62 These studies led to also established.80
realize that there was ‘more to heparin than anticoagulation’,64,65 Among the non-anticoagulant heparins, some glycol-split deriv-
and there were novel drug development opportunities for atives (prepared from partially 2-O-desulfated heparins to obtain
heparin.66,67 up to a total of about 50% glycol-split uronic acids) showed to be
Since most of the emerging therapeutic exploitations of heparin efficient antagonists of FGF2 and potent angiogenesis
do not require the ATBR (in fact, anticoagulant/antithrombotic inhibitors.81,82
B. Casu et al. / Carbohydrate Research 403 (2015) 60–68 65

8.3. Non-anticoagulant heparins as heparanase inhibitors equilibria of internal sulfated iduronate residues.105 As illustrated
in Ref. 106, Mulloy’s structures of heparin are represented by heli-
The multiple functions of heparanase in cancer,83 inflammation ces with alternate triplets of sulfate groups on both sides of the
and other diseases84 are now widely recognized. Most of these chains irrespective of the conformation of the iduronate residues.
functions are associated with the enzymatic activity of heparanase, This shape is intrinsically the result of the a-L-a-D- alternate glyco-
which, acting as a b-D-glucuronidase, cleaves the HS chains at the sidic linkages of iduronate and glucosamine residues in the repeat-
level of GlcA residues. Since the first reports on structural require- ing TSD units. Analysis of available crystal coordinates of heparin
ments of heparin/HS to act as substrate for the enzyme,85 non-anti- oligosaccharide-protein complexes has shown that the linear prop-
coagulant heparins were investigated as inhibitors of the agation of heparin chains is interrupted at the level of the binding
enzymatic activity of heparanase, and some of them (especially sites to proteins by ‘kinks’ generated by changes in local conforma-
glycol-split heparins) were found to be promising as antimetastatic tion as would be observed on going from a 1C4 to 2S0 form.107 A crit-
and anticancer agents.86 These results are discussed in wider con- ical overview of the influence of substitution pattern and cation
texts in Refs. 87,88. Structural requirements of heparin chains to binding on conformation and activity of heparin derivatives is
act as heparanase substrates or inhibitors were also studied using reported in Ref. 108.
chemo-enzymatically synthesized heparin oligosaccharides.89 In
the framework of further studies on heparin-derived heparanase 9.2. Conformation of protein-bound sequences
inhibitors, NMR and LC/MS methods have been developed for
structural characterization of glycol-split heparins90 and glycol-split Complexation with proteins is among the extrinsic factors that
LMWHs.91 Microarray assays,92 and saturation-transfer difference can drive the conformations toward a unique form. Conceivably,
(STD) NMR measurements93 were used for biological evaluation only one of the favored conformations of iduronate residues is rep-
(including inhibition of heparanase) of synthetic heparin mimetic resented in co-crystal structures. Thus, in co-crystals of a complex
hexasaccharides.93 with AT and a sulfated pentasaccharide, the IdoA2SO3 residue was
found to be 100% in the 2S0 form.109 Such a AT-complexation-
9. Conformational aspects induced drive toward the 2S0 conformation was found also in solu-
tion.110 Milestone papers unraveled the influence of structure,
9.1. Conformation of unbound heparin sequences chain length and local conformations in the regulation of thrombin
activity by AT and heparin,111 and provided evidence for an
In mid 1980s, the molecular conformation of GAG component induced-fit model of allosteric activation of AT112 and for the
residues was a matter of debate, especially as regards the IdoA- involvement of the unique trisaccharide toward the nonreducing
containing sequences, such as those of heparin/HS and dermatan end of the ATBR in mediating the early steps of AT activation.113
sulfate, with apparently conflicting results between data obtained A notable example of protein-binding-induced selection of a local
from kinetics of periodate oxidation and from emerging NMR indi- conformation was the finding that in co-crystals of FGF-2 and a
cations from synthetic methyl iduronates20 and urinary dermatan heparin hexasaccharide containing two IdoA2SO3 residues, one of
sulfate.94,95 NMR analysis of the synthetic pentasaccharide, corre- these residues is in the 1C4 and the other in the 2S0 conforma-
sponding to the ATBR of heparin, provided evidence for the confor- tion.114 (See Ref. 4). However, 1C4 is the only conformation in solu-
mational peculiarity of its sulfated iduronate residue.30 A full tion of IdoA2SO3 residues of synthetic heparin/HS tetrasaccharides
analysis of the NMR spectra of heparin and synthetic heparin complexed with both FGF1115 and FGF2.76 A recent, still unex-
mono- and oligosaccharides combined with molecular mechanics plained but certainly meaningful example of different conforma-
calculations clearly showed that IdoA2SO3 and IdoA residues are tions selected in the solid state by apparently similar sulfated
in a dynamic conformational equilibrium among three iso-ener- iduronate residues of heptasaccharidic and tetrasacccharidic sub-
getic forms (1C4, 2S0 and 4C1),96,97 as predicted by a theoretical strates of two different sulfotransferases (3OST-1 and 3OST-3) is
study on iduronate monosaccharides.98 The conformer populations illustrated in Figure 4.116
of the three forms could widely differ depending on sulfation on A series of papers deals with the structural and biochemical
the same residue or in that of neighbor residues, as well as on effects of chain extension of the ATBR. The first of these contribu-
extrinsic factors such as the presence of calcium ions.97 Notably, tions analyzed the crystallographic coordinates of the AT complex
whereas IdoA2SO3 residues in internal ‘regular’ TDS sequences with a synthetic heptasaccharide corresponding to the pentasac-
show a relative population of conformers 1C4:2S0 of about 60:40, charidic active sequence extended by a disaccharidic unit toward
such a ratio is practically reversed (to 40:60) in favor of the 2S0 the reducing end. Its somewhat higher affinity for AT is explainable
form when the adjacent GlcNSO36SO3 residue is 3-O-sulfated, as by additional contacts with AT exerted by the disaccharide elonga-
in the ATBR.96,97,99 Since the relative orientations of the substituent tion.117 Similarly, in NMR/molecular mechanics studies in solution
groups significantly differ in the two conformations, involving, i.e., on octa- and higher heparin oligosaccharides isolated from the
different spacing between sulfate and/or carboxylate groups, such LMWH enoxaparin (reviewed in Ref. 51), elongation (with disac-
a behavior was soon perceived to have an important role on bind- charidic units unmodified by the depolymerization process) of
ing and associated biological properties of iduronic acid-containing the ATBR toward both the reducing and the non-reducing end
GAGs.100 Studies on synthetic pentasaccharides with iduronate allowed better contacts with AT and higher affinity for the protein.
residues ‘locked’ in the 2S0 conformation reinforced the concept Noteworthy, an octasaccharide featuring an unusual GlcA resi-
that this local internal flexibility (also referred to as ‘plasticity’) due just preceding the ATBR showed to have a tenfold higher affin-
favors the adjustment of IdoA/IdoA2SO3-containing sequences to ity and more favorable contacts with the AT basic residues than the
relevant binding sites of proteins.101 As exemplified for the penta- homologous octasaccharide with IdoA in the same position as nor-
saccharide fondaparinux, 15N NMR chemical shifts of the sulfamido mally found in HA chains isolated from porcine mucosal hepa-
groups can provide additional conformational information.102 rin.118 Also, as expected from X-ray data on a synthetic variant of
NMR, molecular dynamics and modeling studies by Mulloy’s group the ATBR pentasaccharide,109 an octasaccharide containing a 3-O-
generated the presently accepted 3D structure for the ‘regular’ sulfate group also in the other GlcNSO36SO3 residue of the ATBR
regions of heparin chains with iduronate residues in different local had higher affinity and better contacts than its homologous ‘nor-
conformations.103,104 NMR and molecular dynamics studies on a mal’ octasaccharide.119 Although some literature data (reviewed
heparin-derived hexasaccharide further confirmed conformational in Ref. 46) indicated that some chains of LMWHs could contain
66 B. Casu et al. / Carbohydrate Research 403 (2015) 60–68

(A)

(B)

Figure 4. Conformation of heparin/HS oligosaccharides in co-crystals with 3-O-sulfotransferases.116 (A) Chemoenzymatically synthesised heptasaccharide substrate
crystallized with 3OST-1; (B) tetrasaccharide generated by cleavage of A with heparin lyase-I, crystallized with 3OST-3. Note that the conformation of the central IdoA2SO3
residue of A is 1C4, while in B is 2S0.

more than one ATBR,49 rather unexpectedly a dodecasaccharide ing whether such an approach could afford heparin/HS-like spe-
containing two pentasaccharidic ATBR separated by a nonsulfated cies. Our group participated in the project by investigating the
IdoA residue was isolated from enoxaparin; interestingly, one of possibility of sulfation at specific positions both of unmodified
the two pentasaccharide sequences binds to AT more strongly than K5 backbone and derivatives obtained by enzymatic partial C-5-
the other and the two types of complexes are in a dynamic equilib- epimerization of GlcA residues. After chemical N-deacetylation
rium120 On the other hand, in octasaccharides terminating with the and N-sulfation, a selective O-sulfation at position 6 of the GlcNAc
1,6-anhydro residues typical of enoxaparin, the terminal units residues was achieved. Under different conditions, a selective 3-O-
were found not contribute to the AT binding, thus rationalizing sulfation of GlcA, and complete O-sulfation of the polysaccharide
the lower affinity for AT (and lower anti-Xa activity) of these spe- were obtained.126 Some of these K5 derivatives inhibited Factor
cies.121 As regards the conformational aspects, all the above-men- Xa, with potential antithrombotic properties.127 In a parallel study,
tioned ATBR-containing oligosaccharides were found to bind to AT conditions for C-5 epimerization of GlcA residues were optimized
with the same geometry as the (core) pentasaccharide; in all of to achieve up to 50% conversion to IdoA.128 Chemical O-sulfation
them, the IdoA2SO3 residue is in the 2S0 form. However, in the of N-sulfated, C-5-epimerized HMW and LMW K5 polysaccharide
AT-binding oligosaccharides so far studied, the iduronate residues, afforded several derivatives, which were significantly 3-O-sulfated
irrespective of being sulfated or nonsulfated, select in the bound at the aminosugar, but almost invariably also at the GlcA residue.
state either the 2S0 or the 1C4 conformation depending on their Efforts to reduce the unnatural 3-O-sulfation of GlcA and increase
position along the oligosaccharide chain even in those cases in the content of N,3-O-sulfated GlcN by exploiting a strategy of
which they preferred in the unbound state the other conforma- graded solvolyic desulfation of fully sulfated, partially 5-O-epimer-
tion.51 While confirming that the conformational plasticity plays ized K5, have generated products with biological properties similar
a decisive role in modulating the protein binding of heparin to those of heparin and LMWHs.129 Thus, although the structure of
sequences, these results imply that the conformation of iduronate the obtained ‘neoheparins’ significantly differed from that of hep-
residues that predominate in the absence of binding proteins is not arins of animal origin, the above studies showed that some of the
necessarily the same adopted in the bound state. Conceivably, the biological properties of these latter could be simulated and
most favored conformation of heparin/HS sequences in the bound modulated.
state is determined by a balance of several interactions, to which A more recent project of an U.S.A. consortium exploiting a full
the plastic iduronate residue contributes in different ways depend- series of recombinant biosynthetic enzymes had afforded homoge-
ing on the structure and conformation of the active site of the neous ULMW heparins and has great potential for generating hep-
protein. arin/HS oligosaccharides with almost unlimited possibilities for
Structural data and concepts on heparin complexes with pro- tailoring specific sequences.130
teins other than AT are presented and discussed in recent mini-
reviews.122–125 11. The ‘heparin crisis’ and its (beneficial) fall-outs

10. Toward a biotechnological heparin In late 2007 and early 2008 a series of adverse events in patients
receiving heparin injections occurred in U.S.A. and in some Euro-
Since early 1990, concerns on possible shortage of heparin of pean countries, causing more than one hundred deaths. As a reac-
animal origin stimulated studies aimed at generating heparins by tion to these dramatic events, the U.S. Food and Drug
exploiting available biosynthetic enzymes in combination with Administration (FDA) recalled suspected heparin vials and orga-
regioselective chemical modification reactions. Discovery of the nized their analysis by a consortium of academic groups (including
microbial polysaccharide K5, which is exclusively constituted by our own), coordinated by Ram Sasisekharan. It was soon shown
(GlcA–GlcNAc)n sequences as the heparin biosynthetic precursor that the adverse events were caused by an ‘unnatural’ contami-
N-acetyl heparosan, triggered an European project aimed at explor- nant, a fully O-sulfated chondroitin sulfate.131
B. Casu et al. / Carbohydrate Research 403 (2015) 60–68 67

The realization that the current methods of pharmacopoeial 5. Linhardt, R. J. J. Med. Chem. 2003, 46, 2551–2564.
6. Chemistry and Biology of Heparin and Heparan Sulfate; Garg, H. G., Linhardt, J.,
controls for heparin were inadequate to detect unexpected con-
Hales, C. A., Eds.; Elsevier: Amsterdam, 2005.
taminants triggered an explosion of analytical studies and propos- 7. Heparin—A Century of Progress; Lever, R., Mulloy, B., Page, C. P., Eds.; Springer:
als for orthogonal approaches, most of which involving Berlin, 2012.
combination of NMR and separation methods. A survey of these 8. Bigelow, W. G. Misterious Heparin; McGraw-Hill Ryerson: Toronto, 1990.
9. Rodén, L.; Ananth, S.; Campbell, P.; Crenton, T.; Ekborg, G.; Manzella, S.;
novel analytical methods (examples of which are reported in Refs. Pillion, D.; Meezan, E. In Ref. 2, 1992; pp 1–20
132–135) and on their expected contribution to an increased safety 10. Barrowcliffe, T. W. Ref. 7, 2012; pp 3–22.
in the use of pharmaceutical heparins is outside the scope of this 11. Wolfrom, M. L.; Rice, F. A. J. Am. Chem. Soc. 1946, 68, 532.
12. Helting, T.; Lindahl, U. J. Biol. Chem. 1971, 246, 5442–5447.
review. However, it is worth mentioning that some sophisticated 13. Cifonelli, J. A.; Dorfman, A. Biochem. Biophys. Res. Commun. 1962, 7, 41–46.
methods for obtaining and analyzing NMR data, such as the two- 14. Perlin, A. S.; Mazurek, M.; Jaques, L. B.; Kavanagh, L. W. Carbohydr. Res. 1968,
dimensional correlation spectroscopy-filtering with iterative radia- 7, 369–379.
15. Perlin, A. S.; Casu, B.; Sanderson, G. R.; Johnson, L. F. Can. J. Chem. 1970, 48,
tion sampling136 and statistical correlation two-dimensional 2260–2268.
(HSQCcos) spectroscopy,137 contributed not only to detect 16. Perlin, A. S.; Sanderson, G. S. Carbohydr. Res. 1970, 12, 183–192.
unknown contaminants, but also to identify previously undetected, 17. Lindahl, U.; Axelsson, O. J. Biol. Chem. 1971, 246, 74–82.
18. Wolfrom, M. L.; Honda, S.; Wang, P. Y. Carbohydr. Res. 1969, 10, 185–259.
very minor components of heparin. Among these components, 19. Perlin, A. S.; Mackie, D. M.; Dietrich, C. P. Carbohydr. Res. 1971, 18, 145–194.
internal N-unsubstituted GlcN,6SO3 residues and 3-O-sulfated 20. Perlin, A. S.; Casu, B.; Tse, J.; Sanderson, G. R. Carbohydr. Res. 1972, 21, 123–
GlcNSO3,6SO3 residues at the nonreducing terminal of heparin 132.
21. Gatti, G.; Casu, B.; Hamer, A. S.; Perlin, A. S. Macromolecules 1979, 12, 1001–
chains were recently identified in unfractionated heparin.137 New
1007.
criteria for characterization of currently marketed heparin prod- 22. Carlsson, P.; Kjellén, L. Ref. 7, 2012; pp 23–41.
ucts were reported by the Federal Food and Drug Administration 23. Mulloy, B. Ref. 7, 2012; pp 77–98.
(FDA) agency.138 24. Bertini, S.; Bisio, A.; Torri, G.; Bensi, D.; Terbojevich, M. Biomacromolecules
2005, 6, 168–173.
25. Lindahl, U.; Bäckstro } m, G.; Thunberg, L.; Leder, I. G. Proc. Natl. Acad. Sci. U.S.A.
12. Open issues and perspectives 1980, 77, 6551–6555.
26. Thunberg, L.; Bäckstro }m, G.; Lindahl, U. Carbohydr. Res. 1982, 100, 393–410.
27. Lindahl, U.; Thunberg, L.; Bäckstro } m, G.; Riesenfeld, J.; Nordling, K.; Bjo
}rk, I. J.
In spite of its being almost 100 years old, heparin has success- Biol. Chem. 1984, 259, 12368–12376.
fully overcome critical times such as the just mentioned crisis 28. Casu, B.; Oreste, P.; Torri, G.; Zoppetti, G.; Lormeau, J. C.; Petitou, M.; Sinaÿ, P.
Biochem. J. 1981, 197, 599–609.
and has not lost its reputation as a life-saving drug.139 Although,
29. Loganathan, D.; Wang, H. M.; Mallis, L. M.; Linhardt, R. J. Biochemistry 1990,
for one side, heparin is facing competition from novel synthetic 29, 4362–4368.
antithrombotics, from the other side most of the prospective devel- 30. Choay, J.; Petitou, M.; Lormeau, J.-C.; Sinaÿ, P.; Casu, B.; Gatti, G. Biochem.
Biophys. Res. Commun. 1983, 116, 492–499.
opments for heparin-related oligo- and polysaccharides as HS
31. Petitou, M.; Casu, B.; Lindahl, U. Biochimie 2003, 85, 83–89.
mimics140 are widely open. In spite of the steady advances in 32. Van Boeckel, C. A. A.; Petitou, M. Angew. Chem., Int. Ed. 1993, 32, 1671–1688.
methods of structural characterization of heparin and understand- 33. Petitou, M.; van Boeckel, C. A. Angew. Chem., Int. Ed. 2004, 43, 3118–3133.
ing of its interactions with proteins mostly associated with increas- 34. Dementiev, A.; Petitou, M.; Herbert, J. M.; Gettings, P. G. Nat. Struct. Biol. 2004,
11, 863–867.
ing interest in structure and functions of HS and HS 35. Yamada, S.; Murakami, T.; Tsuda, H.; Yoshida, K.; Sugahara, K. J. Biol. Chem.
proteoglycans,141 there are still several open issues in the field that 1995, 270, 8696–8705.
could easily fill the professional life of other generations of 36. Chai, W.; Hounsell, E. F.; Bauer, C. J.; Lawson, A. M. Carbohydr. Res. 1995, 269,
139–156.
researchers. For example, understanding of biological function 37. Iacomini, M.; Casu, B.; Guerrini, M.; Naggi, A.; Pirola, A.; Torri, G. Anal.
and potential pharmaceutical exploitation of 3-O-sulfated GlcN Biochem. 1999, 274, 50–58.
residues in different sequences of heparin and HS is still at its 38. Casu, B.; Torri, G.; Vercellotti, J. R. Pharmacol. Res. Comm. 1979, 11, 297–309.
39. Venkataraman, G.; Shriver, Z.; Raman, R.; Sasisekharan, R. Science 1999, 86,
beginning.142 In fact, in spite of the many pathophysiological func- 537–542.
tions continuously discovered for HSs, only a few drugs exploiting 40. Turnbull, J. E.; Hopwood, J. J.; Gallagher, J. T. Proc. Natl. Acad. Sci. U.S.A. 1999,
these functions are being developed.140 96, 2698–2703.
41. Linhardt, R. J.; Rice, K. G.; Kim, Y. S.; Loshe, D. L.; Wang, H. M.; Loganathan, E.
D. Biochem. J. 1988, 254, 781–787.
Acknowledgements 42. Shriver, Z.; Capila, I.; Venkataraman, R. Ref. 7, 2000, pp 159–175.
43. Xiao, Z.; Tappen, B. R.; Ly, M.; Zhao, W.; Canova, L. P.; Guan, H.; Linhardt, R. J. J.
Med. Chem. 2011, 54, 603–610.
The authors gratefully thank Ulf Lindahl and Maurice Petitou for 44. Gray E.; Hogwood, J.; Mulloy, B. Ref. 7, 2012; pp 43–61.
critical reading of the manuscript and Antonella Bisio, Giuseppe 45. Andersson, L.-O.; Barrowcliffe, T. V.; Holmer, E.; Johnson, E. A.; Sims, G. E.
Thromb. Res. 1976, 6, 575–583.
Cassinelli and Marco Guerrini for useful discussions. Two of them 46. Guerrini, M.; Bisio, A. Ref. 7, 2012; pp 127–157.
(B.C. and G.T.) wish to express their gratitude to Arthur Perlin for 47. Linhardt, R. J.; Loganathan, D.; Al-Hakim, A.; Wang, H.-M.; Walenga, J. M.;
introducing them into the heparin field. Part of the work described Hoppensteadt, D.; Fareed, J. J. Med. Chem. 1990, 33, 1639–1645.
48. Guerrini, M.; Guglieri, S.; Naggi, A.; Sasisekharan, R.; Torri, G. Semin. Thromb.
in this mini-review has been financially supported by several agen- Hemost. 2007, 33, 478–487.
cies. Among current Grants, those from National Institutes of 49. Bisio, A.; Vecchietti, D.; Citterio, L.; Guerrini, M.; Raman, R.; Bertini, S.; Eisele,
Health, United States (Number R01-CA138535) and Finlombardia G.; Naggi, A.; Sasisekharan, R.; Torri, G. Thromb. Haemost. 2009, 102, 865–873.
50. Fareed, J.; Haas, S.; Sahahara, E. Semin. Thromb. Hemost. 1999, 3, 1–14.
SpA, Italy (Fondo promozione accordi istituzionali) are acknowl-
51. Guerrini, M.; Mourier, P.; Torri, G.; Viskov, C. Glycoconjugate J. 2014. in press.
edged. The authors wish to thank also the Villa Vigoni Foundation 52. Shriver, Z.; Sundaram, M.; Venkataraman, G.; Fareed, J.; Linhardt, J. R.;
for hosting, since more than twenty years, yearly Glycosaminogly- Biemann, K.; Sasisekharan, R. Proc. Natl. Acad. Sci. U.S.A. 2000, 97, 10365–
can Symposia, with special emphasis on heparin. 10370.
53. Xiao, Z.; Zhao, W.; Yang, Z.; Zhang, Z.; Guan, H.; Linhardt, R. J. Glycobiology
2011, 21, 13–22.
54. Harenberg, J.; Walenga, J.; Torri, G.; Dahl, O. E.; Drouet, L.; Fareed, J. J. Thromb.
References Haemost. 2013, 334, 1421–1425.
55. Sundaram, M.; Shriver, Z.; Zhao, G.; Venkataraman, G.; Langer, R.;
1. Casu, B. In Adv. Carbohydr. Chem. Biochem.; Stuart Tipson, R., Derek Horton, Sasisekharan, R. Proc. Natl. Acad. Sci. U.S.A. 2003, 100, 651–656.
Eds.; , 1985; Vol. 43, pp 51–134. 56. Xu, Y.; Masuko, S.; Takieddin, M.; Xu, H.; Liu, R.; Jing, J.; Mousa, S. A.; Linhardt,
}rk, I., Lindahl, U., Eds.; Plenum
2. Adv. Carbohydr. Chem. Biochem.; Lane, D. A., Bjo R. J.; Liu, J. Science 2011, 334, 498–501.
Press: New York, 1992. 57. Lima, M. A.; Viscov, C.; Herrman, F.; Gray, A. L.; de Faris, E. H.; Cavalheir, R. P.;
3. Conrad, E. Heparin Binding Proteins; Academic Press: San Diego, CA, 1998. Sassaki, G. L.; Hoppensteadt, D.; Fareed, J.; Nader, H. B. Thromb. Haemost. 2013,
4. Casu, B.; Lindahl, U. Adv. Carbohydr. Chem. Biochem. 2001, 57, 159–206. 109, 471–478.
68 B. Casu et al. / Carbohydrate Research 403 (2015) 60–68

58. Mourier, P. A.; Guichard, O. Y.; Herman, F.; Viskov, C. Anal. Biochem. 2014, 15, 103. Mulloy, B.; Forster, M. J.; Jones, C.; Davies, D. B. Biochem. J. 1993, 293, 849–858.
453–457. 104. Mulloy, B.; Forster, M.; Jones, C.; Davies, D. B. Glycobiology 2000, 10, 1147–
59. Capila, I.; Linhardt, R. J. Angew. Chem., Int. Ed. 2002, 41, 391–412. 1158.
60. Kreuger, J.; Spillmann, D.; Li, J. P.; Lindahl, U. J. Cell Biol. 2006, 174, 323–327. 105. Mikhailov, D.; Linhardt, R. J.; Mayo, K. H. Biochem. J. 1997, 328, 51–61.
61. Lindahl, U. Thromb. Haemost. 2007, 96, 109–115. 106. Mulloy B.; Ref. 7, 2012; pp 77–98.
62. Gallagher, J.T. Ref. 7, 2012; pp 347–360. 107. Raman, R.; Sasisekharan, V.; Sasisekharan, R. Chem. Biol. 2005, 12, 267–277.
63. Xu, D.; Esko, J. D. Annu. Rev. Biochem. 2014. in press. 108. Rudd, T. R.; Guimond, S. E.; Skidmore, M. A.; Duchesne, R.; Guerrini, M.; Torri,
64. Lindahl, U.; Lidholt, K.; Spillmann, D.; Kjellén, L. Thromb. Res. 1994, 75, 1–32. G.; Cosentino, C.; Brown, A.; Clarke, D. T.; Turnbull, J. E.; Fernig, D. G.; Yates, E.
65. Lindahl, U. Glycoconju. J. 2000, 17, 597–605. A. Glycobiology 2007, 17, 983–993.
66. Lever, L.; Page, C. P. Nat. Rev. Drug Disc. 2002, 57, 159–206. 109. Jin, L.; Abrahams, J. P.; Skinner, R.; Petitou, M.; Pike, R. N.; Carrell, R. W. Proc.
67. Lever, R.; Page, C.P.; Ref. 7, pp 281–305. Natl. Sci. USA 1997, 14683–14688.
68. Yates, E. A.; Santini, F.; Guerrini, M.; Naggi, A.; Torri, G.; Casu, B. Carbohydr. 110. Hricovini, M.; Guerrini, M.; Bisio, A.; Torri, G.; Petitou, M.; Casu, B. Biochem. J.
Res. 1996, 294, 15–27. 2001, 359, 265–272.
69. Fernandéz, C.; Hattan, C. M.; Kerns, R. J. Carbohydr. Res. 2006, 341, 1253–1265. 111. Olson, S. T.; Bjo}rk, I. Semin. Thromb. Hemost. 1994, 20, 373–409.
70. Yates, E. A.; Guimond, S. E.; Turnbull, J. E. J. Med. Chem. 2004, 47, 277–280. 112. Desai, U. R.; Petitou, M.; Bjo }rk, I.; Olson, S. T. Biochemistry 1998, 37, 13033–
71. Naggi, A. Ref. 6, 2005, pp 461–481. 13041.
72. Shute, J. Ref. 7, 2012; pp 307–324. 113. Petitou, M.; Barzu, T.; Herault, J.-P.; Herbert, J.-M. Glycobiology 1997, 7, 323–
73. Schwo }rer, R.; Zubkova, O. V.; Turnbull, J. E.; Tyler, P. C. Chemistry 2013, 19, 327.
6817–6823. 114. Faham, S.; Hileman, R. E.; Fromm, J. R.; Linhardt, R. J.; Rees, D. C. Science 1996,
74. Mohammadi, M.; Olsen, S. K.; Ibrahimi, O. A. Cytokine Growth Factor Rev. 2005, 271, 116–120.
16, 107–137. 115. Guerrini, M.; Agulles, T.; Bisio, A.; Hricovini, M.; Lay, L.; Paoletti, L.; Sturiale,
75. Maccarana, M.; Casu, B.; Lindahl, U. J. Biol. Chem. 1993, 268, 23898–23905. L.; Torri, G.; Casu, B. Biochem. Biophys. Res. Commun. 2002, 292, 222–230.
76. Guglieri, S.; Hricovini, M.; Raman, R.; Polito, L.; Torri, B.; Casu, B.; 116. Liu, J.; Moon, A. F.; Sheng, J.; Pedersen, L. C. Curr. Opin. Struct. Biol. 2012, 22,
Sasisekharan, R.; Guerrini, M. Biochemistry 2008, 47, 13862–13869. 570–577.
77. Sapa, N.; Cabannes, E.; Petitou, M.; Imberty, A. Glycobiology 2011, 21, 1181– 117. Belzar, K. J.; Dafforn, T. R.; Petitou, M.; Carrell, R. W.; Huntington, J. A. J. Biol.
1193. Chem. 2000, 275, 8733–8741.
78. Jastrebova, N.; Varwildemeersch, M.; Lindahl, U.; Spillmann, D. J. Biol. Chem. 118. Guerrini, M.; Guglieri, S.; Casu, B.; Torri, G.; Mourier, P.; Boudier, C.; Viskov, C.
2010, 285, 26842–26851. J. Biol. Chem. 2008, 283, 26662–26675.
79. Brown, A.; Robinson, C. J.; Gallagher, J. T.; Blundell, T. L. Biophys. J. 2013, 104, 119. Guerrini, M.; Elli, S.; Mourier, P.; Rudd, T. R.; Gaudesi, D.; Casu, B.; Boudier, C.;
1720–1730. Torri, G.; Viskov, C. Biochem. J. 2013, 449, 343–351.
80. Xu, R.; Ori, A.; Rudd, T. R.; Uniewicz, K. A.; Ahmed, Y. A.; Guimond, S. E.; 120. Viskov, C.; Elli, S.; Urso, E.; Gaudesi, D.; Mourier, P.; Herman, F.; Boudier, C.;
Skidmore, M. A.; Siligardi, G.; Yates, E. A.; Fernig, D. G. J. Biol. Chem. 2012, 287, Casu, B.; Torri, G.; Guerrini, M. J. Biol. Chem. 2013, 288, 25895–25907.
40061–40073. 121. Guerrini, M.; Elli, S.; Gaudesi, D.; Torri, G.; Casu, B.; Mourier, P.; Herman, F.;
81. Casu, B.; Guerrini, M.; Naggi, A.; Perez, M.; Torri, G.; Ribatti, D.; Carminati, P.; Boudier, C.; Lorenz, M.; Viskov, C. J. Med. Chem. 2010, 53, 8030–8040.
Giannini, G.; Penco, S.; Pisano, C.; Belleri, M.; Rusnati, M.; Presta, M. 122. Imberty, A.; Lortat-Jacob, H.; Pérez, S. Carbohydr. Res. 2007, 342, 430–439.
Biochemistry 2002, 41, 10519–10528. 123. Rudd, T. R.; Skidmore, M. A.; Guerrini, M.; Hricovini, M.; Powell, A. K.;
82. Casu, B.; Guerrini, M.; Guglieri, A.; Naggi, A.; Perez, M.; Torri, G.; Cassinelli, G.; Siligardi, G.; Yates, E. A. Curr. Opin. Struct. Biol. 2010, 20, 567–574.
Ribatti, D.; Carminati, P.; Giannini, G.; Penco, S.; Pisano, C.; Belleri, M.; Rusnati, 124. Casu, B.; Naggi, A.; Torri, G. Matrix Biol. 2010, 29, 442–452.
M.; Presta, M. J. Med. Chem. 2004, 47, 838–848. 125. Gonzáles-Corrochano, J.; Rolando-Horcajo, M.; Javier Cañada, F.; Nieto, P.;
83. Ramani, V. C.; Puroshothaman, A.; Stewart, M. D.; Thompson, C. A.; Vlodavsky, Martin-Lomas, M.; Giménez-Gallego, G.; Jiménez-Barbero, G. ChemBioChem
I.; Au, J. L.; Sanderson, R. D. FEBS J. 2013, 280, 2294–2306. 2013, 14, 1732–1744.
84. Vlodavsky, I.; Iozzo, R. V.; Sanderson, R. D. Matrix Biol. 2013, 32, 220–222. 126. Casu, B.; Grazioli, M.; Guerrini, M.; Naggi, A.; Torri, G.; Oreste, P.; Tursi, F.;
85. Sandbäck-Pikas, D.; Li, J. P.; Vlodavsky, I.; Lindahl, U. J. Biol. Chem. 1998, 273, Zoppetti, G.; Razi, N.; Feizi, E.; Lindahl, U. Carbohydr. Res. 1994, 263, 271–284.
18870–18877. } rk, I.; Naggi, A.; Casu, B.; Lindahl, U. Biochem. J. 1995, 309,
127. Razi, N.; Feizi, E.; Bjo
86. Naggi, A.; Casu, B.; Perez, M.; Torri, G.; Cassinelli, G.; Penco, S.; Pisano, C.; 429–430.
Giannini, G.; Ishai-Michaeli, R.; Vlodavsky, I. J. Biol. Chem. 2005, 280, 12103– 128. Oreste, P.; Zoppetti, G. Ref. 7, 2012; pp 403–422.
12113. 129. Lindahl, U.; Li, J.-P.; Kusche-Gullberg, M.; Salmivirta, M.; Alaranta, S.;
87. Vlodavsky, I.; Ilan, N.; Naggi, A.; Casu, B. Curr. Pharm. Des. 2007, 13, 2057– Veromaa, T.; Emeis, J.; Roberts, I.; Trevor, C.; Oreste, P.; Naggi, A.; Torri, G.;
2073. Casu, B. J. Med. Chem. 2005, 48, 349–352.
88. Casu, B.; Vlodavsky, I.; Sanderson, R. D. Pathophysiol. Haemost. Thromb. 2007, 130. Xu, Y.; Cai, C.; Chandarajoti, K.; Hsieh, P. H.; Li, L.; Pham, T. Q.; Sparkenbaugh,
36, 195–203. J.; Sheng, J.; Key, N. S.; Pawlinski, R.; Harris, E. N.; Linhardt, R. J.; Liu, J. Nat.
89. Peterson, S.; Liu, J. J. Biol. Chem. 2012, 287, 34836–34843. Chem. Biol. 2014, 10, 248–250.
90. Alekseeva, A.; Casu, B.; Torri, G.; Pierro, S.; Naggi, A. Anal. Biochem. 2013, 434, 131. Guerrini, M.; Beccati, D.; Shriver, Z.; Naggi, A.; Viswanathan, K.; Bisio, A.;
112–122. Capila, I.; Lansing, J. C.; Guglieri, S.; Fraser, B.; Al-Akim, A.; Gunay, N. S.; Zhang,
91. Alekseeva, A.; Casu, B.; Cassinelli, G.; Guerrini, G.; Torri, G.; Naggi, A. Anal. Z.; Robinson, L.; Buhse, L.; Nasr, M.; Woodcock, I.; Langer, R.; Venkataraman,
Bioanal. Chem. 2014, 406, 249–265. G.; Linhardt, R. J.; Casu, B.; Torri, G.; Sasisekharan, R. Nat. Biotechnol. 2008, 26,
92. Noti, C.; de Paz, J. L.; Polito, L.; Seeberger, P. H. Chem. Eur. J. 2006, 12, 8664– 669–675.
8686. 132. Guerrini, M.; Zhang, Z.; Shriver, Z.; Naggi, A.; Masuko, S.; Langer, R.; Casu, B.;
93. Roy, S.; El Hadri, A.; Richard, S.; Denis, F.; Holte, K.; Duffner, J.; Yu, F.; Linhardt, R. J.; Torri, G.; Sasisekharan, R. Proc. Natl. Acad. Sci. U.S.A. 2009,
Galcheva-Gargiova, Z.; Capila, I.; Schultes, B.; Petitou, M.; Kaundinya, G. V. J. 10616956–10616961.
Med. Chem. 2014, 115. 133. Korir, A.; Larive, C. K. Anal. Bioanal. Chem. 2009, 393, 155–169.
94. Gatti, G.; Casu, B.; Torri, G.; Vercellotti, J. R. Carbohydr. Res. 1979, 68, C3–C7. 134. Lima, M. A.; Rudd, T. R.; de Farias, E. H.; Ebner, L. F.; Gesteira, T. F.; de Souza, L.
95. Casu, B.; Choay, J.; Ferro, D. R.; Gatti, G.; Jacquinet, J.-C.; Petitou, M.; Provasoli, M.; Mendes, A.; Cordula, C. R.; Martins, J. R.; Hoppensteadt, D.; Fareed, J.;
A.; Ragazzi, M.; Sinaÿ, P.; Torri, G. Nature 1986, 322, 215–216. Sassaki, G. L.; Yates, E. A.; Tersarol, I. L.; Nader, H. B. PlosOne 2011, 6, e15970.
96. Ferro, D. R.; Provasoli, A.; Ragazzi, M.; Torri, G.; Casu, B.; Gatti, G.; Jacquinet, J.- 135. Shriver, Z.; Capila, I, Venkataraman, G.; Sasisekharan, R. Ref. 7, 2012; pp 159–
C.; Sinaÿ, P.; Petitou, M.; Choay, J. J. Am. Chem. Soc. 1986, 1908, 6773–6778. 176.
97. Ragazzi, M.; Ferro, D. R.; Perly, B.; Sinaÿ, P.; Petitou, M.; Choay, J. Carbohydr. 136. Rudd, T. R.; Macchi, E.; Gardini, C.; Muzi, L.; Guerrini, M.; Yates, E. A.; Torri, G.
Res. 1990, 195, 169–185. Anal. Chem. 2012, 84, 6841–6847.
98. Ragazzi, M.; Ferro, D. A.; Provasoli, A. J. Comput. Chem. 1986, 7, 105–112. 137. Rudd, T. R.; Macchi, E.; Muzi, L.; Ferro, M.; Gaudesi, D.; Torri, G.; Casu, B.;
99. Ferro, D. R.; Provasoli, A.; Ragazzi, M.; Casu, B.; Torri, G.; Bossenec, V.; Perly, Guerrini, M.; Yates, E. A. Anal. Chem. 2013, 85, 7487–7493.
B.; Sinaÿ, P.; Petitou, M.; Choay, J. Carbohydr. Res. 1990, 195, 157–167. 138. Ye, H.; Toby, T. K.; Sommers, C. D.; Ghastriani, H.; They, M. L.; Ye, W.; Kolinski,
100. Casu, B.; Petitou, M.; Provasoli, M.; Sinaÿ, P. Trends Biol. Sci. 1988, 13, 221–225. R. R.; Buhse, L. F.; Al-Akim, A.; Keire, D. A. J. Pharm. Biomed. Anal. 2013, 85, 99–
101. Das, S. K.; Mallet, J.-M.; Esnault, J.; Driguez, P.-A.; Duchaussoy, P.; Sizun, P.; 107.
Hérault, J.-P.; Herbert, J.-M.; Petitou, M.; Sinaÿ, P. Angew. Chem., Int. Ed. 2001, 139. Weitz, D. S.; Weitz, J. I. J. Thromb. Thrombolysis 2010, 29, 199–207.
40, 1670–1673. 140. Lindahl, U.; Kjellén, L. J. Int. Med. 2013, 273, 555–571.
102. Larngeslay, D. J.; Beecher, C. N.; Naggi, A.; Guerrini, M.; Torri, G.; Larive, C. K. 141. Lindahl, U. Matrix Biol. 2014, 36, 3–7.
Anal. Chem. 2013, 85, 1247–1255. 142. Thacker, B. E.; Xu, D.; Lawrence, R.; Esko, J. D. Matrix Biol. 2013, 35, 60–72.

You might also like