• This study demonstrates a novel mechanistic role for a specific N-linked glycan in regulating PAR1 proteolysis.

  • We provide the first description of an APC variant with enhanced therapeutic cytoprotective activity in vivo.

Activated protein C (APC) is an anticoagulant protease that initiates cell signaling via protease-activated receptor 1 (PAR1) to regulate vascular integrity and inflammatory response. In this study, a recombinant APC variant (APCN329Q) mimicking the naturally occurring APC-β plasma glycoform was found to exhibit superior PAR1 proteolysis at a cleavage site that selectively mediates cytoprotective signaling. APCN329Q also enhanced integrin αMβ2-dependent PAR1 proteolysis to exert significantly improved antiinflammatory activity on macrophages compared with wild-type APC. Recent therapeutic applications of recombinant APC in ischemic stroke models have used APC variants with limited anticoagulant activity to negate potential bleeding side effects. Using a mouse model of ischemic stroke and late t-PA intervention, the neuroprotective activity of a murine APC variant with limited anticoagulant activity (mAPCPS) was compared with an identical APC variant except for the absence of glycosylation at the APC-β sequon (mAPCPS/N329Q). Remarkably, mAPCPS/N329Q limited cerebral ischemic injury and reduced brain lesion volume significantly more effectively than mAPCPS. Collectively, this study reveals the importance of APC glycosylation in controlling the efficacy of PAR1 proteolysis by APC and demonstrates the potential of novel APC variants with superior cytoprotective signaling function as enhanced therapeutic agents for the treatment of ischemic stroke.

Activated protein C (APC) plays multiple regulatory roles that cumulatively serve to limit thrombus development1  and attenuate inflammation.2  APC engagement with the endothelial cell protein C receptor (EPCR) facilitates proteolysis-dependent activation of protease-activated receptor 1 (PAR1),3  which protects the endothelial cell barrier from disruptive agents,4  inhibits apoptosis,5,6  and limits cytokine expression from cells exposed to proinflammatory stimuli.6,7 

Recombinant APC signaling attenuates neurovascular sequelae associated with experimental murine ischemic stroke8-10  and limits neurotoxicity caused by subsequent thrombolytic tissue plasminogen activator (t-PA) administration.11,12  Recombinant APC therefore represents a promising neuroprotective drug candidate to treat ischemic stroke, and a recombinant nonanticoagulant APC variant is currently being evaluated as an adjunctive therapy to minimize the adverse effects of t-PA administration.13,14 

A recombinant APC variant (APCN329Q) that mimics the glycosylation pattern of the endogenous plasma APC-β glycoform15  exhibits significantly enhanced PAR1-dependent cytoprotective activity on endothelial cells compared with wild-type APC.16  In this study, we sought to determine the molecular basis for superior APC-β cytoprotective signaling and establish whether ablation of the APC-β glycosylation sequon to accelerate cytoprotective signaling represents a novel means by which to increase the therapeutic efficacy of APC in an ischemic stroke model.

Materials

A complete description of the reagents used in this study is included in the supplemental Material, found on the Blood Web site.

Preparation of recombinant APC

Recombinant human and murine wild-type protein C and variants were generated, expressed, and purified largely as previously described.16,17  A detailed description is provided in the supplemental Material.

Detection of PAR1 proteolysis on HEK 293T cells

PAR1 reporter–transfected 293T cells were incubated with APC in serum-free Dulbecco’s modified Eagle medium supplemented with 3 mM CaCl2 and 0.6 mM MgCl2 for 3 hours, after which the supernatant was removed and alkaline phosphatase (AP) activity measured using an AP substrate (QUANTI-Blue, Invivogen) at 650 nm.

APC inhibition of cytokine secretion from LPS-stimulated macrophages

Analysis of APC inhibition of cytokine secretion from RAW264.7 cells was performed as previously described.18  Tumor necrosis factor-alpha (TNFα) and IL-6 were measured by enyme-linked immunosorbent assay (ELISA) (R&D Systems).

Mouse model of ischemic stroke

Ischemic stroke in mice was induced as previously described19  and is further detailed in the supplemental Material. Briefly, 200 µL of 10 mg/kg t-PA (10% bolus, 90% perfusion during 20 minutes) with or without 0.2 mg/kg mAPCPS or mAPCPS/N329Q (50% bolus, 50% perfusion during 20 minutes) were given intravenously 3 hours after vessel occlusion. After 24 hours, mice were killed and their brains removed and frozen in isopentane. 20 μm cryostat-cut coronal brain sections were stained with thionine and analyzed (ImageJ 1.48V). For volume analysis, one section of every 10 was stained covering the entire lesion, and lesion (unstained) areas were measured.

We hypothesized that enhanced cytoprotective PAR1 signaling by APC-β was a consequence of accelerated EPCR-dependent PAR1 proteolysis. To test this, 293T cells cotransfected with EPCR and PAR1 linked to an N-terminal alkaline phosphatase (AP) cleavage reporter20  were treated with recombinant APC-β (APCN329Q) or N-glycosidase PNGase F–treated APC (APCPNG), in which all N-linked glycans were excised.16  Interestingly, both APCPNG and APCN329Q demonstrated up to a fourfold increased PAR1 proteolysis when compared with wild-type APC (Figure 1A) or other APC glycoform variants (data not shown).

Figure 1

APC glycosylation at Asn-329 specifically restricts PAR1 proteolysis and signaling activity. 293T cells transfected with EPCR and either (A) PAR1-AP, (B) PAR1R41Q-AP, (C) PAR1R46Q-AP were treated with APC (black circles), APCPNG (red squares) or APCN329Q (green triangles) (all 6.25-50 nM) for 3 hours, before AP activity was measured using an AP substrate. An extended period of incubation with the AP substrate was required to observe any AP activity in cell supernatants from APC-treated EPCR/PAR1R46Q-AP–transfected 293T cells. Murine macrophages (RAW264.7) were incubated with APC (black bars) or APCPNG (red bars) (5-20 nM) for 3 hours before stimulation with LPS (50 ng/mL) for 18 hours. (D) TNFα and (E) IL-6 secretion were measured by ELISA. Similarly, APC (black bars) and APCN329Q (yellow bars; 10-20 nM) were incubated with RAW264.7 macrophages for 3 hours before LPS (50 ng/mL) incubation for 18 hours. The resultant supernatants were assessed for the presence of (F) TNFα and (G) IL-6 as before. 293T-transfected cells with (H) PAR1-AP alone (black), EPCR/PAR1-AP (red), or αMβ2/PAR1-AP (green) were treated with thrombin (5 nM), wild-type APC, or APCN329Q (both 50 nM) for 3 hours before AP activity was assessed. All experiments were performed at least in triplicate and results are presented as the mean ± standard error of the mean; *P < .05, **P < .01 by Student t test.

Figure 1

APC glycosylation at Asn-329 specifically restricts PAR1 proteolysis and signaling activity. 293T cells transfected with EPCR and either (A) PAR1-AP, (B) PAR1R41Q-AP, (C) PAR1R46Q-AP were treated with APC (black circles), APCPNG (red squares) or APCN329Q (green triangles) (all 6.25-50 nM) for 3 hours, before AP activity was measured using an AP substrate. An extended period of incubation with the AP substrate was required to observe any AP activity in cell supernatants from APC-treated EPCR/PAR1R46Q-AP–transfected 293T cells. Murine macrophages (RAW264.7) were incubated with APC (black bars) or APCPNG (red bars) (5-20 nM) for 3 hours before stimulation with LPS (50 ng/mL) for 18 hours. (D) TNFα and (E) IL-6 secretion were measured by ELISA. Similarly, APC (black bars) and APCN329Q (yellow bars; 10-20 nM) were incubated with RAW264.7 macrophages for 3 hours before LPS (50 ng/mL) incubation for 18 hours. The resultant supernatants were assessed for the presence of (F) TNFα and (G) IL-6 as before. 293T-transfected cells with (H) PAR1-AP alone (black), EPCR/PAR1-AP (red), or αMβ2/PAR1-AP (green) were treated with thrombin (5 nM), wild-type APC, or APCN329Q (both 50 nM) for 3 hours before AP activity was assessed. All experiments were performed at least in triplicate and results are presented as the mean ± standard error of the mean; *P < .05, **P < .01 by Student t test.

Close modal

Generation of unique tethered ligands by APC or thrombin, caused by distinct cleavage events at Arg-46 or Arg-41, respectively, on PAR1, has been proposed as a potential mechanism to explain the disparate outcomes of PAR1 signaling initiated by each protease.21,22  To determine whether the enhanced cytoprotective activity exhibited by APCN329Q was a consequence of enhanced PAR1 proteolysis at the noncanonical Arg-46 cleavage site, 293T cells were cotransfected with EPCR and either PAR1R41Q-AP or PAR1R46Q-AP reporter variants (Figure 1B-C). Notably, cleavage at the Arg-46 site on PAR1 was especially sensitive to the loss of N-linked glycosylation after PNGase F treatment, and particularly loss of glycosylation at the APC-β sequon (Figure 1B). In contrast, Arg-41 proteolysis was limited in the presence of APC and largely insensitive to glycosylation status (Figure 1C). The increased rate of Arg-46 proteolysis on PAR1 by APCPNG and APCN329Q provides a novel molecular explanation for the enhanced cytoprotective activity of APCN329Q on endothelial cells and supports the emerging concept of biased PAR1 agonism to achieve downstream cytoprotective signaling.

αMβ2 can replace EPCR as a coreceptor for PAR1-dependent APC antiinflammatory signaling on macrophages.23  To determine whether APC glycosylation also negatively regulates αMβ2-dependent PAR1 signaling, we tested the ability of APCPNG and APCN329Q to attenuate proinflammatory cytokine release from lipopolysaccharide (LPS)-stimulated macrophages. APCPNG and APCN329Q reduced TNFα and IL-6 release from LPS-treated macrophages significantly more effectively than wild-type APC (Figure 1D-G). 293T cells coexpressing recombinant human αMβ2 and PAR1-AP were used to determine whether enhanced antiinflammatory activity was mediated by increased αMβ2-dependent PAR1 proteolysis by APCN329Q. Interestingly, the αM subunit mediated all coreceptor activity for αMβ2-PAR1 proteolysis by APC, and enabled APC cleavage of PAR1 at both canonical (Arg-41) and noncanonical (Arg-46) sites (supplemental Figure 1). Both APCPNG and APCN329Q significantly enhanced αMβ2-dependent PAR1 proteolysis (Figure 1H), indicating that N-linked glycans at Asn-329 regulate cytoprotective signaling by APC by impairing PAR1 proteolysis, irrespective of the APC coreceptor used.

Thrombolysis with t-PA is the only Food and Drug Administration–approved thrombolytic treatment of ischemic stroke, but can only be safely administered within 3 hours of symptom onset because of significant neurotoxicity.24  Recombinant APC variants with severely attenuated anticoagulant activity reduce t-PA–associated toxicity in murine models of ischemic stroke in a PAR1-dependent manner.11,12  We hypothesized that a recombinant nonanticoagulant APC that consists solely in the β-glycoform would be more efficacious in reducing t-PA neurotoxicity than existing APC variants in a murine model of ischemic stroke with late t-PA intervention. Similar to its human equivalent, in vitro analysis indicated that loss of N-linked glycan attachment at Asn-329 in murine APC (mAPCN329Q) enhanced PAR1 proteolytic and antiinflammatory activity (Figure 2A-C). To determine whether the therapeutic activity of an APC variant with reduced anticoagulant activity could be enhanced by selective de-glycosylation, a signaling-competent mAPC variant (mAPC-D36A/L38D/A39V; mAPCPS) with severely impaired anticoagulant activity in murine plasma25  was prepared. The therapeutic efficacy of this variant was compared with an identical mAPC variant prepared on an APC-β background (mAPCPS/N329Q) (Figure 2D-E). As anticipated, sham-operated mice did not develop cerebral injury (supplemental Figure 2). Like other mAPC variants with reduced anticoagulant activity,12  mAPCPS retained the ability to reduce t-PA–induced cerebral injury 24 hours after stroke and t-PA administration, albeit modestly in this model (Figure 2F-G). mAPCPS/N329Q, however, significantly reduced brain lesion size compared with when t-PA was administered alone after the ischemic event (P = .006; Figure 2F-G). Remarkably, mAPCPS/N329Q was also significantly superior in preventing brain lesions compared with mAPCPS (P = .004; Figure 2F-G), despite their structures only differing by the presence or absence of a specific N-linked glycan.

Figure 2

APC-β exhibits superior inhibition of cerebral injury in murine ischemic stroke. (A) 293T cells transfected with EPCR and PAR1-AP were incubated with mAPC or mAPCN329Q (6.25-50 nM) for 3 hours. AP activity was assessed using QUANTI Blue AP substrate, as before. RAW264.7 macrophages were incubated with mAPC (black bars) or mAPCN329Q (red bars) (12.5-50 nM) for 3 hours before stimulation with LPS (50 ng/mL) for 18 hours. Secretion of (B) TNFα and (C) IL-6 was measured by ELISA. (D) Schematic description of the ischemic stroke model and study protocol. (E) Sodium dodecyl sulfate polyacrylamide gel electrophoresis analysis under reducing conditions of purified recombinant mAPCPS and mAPCPS/N329Q. Proteins were stained with Coomassie Brilliant Blue. The heavy chain of mAPCPS migrates as a diffuse band composed of the APC-α heavy chain and APC-β glycoform heavy chain fractions. The heavy chain of mAPCPS/N329Q is composed solely of the APC-β fraction. (F) Brain infarct volumes post-stroke and t-PA administration were calculated in each mouse by staining 20-μm coronal brain sections with thionine, which is unable to stain lesion areas. For volume analysis, 1 section of every 10 covering the entire lesion was analyzed. Individual values and the median of each group are represented; n = 9-15; *P = .006, **P = .004. (G) Images corresponding to 2 thionine-stained sections of representative mice from each group are shown. Lesion areas are unstained, whereas remaining healthy tissue is stained purple.

Figure 2

APC-β exhibits superior inhibition of cerebral injury in murine ischemic stroke. (A) 293T cells transfected with EPCR and PAR1-AP were incubated with mAPC or mAPCN329Q (6.25-50 nM) for 3 hours. AP activity was assessed using QUANTI Blue AP substrate, as before. RAW264.7 macrophages were incubated with mAPC (black bars) or mAPCN329Q (red bars) (12.5-50 nM) for 3 hours before stimulation with LPS (50 ng/mL) for 18 hours. Secretion of (B) TNFα and (C) IL-6 was measured by ELISA. (D) Schematic description of the ischemic stroke model and study protocol. (E) Sodium dodecyl sulfate polyacrylamide gel electrophoresis analysis under reducing conditions of purified recombinant mAPCPS and mAPCPS/N329Q. Proteins were stained with Coomassie Brilliant Blue. The heavy chain of mAPCPS migrates as a diffuse band composed of the APC-α heavy chain and APC-β glycoform heavy chain fractions. The heavy chain of mAPCPS/N329Q is composed solely of the APC-β fraction. (F) Brain infarct volumes post-stroke and t-PA administration were calculated in each mouse by staining 20-μm coronal brain sections with thionine, which is unable to stain lesion areas. For volume analysis, 1 section of every 10 covering the entire lesion was analyzed. Individual values and the median of each group are represented; n = 9-15; *P = .006, **P = .004. (G) Images corresponding to 2 thionine-stained sections of representative mice from each group are shown. Lesion areas are unstained, whereas remaining healthy tissue is stained purple.

Close modal

This study shows that effective cytoprotective PAR1 signaling by APC is determined by specific N-linked glycans absent in certain naturally occurring APC glycoforms, revealing new mechanistic insights as to how PAR1 proteolysis by APC is regulated. Moreover, APC-β sequon disruption boosts recombinant nonanticoagulant APC amelioration of cerebral ischemic injury after late t-PA treatment in ischemic stroke. Although further studies are required to investigate which of the multiple PAR1-dependent neuroprotective functions mediated by APC in vivo13  are particularly enhanced by APCPS/N329Q in this setting, recombinant APC variants capable of hyperactive cytoprotective signaling represent a novel approach to boost the efficacy of APC-based therapeutic strategies without increasing dosage or compromising safety.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors acknowledge Eva Molina and Maider Esparza for their excellent technical assistance.

This study was funded by a Science Foundation Ireland Starting Investigator Research Grant (09/SIRG/B1590) (R.J.S.P.), the National Children’s Research Centre (R.J.S.P.), a European Research Area Network (ERANET)-Neuron grant (PRI-PIMNEU-2011-1334), and the Instituto de Salud Carlos III (PI10/01432, PI13/00072, PI11/1458, RECAVA RD06/0014/0008).

Contribution: E.M.G., M.G.D., A.S., L.M.Q., C.D., and S.E.R., performed experiments and analyzed data; P.T.W., J.O., J.H., O.P.S., and J.S.O’D. analyzed data and planned experiments; R.M. and R.J.S.P. analyzed data, planned experiments, and conceived the study; and all authors contributed to the preparation of the manuscript.

Conflict-of-interest disclosure: R.J.S.P. has received honoraria from Octapharma and research funding from Bayer Healthcare Pharmaceuticals.

Correspondence: Roger J. S. Preston, School of Medicine, Trinity College Dublin, United Kingdom; e-mail: prestonr@tcd.ie; and Ramón Montes, Center for Applied Medical Research, University of Navarra, Pamplona, Spain; e-mail: montesdiazpamplona@outlook.com.

1
Hanson
 
SR
Griffin
 
JH
Harker
 
LA
Kelly
 
AB
Esmon
 
CT
Gruber
 
A
Antithrombotic effects of thrombin-induced activation of endogenous protein C in primates.
J Clin Invest
1993
, vol. 
92
 
4
(pg. 
2003
-
2012
)
2
Taylor
 
FB
Chang
 
A
Esmon
 
CT
D’Angelo
 
A
Vigano-D’Angelo
 
S
Blick
 
KE
Protein C prevents the coagulopathic and lethal effects of Escherichia coli infusion in the baboon.
J Clin Invest
1987
, vol. 
79
 
3
(pg. 
918
-
925
)
3
Riewald
 
M
Petrovan
 
RJ
Donner
 
A
Mueller
 
BM
Ruf
 
W
Activation of endothelial cell protease activated receptor 1 by the protein C pathway.
Science
2002
, vol. 
296
 
5574
(pg. 
1880
-
1882
)
4
Feistritzer
 
C
Riewald
 
M
Endothelial barrier protection by activated protein C through PAR1-dependent sphingosine 1-phosphate receptor-1 crossactivation.
Blood
2005
, vol. 
105
 
8
(pg. 
3178
-
3184
)
5
Mosnier
 
LO
Griffin
 
JH
Inhibition of staurosporine-induced apoptosis of endothelial cells by activated protein C requires protease-activated receptor-1 and endothelial cell protein C receptor.
Biochem J
2003
, vol. 
373
 
Pt 1
(pg. 
65
-
70
)
6
Joyce
 
DE
Gelbert
 
L
Ciaccia
 
A
DeHoff
 
B
Grinnell
 
BW
Gene expression profile of antithrombotic protein c defines new mechanisms modulating inflammation and apoptosis.
J Biol Chem
2001
, vol. 
276
 
14
(pg. 
11199
-
11203
)
7
Mosnier
 
LO
Zlokovic
 
BV
Griffin
 
JH
The cytoprotective protein C pathway.
Blood
2007
, vol. 
109
 
8
(pg. 
3161
-
3172
)
8
Cheng
 
T
Liu
 
D
Griffin
 
JH
, et al. 
Activated protein C blocks p53-mediated apoptosis in ischemic human brain endothelium and is neuroprotective.
Nat Med
2003
, vol. 
9
 
3
(pg. 
338
-
342
)
9
Shibata
 
M
Kumar
 
SR
Amar
 
A
, et al. 
Anti-inflammatory, antithrombotic, and neuroprotective effects of activated protein C in a murine model of focal ischemic stroke.
Circulation
2001
, vol. 
103
 
13
(pg. 
1799
-
1805
)
10
Wang
 
Y
Zhang
 
Z
Chow
 
N
, et al. 
An activated protein C analog with reduced anticoagulant activity extends the therapeutic window of tissue plasminogen activator for ischemic stroke in rodents.
Stroke
2012
, vol. 
43
 
9
(pg. 
2444
-
2449
)
11
Cheng
 
T
Petraglia
 
AL
Li
 
Z
, et al. 
Activated protein C inhibits tissue plasminogen activator-induced brain hemorrhage.
Nat Med
2006
, vol. 
12
 
11
(pg. 
1278
-
1285
)
12
Liu
 
D
Cheng
 
T
Guo
 
H
, et al. 
Tissue plasminogen activator neurovascular toxicity is controlled by activated protein C.
Nat Med
2004
, vol. 
10
 
12
(pg. 
1379
-
1383
)
13
Mosnier
 
LO
Zlokovic
 
BV
Griffin
 
JH
Cytoprotective-selective activated protein C therapy for ischaemic stroke.
Thromb Haemost
2014
, vol. 
112
 
5
(pg. 
883
-
892
)
14
Williams
 
PD
Zlokovic
 
BV
Griffin
 
JH
Pryor
 
KE
Davis
 
TP
Preclinical safety and pharmacokinetic profile of 3K3A-APC, a novel, modified activated protein C for ischemic stroke.
Curr Pharm Des
2012
, vol. 
18
 
27
(pg. 
4215
-
4222
)
15
Miletich
 
JP
Broze
 
GJ
Beta protein C is not glycosylated at asparagine 329. The rate of translation may influence the frequency of usage at asparagine-X-cysteine sites.
J Biol Chem
1990
, vol. 
265
 
19
(pg. 
11397
-
11404
)
16
Ní Ainle
 
F
O’Donnell
 
JS
Johnson
 
JA
, et al. 
Activated protein C N-linked glycans modulate cytoprotective signaling function on endothelial cells.
J Biol Chem
2011
, vol. 
286
 
2
(pg. 
1323
-
1330
)
17
Preston
 
RJ
Ajzner
 
E
Razzari
 
C
, et al. 
Multifunctional specificity of the protein C/activated protein C Gla domain.
J Biol Chem
2006
, vol. 
281
 
39
(pg. 
28850
-
28857
)
18
Gleeson
 
EM
O’Donnell
 
JS
Hams
 
E
, et al. 
Activated factor X signaling via protease-activated receptor 2 suppresses pro-inflammatory cytokine production from lipopolysaccharide-stimulated myeloid cells.
Haematologica
2014
, vol. 
99
 
1
(pg. 
185
-
193
)
19
Orbe
 
J
Barrenetxe
 
J
Rodriguez
 
JA
, et al. 
Matrix metalloproteinase-10 effectively reduces infarct size in experimental stroke by enhancing fibrinolysis via a thrombin-activatable fibrinolysis inhibitor-mediated mechanism.
Circulation
2011
, vol. 
124
 
25
(pg. 
2909
-
2919
)
20
Ludeman
 
MJ
Kataoka
 
H
Srinivasan
 
Y
Esmon
 
NL
Esmon
 
CT
Coughlin
 
SR
PAR1 cleavage and signaling in response to activated protein C and thrombin.
J Biol Chem
2005
, vol. 
280
 
13
(pg. 
13122
-
13128
)
21
Mosnier
 
LO
Sinha
 
RK
Burnier
 
L
Bouwens
 
EA
Griffin
 
JH
Biased agonism of protease-activated receptor 1 by activated protein C caused by noncanonical cleavage at Arg46.
Blood
2012
, vol. 
120
 
26
(pg. 
5237
-
5246
)
22
Schuepbach
 
RA
Madon
 
J
Ender
 
M
Galli
 
P
Riewald
 
M
Protease-activated receptor-1 cleaved at R46 mediates cytoprotective effects.
J Thromb Haemost
2012
, vol. 
10
 
8
(pg. 
1675
-
1684
)
23
Cao
 
C
Gao
 
Y
Li
 
Y
Antalis
 
TM
Castellino
 
FJ
Zhang
 
L
The efficacy of activated protein C in murine endotoxemia is dependent on integrin CD11b.
J Clin Invest
2010
, vol. 
120
 
6
(pg. 
1971
-
1980
)
24
Tissue plasminogen activator for acute ischemic stroke. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group.
N Engl J Med
1995
, vol. 
333
 
24
(pg. 
1581
-
1587
)
25
Andreou
 
AP
Efthymiou
 
M
Yu
 
Y
, et al. 
Protective effects of non-anticoagulant activated protein C variant (D36A/L38D/A39V) in a murine model of ischaemic stroke.
PLoS One
2015
, vol. 
10
 
4
pg. 
e0122410
 

Author notes

E.M.G. and M.G.D. contributed equally to this study.

Sign in via your Institution