PreprintPDF Available
Preprints and early-stage research may not have been peer reviewed yet.

Abstract and Figures

Haemonchus contortus is the most pathogenic nematode of small ruminants. Infection in sheep and goats results in anaemia that decreases animal productivity and can ultimately cause death. The involvement of ruminant-specific galectin-11 (LGALS-11) and galectin-14 (LGALS-14) has been postulated to play important roles in protective immune responses against parasitic infection; however, their ligands are unknown. In the current study, LGALS-11 and LGALS-14 ligands in H. contortus were identified from larval (L4) and adult parasitic stages extracts using immobilised LGALS-11 and LGALS-14 affinity column chromatography and mass spectrometry. Both LGALS-11 and LGALS-14 bound more putative protein targets in the adult stage of H. contortus (43 proteins) when compared to the larval stage (2 proteins). Of the 43 proteins identified in the adult stage, 34 and 35 proteins were bound by LGALS-11 and LGALS-14, respectively, with 26 proteins binding to both galectins. Interestingly, hematophagous stage-specific sperm-coating protein and zinc metalloprotease (M13), which are known vaccine candidates, were identified as putative ligands of both LGALS-11 and LGALS-14. The identification of glycoproteins of H. contortus by LGALS-11 and LGALS-14 provide new insights into host-parasite interactions and the potential for developing new interventions. PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec
No caption available
… 
Content may be subject to copyright.
Proteomic identification of Galectin-11 and 14 ligands from
Haemonchus contortus
Dhanasekaran Sakthivel 1, 2, 3 , Jaclyn Swan 1 , Sarah Preston 4 , MD Shakif-Azam 3 , Pierre Faou 5 , Yaqing Jiao 4
, Rachel Downs 5 , Harinda Rajapaksha 5 , Robin Gasser 4 , David Piedrafita Corresp., 3 , Travis Beddoe Corresp. 1
1 Department of Animal, Plant and Soil Science and Centre for AgriBioscience (AgriBio), La Trobe University, Bundoora, Victoria, Australia
2 Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
3 School of Applied and Biomedical Sciences, Federation University, Churchill, Australia
4 Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, Australia
5 Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
Corresponding Authors: David Piedrafita, Travis Beddoe
Email address: david.piedrafita@federation.edu.au, t.beddoe@latrobe.edu.au
Haemonchus contortus is the most pathogenic nematode of small ruminants. Infection in
sheep and goats results in anaemia that decreases animal productivity and can ultimately
cause death. The involvement of ruminant-specific galectin-11 (LGALS-11) and galectin-14
(LGALS-14) has been postulated to play important roles in protective immune responses
against parasitic infection; however, their ligands are unknown. In the current study,
LGALS-11 and LGALS-14 ligands in H. contortus were identified from larval (L4) and adult
parasitic stages extracts using immobilised LGALS-11 and LGALS-14 affinity column
chromatography and mass spectrometry. Both LGALS-11 and LGALS-14 bound more
putative protein targets in the adult stage of H. contortus (43 proteins) when compared to
the larval stage (2 proteins). Of the 43 proteins identified in the adult stage, 34 and 35
proteins were bound by LGALS-11 and LGALS-14, respectively, with 26 proteins binding to
both galectins. Interestingly, hematophagous stage-specific sperm-coating protein and
zinc metalloprotease (M13), which are known vaccine candidates, were identified as
putative ligands of both LGALS-11 and LGALS-14. The identification of glycoproteins of H.
contortus by LGALS-11 and LGALS-14 provide new insights into host-parasite interactions
and the potential for developing new interventions.
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
              
Haemonchus contortus
Dhanasekaran Sakthivel1,2,3, Jaclyn Swan3, Sarah Preston2,4, MD Shakif-Azam2, Pierre Faou5,
Yaqing Jiao4, Rachel Downs5, Harinda Rajapaksha5, Robin B Gasser4, David Piedrafita2* and
Travis Beddoe3
1Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800,
Australia.
2School of Applied and Biomedical Sciences, Federation University, Churchill, Victoria 3842,
Australia.
3Department of Animal, Plant and Soil Science and Centre for AgriBioscience (AgriBio), La
Trobe University, Victoria 3086, Australia.
4 Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University
of Melbourne, Victoria, 3010, Australia.
5Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science La Trobe
University, Victoria 3086, Australia.
*Corresponding authors
Travis Beddoe; email: t.beddoe@latrobe.edu.au
David Piedrafita; email: david.piedrafita@federation.edu.au
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017

 is the most pathogenic nematode of small ruminants. Infection in sheep
and goats results in anaemia that decreases animal productivity and can ultimately cause death.
The involvement of ruminant-specific galectin-11 (LGALS-11) and galectin-14 (LGALS-14) has
been postulated to play important roles in protective immune responses against parasitic
infection; however, their ligands are unknown.In the current study, LGALS-11 and LGALS-14
ligands in  were identified from larval (L4) and adult parasitic stages extracts using
immobilised LGALS-11 and LGALS-14 affinity column chromatography and mass spectrometry.
Both LGALS-11 and LGALS-14 bound more putative protein targets in the adult stage of 
 (43 proteins) when compared to the larval stage (2 proteins). Of the 43 proteins
identified in the adult stage, 34 and 35 proteins were bound by LGALS-11 and LGALS-14,
respectively, with 26 proteins binding to both galectins. Interestingly, hematophagous stage-
specific sperm-coating protein and zinc metalloprotease (M13), which are known vaccine
candidates, were identified as putative ligands of both LGALS-11 and LGALS-14.The
identification of glycoproteins of  by LGALS-11 and LGALS-14 provide new
insights into host-parasite interactions and the potential for developing new interventions.
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017

 is a dominant blood feeding gastrointestinal nematode (GIN) parasite of
small ruminants. Blood feeding by  results in haemorrhagic gastritis, oedema,
diarrhoea and death in severe infections, leading to significant economic impact through
decreased livestock production (Mavrot et al. 2015; McLeod 1995; Roeber et al. 2013). Sheep
can develop effective immunity to  infection and vaccine-induced protection using
-derived molecules has been demonstrated, suggesting that the control of this
parasite through vaccination is possible (Nisbet et al. 2016). However what host molecules
recognise these glycoproteins are poorly understood. Recently it has been shown that galectins
have been showed to play major roles in host defence against microbial pathogens. Galectins are
a family of carbohydrate-binding molecules with characteristic domain organization and affinity
for β-galactosides mediate a variety of important cellular functions, including inflammation and
immune responses due to binding both self and non- self-glycans.
In particular, ruminants highly upregulate two specific galectins (LGALS-11 and LGALS-14
upon infection by various parasites such as  and  (Dunphy et al.
2000; Dunphy et al. 2002; Hoorens et al. 2011; Meeusen et al. 2005). LGALS-14 is secreted by
eosinophil immune cells that are critical for immunity through killing the larval stages of 
 (Balic et al. 2006; Dunphy et al. 2002; Young et al. 2009). LGALS-14 is thought to be
the homologue of human galectin-10, which is also secreted by eosinophils (Ackerman et al.
2002). Analysis of  infected sheep demonstrated release of LGALS-14 into the
gastrointestinal mucus, the interface of host and parasite interaction (Dunphy et al. 2002). In
addition, kinetic studies of LGAL-14 showed that release into the mucus occurred soon after
challenge infection, and correlated with a reduction in parasitic burden (Robinson et al. 2011).
Additional it has been shown that LGAL-14 can bind directly to another parasite 
 suggesting it can inhibit infection.
The second galectin (LGALS-11) is specifically expressed and secreted during 
infections in previously infected sheep that had developed resistance to the parasite (Dunphy et
al. 2000). Immunohistochemistry revealed that LGALS-11 was secreted by epithelial cells lining
the gastrointestinal tract, where it was localised to the nucleus and cytoplasm of cells. Analysis of
the mucosal contents lining the gastrointestinal tract also revealed secretion of LGALS-11 into
the mucus. An observation of increased mucus stickiness corresponding with the production of
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
LGALS-11 suggested that LGALS-11 might work by interacting with the mucus to impede 
 motility (Robinson et al. 2011). Recent immunofluorescent staining techniques using a
recombinant form of galectin-11 have revealed binding to the fourth larval stage and adult 
that has resulted in impaired developmentThese studies suggest a more direct or
additional role for LGALS-11 during  infections.
It appears that both LGALS-11 and LGALS-14 mediate critical immune regulatory effects and/or
mediate direct parasite stage-specific killing (Haslam et al. 1998; Preston et al. 2015b). Although
the interactions of these host galectin-parasite glycoconjugates are likely to be critical for parasite
control, the parasite glycoconjugate molecules that they recognise are unknown. For the first
time, this study describes the ligands of sheep LGALS-11 and LGALS-14 in larval and adult
stages of .


(Haecon-5 strain) was maintained in Professor Gasser’s laboratory, Melbourne
Veterinary School, The University of Melbourne and was used in this study. Mature fourth stage
larvae (L4 stage) and adults of were prepared using established protocols (Preston et
al. 2015a). Briefly, third-stage larvae (L3) were isolated from faeces from -infected
sheep. The cuticle was removed from the L3s by using sodium hypochlorite, the exsheathed L3
(xL3) worms were washed three times with 0.9% (w/v) biological saline. Approximately 2000
xL3 / ml worms were resuspended in Dulbecco’s modified Eagle Medium+GlutaMax (DMEM)
(Gibco-Invitrogen, USA) containing 10,000 IU/ml of penicillin and 10,000 µg/ml of
streptomycin (Gibco-Invitrogen, USA) and 0.5 % (v/v) fungicide (GE Healthcare, UK). Medium
containing xL3s was incubated at 37 °C with 10 % (v/v) CO2 for 7 days. Fresh DMEM was
substituted at two-day intervals and larval development was examined each day. The xL3 and L4
stages were differentiated based on distinctive morphological characteristics (see Preston et al.,
2015a). Animal experimental procedures were approved by the Monash University Animal Ethics
Committee (Ethics # SOBSA/P/2009/44). Adults of were collected from Merino
ewes (8-12 months old) which were experimentally infected with 10,000 L3s and the infected
animals were euthanised 52 days post infection by injection of pentobarbitone (Lethobarb®,
Virbac Pty Ltd, Australia). Approximately 5,000 adult worms of mixed sex were collected from
66
67
68
69
70
71
72
73
74
75
76
77
78
79
80
81
82
83
84
85
86
87
88
89
90
91
92
93
94
95
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
the abomasal content and washed five times with 0.9 % (v/v) biological saline (Baxter, Australia).
Immediately after washing, the worms were snap frozen in liquid nitrogen and stored at -80 °C
until further use.
 
Lysates were prepared using radioimmunoprecipitation assay buffer (RIPA) as previously
described with minor modifications (Maduzia et al. 2011). Briefly, 500 mg of larval or adult 
 were incubated with 100 mM β-D-galactose containing 0.9 % (v/v) biological saline
for 12 h to remove native galectins (bound to the adult parasite surface recovered from infected
sheep) and washed three times with normal saline. Larval or adult  were then
resuspended in 5 ml of ice-cold RIPA buffer [20 mM Tris-HCL pH 7.2, 100 mM NaCl, 1% (v/v)
Nonidet P-40, 0.1 % (w/v) sodium deoxycholate (DOC), 0.05 % (w/v) sodium dodecyl sulphate
(SDS), 1 % (v/v) Triton X-100, 10 mM TCEP (Tris (2-carboxyethyl) phosphine)] and lysed by
sonication (30 sec, 8 times with three min interval at 40 % amplitude). Cellular debris was
removed by centrifugation (15000 x for 20 min) at 4 °C, and any particles in the supernatant
removed by filtering through a 0.22 µm filter. Lysates were dialysed using 3 kDa molecular
weight cut-off against binding buffer [(20 mM Tris-HCl pH 7.5, 100 mM NaCl, 0.5 % (v/v)
Nonidet P-40, 0.1 % (w/v) DOC, 0.05 % (w/v) SDS, 1% (v/v) Triton X-100, 10 mM TCEP)].
SDS–PAGE
!! 
Recombinant LGALS-11 and LGALS-14 were expressed and purified as described previously
((Sakthivel et al. 2015); Fig. 1). The recombinant protein (5 mg/ml) was buffer-exchanged into
HEPES buffer (10 mM HEPES-NaOH pH 7.5, 100 mM NaCl, 10 mM TCEP) and immobilised
by coupling to N-hydroxysuccinamide (NHS)-activated sepharose (GE Healthcare, UK)
following the manufacturer’s protocol. Briefly, 4 ml of NHS-activated sepharose was washed
with 15 column-volumes of ice-cold 1 mM HCl. The washed Sepharose beads were equilibrated
with 20 ml of coupling buffer (10 mM HEPES-NaOH pH 7.5, 100 mM NaCl, 10 mM TCEP).
Following equilibration, LGALS-11 and LGALS-14 were added separately to the activated
Sepharose and allowed to couple for 5 h at 22 °C. Following the coupling reaction, the unused,
activated sites were blocked using 15 column-volumes of blocking buffer (100 mM Tris-HCl pH
8.0, 100 mM NaCl) for 3 h. Following blocking, the sepharose beads were washed alternatively
six times with 15 column-volumes of 100 mM Tris-HCL pH 8.0 and 100 mM sodium acetate pH
96
97
98
99
100
101
102
103
104
105
106
107
108
109
110
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
126
127
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
5.0 and 250 mM NaCl. The galectin affinity column was maintained in storage buffer (20 mM
Tris-HCl pH 8.0, 100 mM NaCl, 10 mM TCEP, NaAc 0.02 % (w/v)) until further use. A control
resin was also prepared without any protein ligand.
!!
Immobilised LGALS-11, -14 or control slurry (1 ml) was loaded into individual columns. Larval
and adult  lysates were diluted with 5 ml of binding buffer (20 mM Tris-HCl pH 7.5,
100 mM NaCl, 0.5 % (v/v) Nonidet P-40, 0.1 % (w/v) DOC, 0.05 % (w/v) SDS, 1% (v/v) Triton
X-100, 10 mM TCEP) and applied to the galectin affinity column and incubated for 16 h at 4°C.
Thereafter, columns were washed three times with 15 ml of RIPA buffer, the captured protein
fractions were eluted by incubating for 2 h with galactose elution buffer (250 mM β-D-Galactose
20 mM Tris-HCl pH 8.0, 100 mM NaCl, 10 mM TCEP) and the resultant supernatant was
subjected to LC-MS/MS analysis to identify the protein molecules present. The eluted protein
products were analysed by 12% SDS-PAGE stained with nitrate. The unbound fractions, column
wash and eluted proteins fractions were concentrated using sodium deoxycholate/trichloroacetic
acid precipitation method to allow the visualisation of protein products as previously described
(Arnold & Ulbrich-Hofmann 1999).
"#!$%$!&!' 
Eluted protein samples were dissolved in digestion buffer (8 M urea, 50 mM NH4HCO3, 10 mM
dithiothreitol) and incubated at 25 (C for 5 h. Following incubation, iodoacetamide (IAA) was
added to final concentration of 55 mM to alkylate thiol groups and incubated for 35 min at 20 (C
in the dark. The alkylated protein preparation was diluted with 1M urea in 25 mM ammonium
bicarbonate (pH 8.5) and sequencing-grade trypsin (Promega) was added to a final concentration
of 5 µM. The reaction was incubated for 16 h at 37 (C in the dark. The digests were acidified
with 1% (v/v) trifluoroacetic acid (TFA) and the peptides desalted on poly(styrene-
divinylbebzebe) copolymer (SDB) (Empore) StageTips as described previously (Rappsilber et al.
2007).
Trypsin-digested peptides were reconstituted in 0.1% (v/v) TFA and 2% (v/v) acetonitrile (ACN)
and then loaded onto a guard column (C18 PepMap 100 µm ID × 2 cm trapping column, Thermo-
128
129
130
131
132
133
134
135
136
137
138
139
140
141
142
143
144
145
146
147
148
149
150
151
152
153
154
155
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Fisher Scientific) at 5 µl/min and washed for 6 min before switching the guard column, in line
with the analytical column (Vydac MS C18, 3 µm, 300 Å and 75 µm ID × 25 cm). The separation
of peptides was performed at 300 nl/min using a non-linear ACN gradient of buffer A (0.1% (v/v)
formic acid, 2 % (v/v) ACN) and buffer B (0.1% (v/v) formic acid, 80 % (v/v) ACN), starting at
5% (v/v) buffer B to 55% for 120 min. Data were collected on an Orbitrap Elite (Thermo-Fisher
Scientific) in a Data-Dependent Acquisition mode using m/z 300–1500 as MS scan range, CID
MS/MS spectra and were collected for the 20 most intense ions. Dynamic exclusion parameters
were set as described previously (Nguyen et al. 2016). The Orbitrap Elite was operated in dual
analyser mode, with the Orbitrap analyser being used for MS and the linear trap being used for
MS/MS. Pull-down and LC-MS/MS analysis were performed three times on different days.
)
The MS/MS spectra obtained from the Orbitrap analyser was used to search against the Swiss-
Prot   FASTA database (downloaded on 07.31.2016, 21,201 protein entries)
together with common contaminants were used for this analysis using the Mascot search engine
(Matrix Science Ltd., London, UK) as described previously (Perkins et al. 1999). Briefly,
carbamidomethylation of cysteines was set as a fixed modification, acetylation of protein N-
termini, methionine oxidation was included as variable modifications. Precursor mass tolerance
was 10 ppm, product ions were searched at 0.5 Da tolerances, minimum peptide length defined at
6, maximum peptide length 144, and Peptide spectral matches (PSM) were validated using
Percolator based on q-values at a 1% false discovery rate (FDR). Both peptide and protein
identifications were reported at a false discovery rate (FDR) of 1%. The mass spectrometry
proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner
repository with the data set identifier PXD008435 and 10.6019/PXD008435.
 
Normalized spectral abundance factor (NSAF) scores were calculated for the identified proteins
using the Scaffold software v4.7.2 (Searle 2010). Then proteins were subjected to the significance
analysis of interactome' (SAINT) (Choi et al. 2011) to identify  protein-protein
interactions after removing all zero or missing rows. Proteins with a SAINT probability greater
than 0.9 were selected as high probability interactions. Finally, the resulting interaction network
was visualised using the Cytoscape v3.4.0 (Shannon et al. 2003).
156
157
158
159
160
161
162
163
164
165
166
167
168
169
170
171
172
173
174
175
176
177
178
179
180
181
182
183
184
185
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
   
The N- and O-linked glycosylation pattern and the signal peptides of eluted proteins were
analysed following the instructions provided in the glycosylation analysis server. Briefly, N-
glycosylation and Signal peptide was analysed using NetNGlyc 1.0 server
(http://www.cbs.dtu.dk/services/NetNGlyc/). Whereas the O-glycosylation pattern was analysed
using NetOGlyc 4.0 Server (http://www.cbs.dtu.dk/services/NetOGlyc/). The results obtained
from the N and O glycosylation servers were provided as supplementary results.
*)
H. contortus +!!
The overall experimental procedure to map interacting proteins of LGALS-11 and LGALS-14
from  is given in Fig. 1. Lysates of L4 and adult stages were assessed before loading
onto the columns containing the Sepharose immobilised LGALS-11 and LGALS-14 and are
shown in Fig. 2a. Multiple bands were observed, with both larval and adult lysates containing a
broad range of molecules of differing molecular weights. Following the application of L4 and
adult lysates to affinity columns, containing immobilised galectins, bound parasite molecules
were eluted with galactose (Fig. 2b & 2c). The eluted molecules from both affinity columns and
an control column were subjected to LC-MS/MS. Proteins that were identified in 2 of the 3
biological replicates were included for further analysis and the proteins that were bound to
control resin (S1) were removed from the analysis. Overall, 43 individual proteins were identified
and grouped based on their respective known or putative biological function(s) (Table 1). The
greatest number of proteins identified was in the adult stage of ; with 34 proteins
binding to LGALS-11 and 35 proteins binding to LGALS-14. Of those identified proteins, 26
proteins were found to bind to both LGALS-11 and LGALS-14 (Fig. 3; Table 2). In the L4 larval
stage, LGALS-11 and LGALS-14 could bind to 0 and 2 proteins respectively.
%!!
Approximately 69% of proteins in that bound specifically to LGALS-11 and/or
LGALS-14 were inferred to be involved in metabolic and regulatory processes (Table 1, Fig. 4).
Most of these proteins (~ 70 %) were likely involved in metabolic activities, such as energy
metabolism, transcription andtranslation. These proteins predominantly included regulatory
186
187
188
189
190
191
192
193
194
195
196
197
198
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
enzymes, such as peptidases, carboxyl transferases, aldo-keto reductases, deoxynucleoside
kinase, dehydrogenase, amidinotransferase and RNA polymerase. Another protein group (~9 %)
identified represented structural proteins, such as actin, myosin and collagen (Table 1, Fig. 4).
Other proteins identified had putative roles in molecular transport (e.g., lipid and amino acid
transport) or had no assigned function(s) (Fig. 4).  analysis revealed that approximately
65% of the proteins of the adult stagethat bound specifically to LGALS-11 and LGALS-14 had
one or more potential glycosylation site (Table 2). On the contrary, about 35 % of adult stages
specific proteins that bound to LGALS-11 and LGALS-14 were predicted as non-glycosylated.
Though animal lectins have a primary preference for glycoconjugates, it is believed that the
LGALS-11 and LGALS-14 might also display a glycan independent protein-protein interaction
activity similar to previously reported for galectin-1 and galectin-3 (Bawumia et al. 2003; Camby
et al. 2006; Menon et al. 2000; Paz 2001).
!!
More putative ligands (n = 43) were identified in the adult stage of  compared with
larval stages (n = 2) following galectin pull-down assays. Although the L4 stage is a histotropic
stage (in glands of the stomach) and would be expected to be in intimate contact with
inflammatory mediators, including galectins, it moults (with a change in antigenic profile) within
48-72 h into the immature adult (Meeusen et al. 2005). This would be expected to limit the
antigenic exposure of these parasite antigens to the host. Compared to the adult stage that is
relatively long-lived (6-8 weeks), allowing a sustained interaction of host molecules with parasite
antigens (Nikolaou & Gasser 2006; Veglia 1915). This interaction might be reflected in the
specific and localised binding of LGALS-11 in the larvae and the significant staining of LGALS-
11 on the surface of adult  (see Preston et al., 2015b). In addition, the L4 stage is
relatively small (750 – 850 µm long), whereas the adult stage is usually 10-30 mm long.
A protein-protein interaction network was drawn for LGALS-11 and LGALS-14 affinity purified
proteins specific to adult parasitic stage revealed that, LGALS-11 and LGALS-14 found to
interact 5 unique proteins individually. Whereas 9 proteins were found to interact with both
LGALS-11 and LGALS-14 (Fig. 5). Carboxyl transferase, Aldo keto reductase and myosin
displayed unique interaction with LGALS-11. Whereas Zinc metallopeptidase M13, Porin
domain containing protein, von Willebrand factor and mitochondrial solution substrate carrier
215
216
217
218
219
220
221
222
223
224
225
226
227
228
229
230
231
232
233
234
235
236
237
238
239
240
241
242
243
244
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
protein displayed an interaction network unique to LGALS-14. Peptidase S28, Alpha beta
hydrolase fold-1, Glutamate phenylalanine leucine valine dehydrogenase, Nematode cuticle
collagen, Lipid transport protein, Vitellinogen and von Willebrand factor domain were found to
interact both LGALS-11 and LGALS-14 (Fig. 5).

A significant number of proteins (n = 19) with enzyme activity in adults were identified, and
similar proteins have been described in other ‘omic studies, suggesting that many of these
enzymes of the protease family are conserved and evolutionarily related in nematodes (Campbell
et al. 2011; Ghedin et al. 2007; Schwarz et al. 2013). A notable protease identified in the adult
stage, is zinc metallopeptidase (M13 protease or neprilysin). Zinc metallopeptidases have been
reported as the major protein fraction of host protective glycoprotein complex H-gal-GP
( galactose containing glycoprotein). Several studies isolated zinc metallopeptidases
from crude extracts of  using lectins that have a binding preference to β-D-galactose
and, following vaccination of sheep, led to reduced worm burdens following challenge infection
(Dicker et al. 2014; Newlands GFJ 2006; Smith et al. 1999; Smith et al. 2000).
,
A number of parasite molecules were identified that interact with host galectins and are
potentially involved in manipulating the host blood function in the adult stage but not larvae of
. That the adult stage of this nematode is primary a blood feeder may explain the lack
of such molecules identified in the larvae. Blood feeding parasites are known to use several
mechanisms to suppress platelet aggregation, allowing prolonged blood feeding by retarding
blood clotting (Liu & Weller 1992). The von-Willebrant factor (VWF) domain is a well-known
protein domain reported in integrin and other extracellular proteins (Whittaker & Hynes 2002).
The binding of a C-type lectin (CLEC4M), with VWF has previously been shown to enhance the
internalisation of VWF by the host cells and alter plasma levels of VWF (Rydz et al. 2013). In
previous reports, proteins containing the VWF domain are localised in nematode intestine and
suggested to play critical roles in cell adhesion and platelet aggregation (Wohner et al. 2012). A
multimeric glycoprotein containing VWF domain was identified previously in adult 
that can suppress platelet aggregation (Crab et al. 2002). In this study, a protein containing a
VWF domain was eluted from the LGAL14 column, which might suggest that this host galectin
245
246
247
248
249
250
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
272
273
274
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
plays a role in potential modulating the ability of the parasite to suppress blood clotting. This
protein was not detected in larvae by both LGALS-11 and LGALS-14. However, the functional
significance of VWF in parasitised animals remains unknown, warranting further study.
!"!%'
The stage-specific sperm-coating protein (SCP) identified by host galectins in this study are
common to many nematode species (Cantacessi & Gasser 2012) and are suggested to play critical
roles in infection and immunomodulatory events such as neutrophil inhibition (Cantacessi et al.
2012; Gadahi et al. 2016; Hewitson et al. 2009). Transcriptomic studies of  have
identified that 54 genes containing one or more SCP-like domains are upregulated in the blood-
feeding adult, suggesting that SCP proteins have active and stage-specific involvement at the
onset of blood feeding (Wang & Kim 2003). Similar SCP domain containing proteins (Hc24 and
Hc40) were reported in excretory/secretory proteins of  .
Although there is some information for SCP domain-containing proteins in  (O’Rourke
et al. 2006; Wang & Kim 2003), their biological functions in  needs experimental
investigation.
%
Recently, host galectins have been hypothesised to interact with molecules to modulate host-
pathogen interactions in ruminants (Hoorens et al. 2011; Kemp et al. 2009; Preston et al. 2015b).
The finding that LGAL-14 is concentrated within eosinophils (an immune cell considered a major
mediator of parasite killing, including of ) suggested the possibility of a direct role
for ruminant galectins in mediating parasite-killing (Meeusen & Balic 2000; Robinson et al.
2011). The subsequent demonstration of direct binding of LGAL-11 to  and their
ability to inhibit larval development and growth ! has confirmed the roles of galectins and
ability to directly kill relatively large multicellular pathogens (Preston et al. 2015b).
The parasite surface is the key contact with the host and is often considered important source of
potential vaccine molecules. Correspondingly, 45% of the glycoproteins that the two galectins
bound were membrane proteins of the adult stage of and included vitelline, myosin
and M13 protein (neprilysin); these proteins have been previously assessed as vaccine candidates
(Knox 2011; Strube et al. 2015; Tellam et al. 2002). This evidence would indicate that other
275
276
277
278
279
280
281
282
283
284
285
286
287
288
289
290
291
292
293
294
295
296
297
298
299
300
301
302
303
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
putative glycoproteins identified here by these ruminant galectins might facilitate the
identification of new intervention targets and, thus, warrant further investigation. In conclusion,
the analysis of parasite proteins recognised by galectins that are involved in resistance to
parasites (Guo et al. 2016; Preston et al. 2015a; Preston et al. 2015b), has identified several
interesting stage-specific proteins. Exploring the possible biological roles and potential
anthelminthic activities of these proteins has significant potential to advance our understanding of
the host-parasite interplay and inform future parasite control strategies.
-+
DS also thanks Jyostna Nagpal for supporting DS in making high quality images. DS and DP
thank Fiona Tegart for supporting in animal maintenance during the experiment.
304
305
306
307
308
309
310
311
312
313
314
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
*
Ackerman SJ, Liu L, Kwatia MA, Savage MP, Leonidas DD, Swaminathan GJ, and Acharya KR.
2002. Charcot-Leyden Crystal Protein (Galectin-10) Is Not a Dual Function Galectin with
Lysophospholipase Activity but Binds a Lysophospholipase Inhibitor in a Novel
Structural Fashion."#$ 277:14859-14868.
Arnold U, and Ulbrich-Hofmann R. 1999. Quantitative Protein Precipitation from Guanidine
Hydrochloride-Containing Solutions by Sodium Deoxycholate/Trichloroacetic Acid.
%$#$ 271:197-199. https://doi.org/10.1006/abio.1999.4149
Balic A, Cunningham CP, and Meeusen ENT. 2006. Eosinophil interactions with Haemonchus
contortus larvae in the ovine gastrointestinal tract. &  $ 28:107-115.
10.1111/j.1365-3024.2006.00816.x
Bawumia S, Barboni Eminia AM, Menon Rajesh P, and Colin Hughes R. 2003. Specificity of
interactions of galectin-3 with Chrp, a cysteine- and histidine-rich cytoplasmic protein.
# 85:189-194. https://doi.org/10.1016/S0300-9084(03)00007-5
Camby I, Le Mercier M, Lefranc F, and Kiss R. 2006. Galectin-1: a small protein with major
functions.'$$ 16:137R-157R. 10.1093/glycob/cwl025
Campbell BE, Hofmann A, McCluskey A, and Gasser RB. 2011. Serine/threonine phosphatases
in socioeconomically important parasitic nematodes—Prospects as novel drug targets?
#$%! 29:28-39. http://dx.doi.org/10.1016/j.biotechadv.2010.08.008
Cantacessi C, and Gasser RB. 2012. SCP/TAPS proteins in helminths Where to from now?
(& 26:54-59. https://doi.org/10.1016/j.mcp.2011.10.001
Cantacessi C, Hofmann A, Young ND, Broder U, Hall RS, Loukas A, and Gasser RB. 2012.
Insights into SCP/TAPS proteins of liver flukes based on large-scale bioinformatic
analyses of sequence datasets.&)*+, 7:e31164. 10.1371/journal.pone.0031164
Choi H, Larsen B, Lin Z-Y, Breitkreutz A, Mellacheruvu D, Fermin D, Qin ZS, Tyers M, Gingras
A-C, and Nesvizhskii AI. 2011. SAINT: Probabilistic Scoring of Affinity Purification -
Mass Spectrometry Data.+ 8:70-73. 10.1038/nmeth.1541
315
316
317
318
319
320
321
322
323
324
325
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Crab A, Noppe W, Pelicaen C, Hoorelbeke KV, and Deckmyn H. 2002. The parasitic
hematophagous worm   inhibits human platelet aggregation and
adhesion: partial purification of a platelet inhibitor.- 87:899-
904.
Dicker AJ, Inglis NF, Manson EDT, Subhadra S, Illangopathy M, Muthusamy R, and Knox DP.
2014. Proteomic analysis of ( and  intestinal
protein extracts and subsequent efficacy testing in a vaccine trial. &)*  +
-. 8:e2909. 10.1371/journal.pntd.0002909
Dunphy JL, Balic A, Barcham GJ, Horvath AJ, Nash AD, and Meeusen ENT. 2000. Isolation and
characterization of a novel inducible mammalian galectin. "    #
$ 275:32106-32113. 10.1074/jbc.M003739200
Dunphy JL, Barcham GJ, Bischof RJ, Young AR, Nash A, and Meeusen ENT. 2002. Isolation and
characterization of a novel eosinophil-specific galectin released into the lungs in response
to allergen challenge."#$ 277:14916-14924.
Gadahi JA, Wang S, Bo G, Ehsan M, Yan R, Song X, Xu L, and Li X. 2016. Proteomic analysis
of the excretory and secretory proteins of  (HcESP) binding to goat
PBMCs   !! revealed stage-specific binding profiles. &)*  +, 11:e0159796.
10.1371/journal.pone.0159796
Ghedin E, Wang S, Spiro D, Caler E, Zhao Q, Crabtree J, Allen JE, Delcher AL, Guiliano DB,
and et al. 2007. Draft Genome of the Filarial Nematode Parasite Brugia malayi.*
317:1756.
Guo Z, Gonzalez JF, Hernandez JN, McNeilly TN, Corripio-Miyar Y, Frew D, Morrison T, Yu P,
and Li RW. 2016. Possible mechanisms of host resistance to Haemonchus contortus
infection in sheep breeds native to the Canary Islands. *  / 6:26200.
10.1038/srep26200
Haslam SM, Coles GC, Reason AJ, Morris HR, and Dell A. 1998. The novel core fucosylation of
   N-glycans is stage specific. (    #
&$ 93:143-147. http://dx.doi.org/10.1016/S0166-6851(98)00020-6
342
343
344
345
346
347
348
349
350
351
352
353
354
355
356
357
358
359
360
361
362
363
364
365
366
367
368
369
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Hewitson JP, Grainger JR, and Maizels RM. 2009. Helminth immunoregulation: The role of
parasite secreted proteins in modulating host immunity. (    #
&$ 167:1-11. http://dx.doi.org/10.1016/j.molbiopara.2009.04.008
Hoorens P, Rinaldi M, Mihi B, Dreesen L, Grit G, Meeusen E, Li RW, and Geldhof P. 2011.
Galectin-11 induction in the gastrointestinal tract of cattle following nematode and
protozoan infections. &  $ 33:669-678. 10.1111/j.1365-
3024.2011.01336.x
Kemp JM, Robinson NA, Meeusen ENT, and Piedrafita DM. 2009. The relationship between the
rapid rejection of   larvae with cells and mediators in abomasal
tissues in immune sheep.   "    &$ 39:1589-1594.
http://dx.doi.org/10.1016/j.ijpara.2009.05.015
Knox D. 2011. Proteases in blood-feeding nematodes and their potential as vaccine candidates.
%!,0(# 712.
Liu LX, and Weller PF. 1992. Intravascular filarial parasites inhibit platelet aggregation. Role of
parasite-derived prostanoids."! 89:1113-1120.
Maduzia LL, Yu E, and Zhang Y. 2011. Caenorhabditis elegans Galectins LEC-6 and LEC-10
Interact with Similar Glycoconjugates in the Intestine. "  # $
286:4371-4381.
Mavrot F, Hertzberg H, and Torgerson P. 2015. Effect of gastro-intestinal nematode infection on
sheep performance: a systematic review and meta-analysis. & 1 2 8:557.
10.1186/s13071-015-1164-z
McLeod RS. 1995. Cost of major parasites to the Australian livestock industries. "&
25. 10.1016/0020-7519(95)00071-9
Meeusen ENT, and Balic A. 2000. Do Eosinophils have a Role in the Killing of Helminth
Parasites?&$-$ 16:95-101. 10.1016/S0169-4758(99)01607-5
370
371
372
373
374
375
376
377
378
379
380
381
382
383
384
385
386
387
388
389
390
391
392
393
394
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Meeusen ENT, Balic A, and Bowles V. 2005. Cells, cytokines and other molecules associated
with rejection of gastrointestinal nematode parasites. 2$  $  
$ 108:121-125. http://dx.doi.org/10.1016/j.vetimm.2005.07.002
Menon RP, Strom M, and Hughes RC. 2000. Interaction of a novel cysteine and histidine-rich
cytoplasmic protein with galectin-3 in a carbohydrate-independent manner.,#*)
470:227-231. 10.1016/S0014-5793(00)01310-7
Newlands GFJ SP, Nisbet AJ, Redmond DL, Smith SK, Petitit D, Smith WD. 2006. Molecular
characterization of a family of metalloendopeptidases from the intestinal brush border of
Haemonchus contortus.&$ 133:357–368.
Nguyen VA, Carey LM, Giummarra L, Faou P, Cooke I, Howells DW, Tse T, Macaulay SL, Ma
H, Davis SM, Donnan GA, and Crewther SG. 2016. A Pathway Proteomic Profile of
Ischemic Stroke Survivors Reveals Innate Immune Dysfunction in Association with Mild
Symptoms of Depression A Pilot Study.     +$ 7:85.
10.3389/fneur.2016.00085
Nikolaou S, and Gasser RB. 2006. Prospects for exploring molecular developmental processes in
  .   "    &$ 36:859-868.
http://dx.doi.org/10.1016/j.ijpara.2006.04.007
Nisbet AJ, Meeusen EN, González JF, and Piedrafita DM. 2016. Chapter Eight - Immunity to
Haemonchus contortus and Vaccine Development. In: Robin BG, and Georg Von S-H,
eds. %!&$: Academic Press, 353-396.
O’Rourke D, Baban D, Demidova M, Mott R, and Hodgkin J. 2006. Genomic clusters, putative
pathogen recognition molecules, and antimicrobial genes are induced by infection of C.
elegans with M. nematophilum.'/ 16:1005-1016. 10.1101/gr.50823006
Paz AH, R.; Elad-Sfadia, G.; Ballan, E.; Kloog, Y. 2001. Galectin-1 binds oncogenic H-Ras to
mediate Rasmembrane anchorage and cell transformation. 20:7486-7493.
Perkins DN, Pappin DJC, Creasy DM, and Cottrell JS. 1999. Probability-based protein
identification by searching sequence databases using mass spectrometry data.
,),-/&/,** 20:3551-3567.
395
396
397
398
399
400
401
402
403
404
405
406
407
408
409
410
411
412
413
414
415
416
417
418
419
420
421
422
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Preston S, Dunphy J, Beddoe T, Meeusen E, and Young A. 2015a. Evaluation of the role of
galectins in parasite immunity. In: Stowell SR, and Cummings RD, eds. '3
(&. New York, NY: Springer New York, 371-395.
Preston SJM, Beddoe T, Walkden-Brown S, Meeusen E, and Piedrafita D. 2015b. Galectin-11: A
novel host mediator targeting specific stages of the gastrointestinal nematode parasite,
Haemonchus contortus.   "    &$ 45:791-796.
http://dx.doi.org/10.1016/j.ijpara.2015.06.003
Rappsilber J, Mann M, and Ishihama Y. 2007. Protocol for micro-purification, enrichment, pre-
fractionation and storage of peptides for proteomics using stage tips. +  &
2:1896-1906.
Robinson N, Pleasance J, Piedrafita D, and Meeusen EN. 2011. The kinetics of local cytokine and
galectin expression after challenge infection with the gastrointestinal nematode,
Haemonchus contortus.   "    &$ 41:487-493.
10.1016/j.ijpara.2010.11.006
Roeber F, Jex AR, and Gasser RB. 2013. Impact of gastrointestinal parasitic nematodes of sheep,
and the role of advanced molecular tools for exploring epidemiology and drug resistance -
an Australian perspective.&12 6:153-153. 10.1186/1756-3305-6-153
Rydz N, Swystun LL, Notley C, Paterson AD, Riches JJ, Sponagle K, Boonyawat B,
Montgomery RR, James PD, and Lillicrap D. 2013. The C-type lectin receptor CLEC4M
binds, internalizes, and clears von Willebrand factor and contributes to the variation in
plasma von Willebrand factor levels. # 121:5228-5237. 10.1182/blood-2012-10-
457507
Sakthivel D, Littler D, Shahine A, Troy S, Johnson M, Rossjohn J, Piedrafita D, and Beddoe T.
2015. Cloning, expression, purification and crystallographic studies of galectin-11 from
domestic sheep (!  ). %  $  *   71:993-997.
doi:10.1107/S2053230X15010195
Schwarz EM, Korhonen PK, Campbell BE, Young ND, Jex AR, and Jabbar A. 2013. The genome
and developmental transcriptome of the strongylid nematode Haemonchus contortus.
'# 14. 10.1186/gb-2013-14-8-r89
423
424
425
426
427
428
429
430
431
432
433
434
435
436
437
438
439
440
441
442
443
444
445
446
447
448
449
450
451
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Searle BC. 2010. Scaffold: A bioinformatic tool for validating MS/MS-based proteomic studies.
&/-,(* 10:1265-1269. 10.1002/pmic.200900437
Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, and
Ideker T. 2003. Cytoscape: A Software Environment for Integrated Models of
Biomolecular Interaction Networks. '  / 13:2498-2504.
10.1101/gr.1239303
Smith SK, Pettit D, Newlands GF, Redmond DL, Skuce PJ, Knox DP, and Smith WD. 1999.
Further immunization and biochemical studies with a protective antigen complex from the
microvillar membrane of the intestine of Haemonchus contortus. &  
21:187-199.
Smith WD, Smith SK, Pettit D, Newlands GF, and Skuce PJ. 2000. Relative protective properties
of three membrane glycoprotein fractions from Haemonchus contortus.&
22:63-71.
Strube C, Haake C, Sager H, Schorderet Weber S, Kaminsky R, Buschbaum S, Joekel D, Schicht
S, Kremmer E, Korrell J, Schnieder T, and von Samson-Himmelstjerna G. 2015.
Vaccination with recombinant paramyosin against the bovine lungworm Dictyocaulus
viviparus considerably reduces worm burden and larvae shedding. & 1 2
8:119. 10.1186/s13071-015-0733-5
Tellam RL, Kemp D, Riding G, Briscoe S, Smith D, Sharp P, Irving D, and Willadsen P. 2002.
Reduced oviposition of Boophilus microplus feeding on sheep vaccinated with vitellin.
2$&$ 103:141-156. http://dx.doi.org/10.1016/S0304-4017(01)00573-8
Veglia F. 1915. The anatomy and life history of the  (Rud). /
2$/.%4*% 3:349-500.
Wang J, and Kim SK. 2003. Global analysis of dauer gene expression in Caenorhabditis elegans.
.! 130:1621.
Whittaker CA, and Hynes RO. 2002. Distribution and evolution of von willebrand/integrin A
domains: widely dispersed domains with roles in cell adhesion and elsewhere.(
#$ 13:3369-3387.
452
453
454
455
456
457
458
459
460
461
462
463
464
465
466
467
468
469
470
471
472
473
474
475
476
477
478
479
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Wohner N, Kovács A, Machovich R, and Kolev K. 2012. Modulation of the von Willebrand
factor-dependent platelet adhesion through alternative proteolytic pathways.-
/ 129:e41-e46. 10.1016/j.thromres.2011.11.021
Yatsuda AP, Krijgsveld J, Cornelissen AW, Heck AJ, and de Vries E. 2003. Comprehensive
analysis of the secreted proteins of the parasite Haemonchus contortus reveals extensive
sequence variation and differential immune recognition."# 278:16941-16951.
10.1074/jbc.M212453200
Young AR, Barcham GJ, Kemp JM, Dunphy JL, Nash A, and Meeusen EN. 2009. Functional
characterization of an eosinophil-specific galectin, ovine galectin-14. '$5
" 26:423-432. 10.1007/s10719-008-9190-0
480
481
482
483
484
485
486
487
488
489
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Table 1(on next page)
Identification by mass spectroscopy of larval and adult Haemonchus contortus proteins
eluted from LGALS-11 and LGALS-14 columns
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Accession
code Gene description CV PSMs UP
Mascot
Score Groups
Log-
odds
1 2 3 4
Metabolic process
W6NE18 Peptidase S28 GN=HCOI_01497800 45.21 634 20 7058 No Yes Yes Yes 145.7
W6NFG0 Alpha beta hydrolase fold-1 GN=HCOI_00457700 58.65 504 16 6681 No Yes Yes Yes 128.68
W6NFT9 Peptidase S28 GN=HCOI_01562400 23.27 222 18 3151 No Yes No Yes 11.26
W6NJ96 Carboxyl transferase GN=HCOI_00766200 23.39 123 10 1146 No Yes No Yes 16.01
W6NLA8 Glutamate phenylalanine leucine valine dehydrogenase 42.50 102 10 957 No Yes No Yes 20.61
W6NG90 Peptidase S28 GN=HCOI_01624000 4.88 101 3 1218 No Yes No Yes 29.6
W6NC58 Aldo keto reductase GN=HCOI_00043700 39.64 83 10 789 No Yes No Yes -0.18
W6NAV8 Aldo keto reductase GN=HCOI_00043500 27.22 46 6 477 No Yes No Yes -0.18
W6NU27 Carboxyl transferase GN=HCOI_00766300 25.83 67 6 909 No Yes No Yes 4.35
W6NKM1 Succinate dehydrogenase iron-sulfur subunit,
GN=HCOI_01735500 32.61 64 8 521 No Yes No Yes 34.83
W6NF70 Deoxynucleoside kinase 35.42 42 6 242 No No No Yes -0.18
U6NNG6 Ribosomal protein L7 L12 GN=HCOI_00340500 11.41 30 1 783 No Yes No Yes -0.18
W6NA79 Zinc metallopeptidase M13 GN=HCOI_01030800 37.90 28 3 330 No No No Yes 4.35
W6ND82 von Willebrand factor GN=HCOI_01354500 13.82 26 3 371 No No No Yes 16.01
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
W6NMI7 Proteinase inhibitor I33 GN=HCOI_02015200 18.58 22 5 162 No No No Yes -0.18
W6NKG5 Ribosomal protein L15 GN=HCOI_01717500 5.88 4 1 26 No No No Yes 5.67
W6NEW9 Amidinotransferase GN=HCOI_01556200 23.35 24 4 109 No Yes No No -0.18
W6NI22 Adenylosuccinate lysase GN=HCOI_00436500 6.41 14 3 141 No Yes No No 12.23
W6NF84 Short-chain dehydrogenase reductase GN=HCOI_01467500 3.07 11 1 137 No Yes No Yes 1.71
W6ND43 Acetyltransferase component of pyruvate dehydrogenase
complex GN=HCOI_00576100 1.22 8 1 51 No Yes No No 1.68
Regulation of biological processes
W6NC73 ATPase GN=HCOI_02138200 22.64 23 2 418 No Yes No Yes -0.18
W6NJ12 Filament domain containing protein GN=HCOI_02013700 14.09 20 3 291 No Yes No Yes -0.18
W6NM02 Fumarate lyase GN=HCOI_01914600 4.73 8 1 62 No Yes No No 4.61
W6NGK5 CRE-DHS-15 protein GN=HCOI_00341400 29.31 6 1 210 No No No Yes -0.18
W6NI80 Acyl-CoA-binding protein GN=HCOI_01539100 20.69 6 1 205 No Yes No Yes 6.69
W6NCC1 NIPSNAP GN=HCOI_01963300 7.63 6 1 75 No Yes No No -0.18
W6NWY9 Porin domain containing protein GN=HCOI_01573900 43.97 82 9 1149 No Yes No Yes 4.35
W6NAW4 FG-GAP and Integrin alpha-2/Integrin alpha chain
GN=HCOI_01903100 5.90 33 5 340 No Yes No Yes -0.18
W6NAL4 Heat shock protein 70 GN=HCOI_00589700 7.24 9 1 82 No Yes No No 7.23
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Transport
W6NVQ1 Lipid transport protein and Vitellinogen and von Willebrand
factor domain GN=HCOI_01683400 23.18 209 24 1465 No Yes No Yes 9.62
W6N9I2 Lipid transport protein GN=HCOI_00072100 35.97 139 13 1313 No Yes No Yes 51.51
W6NQZ5 Mitochondrial substrate solute carrier GN=HCOI_01092000 23.10 53 8 334 No No No Yes 6.2
Cytoskeleton
W6NAH7 Nematode cuticle collagen and Collagen triple helix repeat 5.60 85 2 1399 No Yes No Yes 4.35
W6NHH0 Annexin GN=HCOI_01003500 14.01 22 3 135 No Yes No No -0.18
W6NE41 Myosin tail GN=HCOI_01216000 3.99 17 3 59 No Yes No No 13.42
W6NF56 Myosin tail GN=HCOI_01461300 34.48 51 8 675 No Yes No Yes 6.2
Host-parasite interaction
W6NGA7 SCP extracellular domain GN=HCOI_01577700 14.95 4 2 59 No Yes No Yes -0.18
Unknown
W6NAN9 CBN-MLC-3 protein GN=HCOI_01274700 49.67 63 7 465 No Yes No Yes 2.32
W6NX42 Uncharacterized protein GN=HCOI_01051700 25.56 38 3 607 No No No Yes 19.09
W6NFI4 Protein C15F1.2 GN=HCOI_01126300 34.87 20 4 97 No Yes No Yes -0.18
W6NB42 Protein C23H5.8, isoform-c GN=HCOI_00648100 14.43 16 2 170 No Yes No Yes 10.80
W6NPK3 Uncharacterized protein GN=HCOI_00260500 5.20 10 1 45 No No No Yes -0.18
W6NUX4 Uncharacterized protein GN=HCOI_01608400 4.04 12 1 43 No Yes No Yes 4.04
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Abbreviations:
GN = Gene name; CV = Coverage; PSMs = Peptide spectrum matches; UP = Unique peptides;
Log-odds 0 = 50% chance, Positive values = More than 50%, Negative values = Less than 50%
Groups:
1 = LGALS-11 bound protein from L4 larval stage of H. contortus
2 = LGALS-11 bound protein from adult stage of H. contortus
3 = LGALS-14 bound protein from L4 larval stage of H. contortus
4 = LGALS-14 bound protein from adult stage of H. contortus
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Table 2(on next page)
Host galectins LGALS-11 and LGALS-14 ligands common to adult stages of H. contortus
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Accession Gene Description
Signal
peptide
Potential
Glycosylation
         
 !"
 #
$ %&'  (  ')&  *&+
 ,""
# #
$-!         
, 
# #
./
0.&&1
&&(  *      
2 
 
! 0      "
 .
# 
3! (4)&*"  
0% &5  ')&&  &5  1&
')
 #
!         
 
# #
%" 5&&&  
, %&65 2"  #
#!       
,"2!
 #
7" (4)&*"2  #
89
5  ')  :5(;5<  +
5&*5  5(5=  '&
"2,,
 #
%! >+90+2" "  #
$, 9) 2  
%. %&65 2,  #
% $+%&'+&' # #
7 ?(&    0"  0
2 ,
 #
! %*  
"2 %-  #
3 $&      
2"
 
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Abbreviations:
GN = Gene name
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Figure 1(on next page)
Schematic flow of pull-down experiment to identify the interactome.
Lysates of Haemonchus contortus (larval or adult worms) containing glycoproteins were
isolated using immobilised recombinant LGALS-11 and LGALS-14 columns and eluted using a
high concentration of β-D-galactose. The glycoproteins of larval and adult stages that
interact with host galectins were analysed by LC-MS/MS. The spectra obtained from the LC-
MS/MS were analysed using the Mascot (Perkins et al. 1999) and the NCBI protein database.
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Figure 2(on next page)
SDS-PAGE analysis of galectin bound proteins.
(A) Protein profiling of larval and adult stages of Haemonchus contortus. (M)
Molecular weight markers; (Lane 1 & 2) lysates prepared from L4 stage; (Lane 3) lysates
prepared from adult stage. (B) Protein profile of adult stage parasite bound to LGALS-
11 and LGALS-14 and (C) larval stage parasite bound to LGALS-11 and LGALS-14.
M) Molecular weight markers; (Lane 1) Total parasite lysate, (Lane 2 & 3) Unbound protein
fractions of LGALS-11 and LGALS-14 column, (Lane 4 & 5) Column wash of LGALS-11 and
LGALS-14 column and (Lane 6 & 7) eluted protein of LGALS-11 and LGALS-14 column.
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Figure 3(on next page)
Venn diagram of parasite proteins bound by host galectins.
Venn diagram showing the distribution of proteins of the larval (A) and adult (B) stages of
Haemonchus contortus. In larval and adult stages, 0 and 26 proteins were bound by both the
galectins respectively.
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Figure 4(on next page)
Categorisation of proteins in the adult stage of Haemonchus contortus that interacted
with host galectins.
The profiles were categorised based on biological process of LGALS-11-bound proteins (A)
and LGALS-14 bound-proteins (B) and cellular location of LGALS-11-bound proteins (C) and
LGALS-14-bound proteins (D).
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
Figure 5(on next page)
Protein-protein interaction network of adult stage of Haemonchus contortus with host
galectins.
Protein interactions was determined using the software (SAINT) (Choi et al. 2011) and
resulting interaction network was visualised using the Cytoscape v3.4.0.
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
W6NA79
W6ND82
W6NQZ5
14_Adult
W6NWY9
W6NX42
11_Adult
W6NU27
W6NAV8
W6NC58
W6NF56
W6NJ96
W6NG90
W6NFT9
W6NLA8
W6NFG0
W6NAH7
W6NE18
W6N9I2
W6NVQ1
PeerJ Preprints | https://doi.org/10.7287/peerj.preprints.3473v1 | CC BY 4.0 Open Access | rec: 19 Dec 2017, publ: 19 Dec 2017
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Galectins are an evolutionarily conserved family of proteins that translate glycan recognition into cellular effects. Galectin-11 is a unique member of the galectin family that is only expressed in ruminants such as sheep, goat and cattle and that plays a critical role in several important biological processes, such as reproduction and parasite-mediated innate immune responses. Currently, these two areas are of major importance for the sustainability of ruminant livestock production. Despite the emerging biological significance of galectin-11, no structural information is available. It is expected that structural studies will unravel the functional mechanisms of galectin-11 activity. Here, the expression, purification and crystallization of the ruminant-specific galectin-11 from domestic sheep and the collection of X-ray data to 2.0 Å resolution are reported.
Article
Full-text available
Galectin-11 and galectin-14 are ruminant galectins involved in parasitic infections. Although their roles in parasite immunity are still being elucidated, its appears that their functions are parasite specific. In gastrointestinal infections with the nematode Haemonchus contortus, both galectin-11 and galectin-14 appear to be protective. However, in a chronic infection of liver fluke, Fasciola hepatica, these galectins may aid parasite survival. This chapter discusses the methods designed to study parasitic infections in sheep, which have provided us with insight into the functions of galectin-11 and galectin-14 during host-parasite interactions. These methods include parasite cultivation and infection, galectin staining of host and parasite tissue, surface staining of parasites with recombinant galectins and in vitro assays to monitor the effect of galectins on larval development.
Article
Full-text available
The barber's pole worm, Haemonchus contortus, is one of the most economically important parasites of small ruminants worldwide. Although this parasite can be controlled using anthelmintic drugs, resistance against most drugs in common use has become a widespread problem. We provide a draft of the genome and the transcriptomes of all key developmental stages of H. contortus to support biological and biotechnological research areas of this and related parasites. The draft genome of H. contortus is 320 Mb in size and encodes 23,610 protein-coding genes. On a fundamental level, we elucidate transcriptional alterations taking place throughout the life cycle, characterize the parasite's gene silencing machinery, and explore molecules involved in development, reproduction, host-parasite interactions, immunity and disease. The secretome of H. contortus is particularly rich in peptidases linked to blood feeding activity and interactions with host tissues, and a diverse array of molecules is involved in complex immune responses. On an applied level, we predict drug targets and identify vaccine molecules. The draft genome and developmental transcriptome of H. contortus provide a major resource to the scientific community for a wide range of genomic, genetic, proteomic, metabolomic, evolutionary, biological, ecological and epidemiological investigations, and a solid foundation for biotechnological outcomes, including new anthelmintics, vaccines and diagnostic tests. This first draft genome of any strongylid nematode paves the way for a rapid acceleration in our understanding of a wide range of socioeconomically important parasites of one of the largest nematode orders.
Article
Full-text available
Genetic variation in or near the C-type lectin domain family 4 member M (CLEC4M) has been associated with plasma levels of von Willebrand factor (VWF) in healthy individuals. CLEC4M is a lectin receptor with a polymorphic extracellular neck region possessing a variable number of tandem repeats (VNTR). A total of 491 participants (318 patients with type 1 von Willebrand disease [VWD] and 173 unaffected family members) were genotyped for the CLEC4M VNTR polymorphism. Family-based association analysis on kindreds with type 1 VWD demonstrated an excess transmission of VNTR 6 to unaffected individuals (P = .0096) and an association of this allele with increased VWF:RCo (P = .029). CLEC4M-Fc bound to VWF. Immunofluorescence and enzyme-linked immunosorbent assay demonstrated that HEK 293 cells transfected with CLEC4M bound and internalized VWF. Cells expressing 4 or 9 copies of the CLEC4M neck region VNTR showed reduced interaction with VWF relative to CLEC4M with 7 VNTR (CLEC4M 4%-60% reduction, P < .001; CLEC4M 9%-45% reduction, P = .006). Mice expressing CLEC4M after hydrodynamic liver transfer have a 46% decrease in plasma levels of VWF (P = .0094). CLEC4M binds to and internalizes VWF, and polymorphisms in the CLEC4M gene contribute to variable plasma levels of VWF.
Article
Full-text available
SCP/TAPS proteins of parasitic helminths have been proposed to play key roles in fundamental biological processes linked to the invasion of and establishment in their mammalian host animals, such as the transition from free-living to parasitic stages and the modulation of host immune responses. Despite the evidence that SCP/TAPS proteins of parasitic nematodes are involved in host-parasite interactions, there is a paucity of information on this protein family for parasitic trematodes of socio-economic importance. We conducted the first large-scale study of SCP/TAPS proteins of a range of parasitic trematodes of both human and veterinary importance (including the liver flukes Clonorchis sinensis, Opisthorchis viverrini, Fasciola hepatica and F. gigantica as well as the blood flukes Schistosoma mansoni, S. japonicum and S. haematobium). We mined all current transcriptomic and/or genomic sequence datasets from public databases, predicted secondary structures of full-length protein sequences, undertook systematic phylogenetic analyses and investigated the differential transcription of SCP/TAPS genes in O. viverrini and F. hepatica, with an emphasis on those that are up-regulated in the developmental stages infecting the mammalian host. This work, which sheds new light on SCP/TAPS proteins, guides future structural and functional explorations of key SCP/TAPS molecules associated with diseases caused by flatworms. Future fundamental investigations of these molecules in parasites and the integration of structural and functional data could lead to new approaches for the control of parasitic diseases.
Article
Several algorithms have been described in the literature for protein identification by searching a sequence database using mass spectrometry data. In some approaches, the experimental data are peptide molecular weights from the digestion of a protein by an enzyme. Other approaches use tandem mass spectrometry (MS/MS) data from one or more peptides. Still others combine mass data with amino acid sequence data. We present results from a new computer program, Mascot, which integrates all three types of search. The scoring algorithm is probability based, which has a number of advantages: (i) A simple rule can be used to judge whether a result is significant or not. This is particularly useful in guarding against false positives. (ii) Scores can be com pared with those from other types of search, such as sequence homology. (iii) Search parameters can be readily optimised by iteration. The strengths and limitations of probability-based scoring are discussed, particularly in the context of high throughput, fully automated protein identification.
Article
Sheep are capable of developing protective immunity to Haemonchus contortus through repeated exposure to this parasite, although this immune protection is the result of a complex interaction among age, gender, physiological status, pregnancy, lactation, nutrition and innate and adaptive immunity in the host animal. There are multiple effectors of the protective immune response, which differ depending on the developmental stage of the parasite being targeted, and our understanding of the effector mechanisms has developed considerably in the 2000s. The rational design of vaccines based on 'natural' or 'exposed' antigens depends on an understanding of this exposure-induced immunity. However, the most effective current vaccines rely on protection via the induction of high circulating antibody levels to 'hidden' gut antigens of H. contortus. The success of this latter strategy has resulted in the launch of a vaccine, which is based on extracts of the parasite's gut, to aid in the control of Haemonchus in Australia. The development of recombinant subunit vaccines based on the components of the successful native vaccine has not yet been achieved and most of the recent successes with recombinant subunit vaccines have focussed on antigens unrelated to the gut antigens. The future integration of an understanding of the immunobiology of this parasite with advances in antigen identification, expression (or synthesis) and presentation is likely to be pivotal to the further development of these recombinant subunit vaccines. Recent progress in each of the components underpinning this integrated approach is summarized in this review.
Article
Galectin-11 is released from epithelial cells of the gastrointestinal tract, specifically following infection with gastrointestinal parasites including the highly pathogenic nematode, Haemonchus contortus. The function(s) of galectin-11 are currently unknown but seem to be associated with the development of immunity by the host. The aim of the present study was to examine the interaction of galectin-11 with the different parasitic life cycle stages of H. contortus and determine any effects on parasite development. The results of this study showed that galectin-11 binds to the surface of the L4 and adult stages of the parasite but not to the exsheathed L3 stage. In addition, at a lower concentration, binding to the L4 was specifically localised to the pharynx region. Subsequent in vitro assays demonstrated significant inhibition of larval growth and development in the presence of recombinant galectin-11. These results indicate, to our knowledge for the first time, a functional role for galectin-11 in gastrointestinal nematode infection of ruminants and a mechanism of action of galectin-11, targeting the development and growth of the L4 and possibly the adult parasite stage. Copyright © 2015. Published by Elsevier Ltd.