You are on page 1of 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/353415136

Platelet EVs contain an active proteasome involved in protein processing for


antigen presentation via MHC-I molecules

Article  in  Blood · July 2021


DOI: 10.1182/blood.2020009957

CITATIONS READS

22 263

24 authors, including:

Genevieve Marcoux Audrée Laroche


Lund University Laval University
17 PUBLICATIONS   785 CITATIONS    9 PUBLICATIONS   171 CITATIONS   

SEE PROFILE SEE PROFILE

Stephan Hasse Maroua Mbarik


Laval University Centre Hospitalier Universitaire de Québec (CHUQ)
8 PUBLICATIONS   66 CITATIONS    9 PUBLICATIONS   71 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Blood cell-derived microvesicles: clinical impacts and use View project

Cytohesin-1 in human neutrophils View project

All content following this page was uploaded by Eric Boilard on 23 March 2022.

The user has requested enhancement of the downloaded file.


Plenary Paper

PLATELETS AND THROMBOPOIESIS

Platelet EVs contain an active proteasome involved


in protein processing for antigen presentation
via MHC-I molecules
Genevieve Marcoux,1,2 Audr
ee Laroche,1,2 Stephan Hasse,1,2 Marie Bellio,1,2 Maroua Mbarik,1,2 Marie Tamagne,3,4,5 Isabelle Allaeys,1,2
Anne Zufferey,1,2 Tania L
evesque,1,2 Johan Rebetz,6 Annie Karakeussian-Rimbaud,7,8 Julie Turgeon,7,8 Sylvain G. Bourgoin,1,2
Hind Hamzeh-Cognasse,9 Fabrice Cognasse,10,9 Rick Kapur,11 John W. Semple,12,6 Marie-Jos ebert,7,8 France Pirenne,3,4,5
ee H
elanie Dieude,14,7,8 Beno^ıt Vingert,3,4,5,* and Eric Boilard1,2,8
Herman S. Overkleeft,13 Bogdan I. Florea,13 M
1
Centre de Recherche, Centre Hospitalier Universitaire de Qu ebec–Universit ebec, QC, Canada; 2Centre de Recherche Arthrite, Facult
e Laval, Qu e de Medecine
de l'Universit
e Laval, Quebec, QC, Canada; 3Institut Mondor de Recherche Biom edicale (IMRB), University Paris Est Creteil, INSERM, Cr
eteil, France;
4
Etablissement Français du Sang, Ivry sur Seine, France; 5Laboratory of Excellence GR-Ex, Paris, France; 6Division of Hematology and Transfusion Medicine, Lund
University, Lund, Sweden; 7Research Centre, Centre Hospitalier de l'Universit e de Montr eal (CRCHUM), Montr eal, QC, Canada; 8Canadian Donation and
Transplantation Research Program, Edmonton, AB, Canada; 9Universit e de Lyon, Universit 
e Jean Monnet, INSERM U1059, Saint-Etienne, France; 10Etablissement
Français du Sang Auvergne-Rh^ one-Alpes, Saint-Etienne, France; 11Sanquin Research, Department of Experimental Immunohematology, Amsterdam and
Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; 12Departments of Pharmacology and Medicine, University of
Toronto, Toronto, ON, Canada; 13Gorlaeus Laboratories, Leiden Institute of Chemistry and Netherlands Proteomics Centre, Leiden, The Netherlands; and
14
D
epartement Microbiologie, Infectiologie et Immunologie, Facult
e de M
edecine, Universit
e de Montr
eal, Montr
eal, QC, Canada

KEY POINTS
In addition to their hemostatic role, platelets play a significant role in immunity. Once
 Platelet-derived activated, platelets release extracellular vesicles (EVs) formed by the budding of their
extracellular vesicles cytoplasmic membranes. Because of their heterogeneity, platelet EVs (PEVs) are thought to
contain an active
perform diverse functions. It is unknown, however, whether the proteasome is transferred
proteasome.
from platelets to PEVs or whether its function is retained. We hypothesized that functional
 Platelet-derived protein processing and antigen presentation machinery are transferred to PEVs by activated
extracellular vesicles can platelets. Using molecular and functional assays, we found that the active 20S proteasome
process and present
antigen. was enriched in PEVs, along with major histocompatibility complex class I (MHC-I) and
lymphocyte costimulatory molecules (CD40L and OX40L). Proteasome-containing PEVs
were identified in healthy donor blood, but did not increase in platelet concentrates that
caused adverse transfusion reactions. They were augmented, however, after immune complex injections in mice. The
complete biodistribution of murine PEVs after injection into mice revealed that they principally reached lymphoid organs,
such as spleen and lymph nodes, in addition to the bone marrow, and to a lesser extent, liver and lungs. The PEV
proteasome processed exogenous ovalbumin (OVA) and loaded its antigenic peptide onto MHC-I molecules, which
promoted OVA-specific CD81 T-lymphocyte proliferation. These results suggest that PEVs contribute to adaptive
immunity through cross-presentation of antigens and have privileged access to immune cells through the lymphatic
system, a tissue location that is inaccessible to platelets.

Introduction invasion, organ and tissue damage may also favor platelet
Platelets are the second most abundant lineage in the blood and activation and inflammation in chronic inflammatory diseases .2-8
are best known for their role in hemostasis.1 They are small
fragments produced by the large, multinucleated megakaryocyte Albeit anucleate, the platelet cytoplasm includes numerous
in the bone marrow. They bear receptors that permit recruitment molecules comprising the proteasome that are transferred from
of immune cells and carry an extensive set of immune and megakaryocytes to their progeny. The proteasome is a high-
inflammatory molecules (eg, cytokines/chemokines, lipid media- molecular-weight cylindrical protein complex through which
tors, and hormones) stored in their granules or cytoplasm or unwanted or damaged proteins are degraded.9,10 The central
synthesized by messenger RNA translation after platelet activa- complex part, called the 20S proteasome, is made up of 28
tion. Thus, although platelets may mount an innate immune distinct subunits,11 comprising the 3 catalytic subunits necessary
response against injury, which is critical to combating pathogen for the degradation of proteins into peptides of 3 to 15 amino

© 2021 by The American Society of Hematology blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 2607
A B 0.6 C 0.5 D 0.30
PLTs PEVs

Proteasome 20S α/Actin (AU)


*
35 kDa 0.5 0.4 0.25
Proteasome

TSG10/Actin (AU)
CD41/Actin (AU)
subunit 20S α 0.4 0.20
25 kDa 0.3
0.3 0.15
0.2
0.2 0.10
CD41 100 kDa 0.1
0.1 0.05

0.0 0.0 0.00


PLTs PEVs PLTs PEVs PLTs PEVs
48 kDa
TSG101
E F *
2.0 70000 Control

Proteasome activity/
60000 Epoxomicin

μg of protein
TOM20 1.5 50000
TOM20/Actin (AU)
11 kDa **
40000
1.0 30000 ***
Actin 48 kDa 20000
0.5 10000
0
PLTs PEVs PLTs PEVs PLTs PEVs
0.0
PLTs PEVs Trypsin Caspase Chymotrypsin

G H ****
I ****
** ****
100
CD41+ proteasome+ (%)

****

CD41+ proteasome+ PEVs


****
80 100

(% of control)
60 80
60
40 *** 40
20 20
0 0
s

Vs

l
tra in

n tri

la 0
d
et

EV

ox tro

Un -10

le
PE

Ul mic

ito n
el

lP

be
500 nm Tr ce
Ep on
at

X
e

o
al
Pl

C
rg

Sm
La

J
Control Epoxomicin Ultracentri Triton X-100 Unlabeled
α-CD41

CD41
Proteasome + LWA 300 Supernatant

Proteasome unlabeled
PEVs

K
Platelets Platelets-ROI PEVs PEVs-ROI
CD41 WGA CD41 WGA

3.00 μm 3.00 μm 3.00 μm 3.00 μm


LWA300 Merge LWA300 Merge

12.00 μm 3.00 μm 3.00 μm 12.00 μm 3.00 μm 3.00 μm

Figure 1. Platelets and PEVs contain proteasome. (A) Proteasome 20S a subunit, CD41, TOM20, TSG101 and actin in human PEVs (18 000g fraction) and platelet (PLTs)
preparations (20 mg protein per lane) were assessed by immunoblot analysis. Results are representative of 5 distinct preparations. (B-E) Protein quantifications were assessed
by densitometry with laboratory imaging software (BioRad). Results were normalized to actin and expressed as arbitrary units (AU). Mean 6 SEM (n 5 5). (D) *P , .05 (paired
Student t test). (F) Proteasome function was assessed by measuring trypsin-like, caspase-like, or chymotrypsin-like activity of PEVs and platelets treated or not with
epoxomicin using the Proteasome-Glo chymotrypsin-like, trypsin-like, and caspase-like cell-based assays. Twenty and 10 mg of proteins was used for platelets and PEVs, respectively.

2608 blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 MARCOUX et al


acids in length.11,12 Proteasome activity in megakaryocytes is was gently resuspended in 600 mL Tyrode’s buffer (pH 7.4).
necessary for platelet production13,14 and in platelets, the Platelets were either left nonactivated or activated with thrombin
proteasome regulates platelet lifespan,15 activation,16-18 and (0.1 U/mL) after addition of 5 mM of calcium for 90 minutes at
release of platelet EVs (PEVs).19,20 The platelet proteasome can room temperature (RT; time based on the kinetics of
hydrolyze proteins into smaller peptides,11,21,22 thereby enabling CD411proteasome1 PEV release shown in supplemental Figure
peptide loading onto the platelet major histocompatibility com- 3C). PEVs were obtained by 2 rounds of centrifugation of
plex class I (MHC-I) molecules.23-25 Components of the peptide- stimulated platelets at 1300g for 5 minutes at RT. Activated
loading complex are also expressed in platelets and are found in platelets, EVs, or dendritic cells (DCs) were pulsed with 100 mg/mL
proximity with MHC-I during platelet activation.26,27 Platelets can ovalbumin (OVA) protein (Sigma-Aldrich) or 200 mg/mL of OVA
efficiently form an immunological synapse with T lymphocytes to peptide (SIINFEKL [Invivogen]) or were left unpulsed for 4 hours at
activate lymphocyte proliferation,26,28,29 and they are known to RT. These conditions were either left unlabeled for lymphoprolif-
fulfill roles in cross-presentation of antigens in adaptive immunity. eration and intracellular staining experiments or labeled for
In a similar manner, megakaryocytes cross-present antigens to Hs-flow cytometry (FCM) experiments.
CD8 T lymphocytes, thereby suggesting that they may also play a
dual role in innate and adaptive immunity.25,30-32 Five microliters of PEVs or platelet suspensions were labeled with
250 nM LWA300 proteasome probe in a total volume of 100 mL
EVs, produced in abundance by platelets, are small (up to 1 mm in for 90 minutes at 30 C. Samples were then incubated with the
diameter), membrane-bound vesicles released from the plasma following antibodies for 30 minutes at RT before dilution in
membrane or endosomal compartments of activated cells. PEVs annexin V binding buffer and analysis by high sensitivity-FCM (hs-
are heterogeneous in surface molecules and content (eg, nucleic FCM): BUV395 anti-CD41, BV650 anti-CD62p, BUV395 anti-
acids, lipids, transcription factors, enzymes, and mitochondria) and CD41, BV650 anti-CD62p, BV711 annexin V, BV421 anti-OX40L,
may have diverse functions beyond hemostasis.33-35 For instance, BUV737 anti-CD154 (all from BD Biosciences), PeCy7 anti-CD40,
PEVs convey mitochondrial components that are associated with
PeCy7 anti-MHC-I (AF6-88.5), and PE anti-MHC-I bound to OVA
inflammation and adverse transfusion reactions (ATRs).36-38
peptide (25D1.16) (all from Biolegend).
Despite the fact that platelets are restricted to the blood
circulation, PEVs can cross tissue barriers and enter the synovial
fluid,39,40 lymph,41,42 and bone marrow43 where they can deliver Results
platelet-derived molecules and modulate target cells.34 For
PEVs contain functional proteasome
instance, PEVs promote the formation of germinal centers and
After platelet activation by thrombin, remnant platelets were
the production of IgG by B cells.44,45 They also interact with and
eliminated by centrifugation, and larger EVs were isolated by a
modulate regulatory T-cell differentiation and activity.46,47 Thus,
second high-speed centrifugation (18 000g fraction). The super-
PEVs may be able to transport platelet-derived molecules relevant
natant obtained was further centrifuged at 100 000g, and smaller
to adaptive immunity into lymphoid organs. However, it is
EVs (probably exosomes) were obtained from the pellet. We
unknown whether the proteasome and the molecules necessary
found that 98.1% 6 0.5% of proteins were retrieved in the larger
for antigen presentation are also transferred during the budding
EV (18 000g) fraction. Immunoblot analysis confirmed that human
of PEVs. In this study, we sought to find out whether functional
protein processing and antigen presentation machinery are PEVs from this fraction were enriched in the proteasome 20S
transferred to PEVs by activated platelets. a-subunit, in addition to mitochondria (indicated by TOM20
expression) and CD41, but lacked TSG101 (a putative marker of
exosomes; Figure 1A-E). Using platelets as a positive control, we
Material and methods assessed proteasome function in these PEVs. Proteasome-
More details are presented in the supplemental Methods (avail- associated trypsinlike, caspaselike, and chymotrypsinlike activities
able on the Blood Web site). were detectable in platelets and were significatively increased in
the PEV fraction, but were undetectable after treatment with
Labeling of murine platelets, DCs, and PEVs epoxomicin, a proteasome inhibitor (Figure 1F). Visualization of
Platelets were isolated from C57BL/6J mice by retro-orbital or immunogold-labeled proteasome 20S a subunit by transmission
cardiac puncture in 200 mL ACD and 350 mL Tyrode’s buffer (pH electron microscopy confirmed the presence of proteasomes in
6.5). Whole blood was centrifuged at 600g for 3 minutes and then PEVs (Figure 1G). These data suggest that the catalytically active
at 400g for 2 minutes to remove red blood cells. Supernatant was proteasome was transferred to PEVs upon its release from the
spun at 1300g for 5 minutes, and the platelet-containing pellet platelets.

Figure 1 (continued) Mean 6 SEM (n 5 6). *P , .05, **P , .01, ***P , .001 (Mann-Whitney U test). (G) TEM visualization of immunogold labeling of proteasome 20S a
subunit in PEVs released from thrombin (0.5 U/mL)-activated platelets (arrowheads). Data are representative of 3 independent experiments. (H) hs-FCM analysis of resting
platelets and thrombin (0.5 U/mL)–activated platelets. Two distinct populations of PEVs, (larger PEVs [17% of these PEVs contain active proteasome] and smaller PEVs)
that do not contain active proteasome (n 5 20). Data are the mean 6 SEM. **P , .01; ***P , .001; ****P , .0001 (Kruskal–Wallis). (I-J) Controls were performed to assess
the specificity of PEV detection using hs-FCM. Sensitivity of CD411proteasome1 PEVs to competition by epoxomicin, ultracentrifugation (Ultracentri), or 0.05% Triton
X-100 and unlabeled samples are presented as the percentage of untreated (Control). Data are the mean 6 SEM of 5 independent experiments. ****P , .0001 vs control
(paired Student t test). (K) Confocal microscopy visualization of proteasome content associated with platelets (left) and PEVs (right). Visualization of CD41, wheat germ
agglutinin (WGA) to determine plasma membrane surface and proteasome (LWA300), and merger of the stains is displayed in the region of interest (ROI). Populations
originating from dashed-line squares and represented in ROI are triple positives (arrowheads) or CD411 and WGA1 but proteasome2 (arrows). SEM, standard error of the
mean; TEM, transmission electron microscopy.

ACTIVE PROTEASOME IN EXTRACELLULAR VESCICLES blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 2609
A
4.0×107 6.0×106 4.0×107 6.0×108

Proteasome+ mito+ PEVs/mL


Proteasome+ mito– PEVs/mL

Proteasome– mito+ PEVs/mL


Total proteasome+ PEVs/mL
NS NS NS ****
3.0×107 3.0×10 7
4.0×106 4.0×108

1.0×107 1.0×10 7

2.0×106 2.0×108
1.0×107 1.0×107

0 0 0 0
Control ATR Control ATR Control ATR Control ATR

B C
Bronchoalveolar lavages 2.0×107 HA-IgG injected PPP 2.0×106
NS
Proteasome+ PEVs/mL

Proteasome+ PEVs/mL
**
1.5×107 1.5×106

1.0×107 1.0×106
Diluent injected
PPP
0.5×107 0.5×106

Control TRALI-induced
0 0
mice mice Control TRALI Diluent HA-lgG

Figure 2. Identification of proteasome-containing PEVs under physiological and pathological conditions. (A) Proteasome-containing PEVs detected by hs-FCM are
found in PFP from platelet concentrates that caused ATRs in recipients and in control concentrates that did not induce ATRs. The total number of proteasome-containing
PEVs (containing mitochondria [mito] or not; proteasome1mito1 PEVs or proteasome1mito2 PEVs) did not significantly differ between control and ATR, whereas
proteasome2mito1 PEVs increased in ATR (no ATR group [n 5 33] vs the ATR reaction group [n 5 34], matched in duration of storage; data are the mean 6 standard error
of the mean [SEM]; ****P , .0001 (Student t test). NS, nonsignificant. (B) Proteasome-containing PEVs detected by hs-FCM are found in bronchoalveolar lavage fluid from
mice after induction of transfusion-related acute lung injury (TRALI) with the 34-1-2s and AF6-88.5.5.3 antibodies and in control mice (n 5 5) Data are the mean 6 SEM;
Student t test. NS, nonsignificant. (C) Proteasome-containing PEVs were detected at significantly higher levels in mice 1 hour after IV injection of HA-IgG vs control (diluent)
mice (n 5 3). Data are the mean 6 SEM. **P, .01 (Student t test). PFP, platelet-free plasma.

LWA300 is a conjugate between epoxomicin and BODIPY FL release of active proteasome1 PEVs revealed that the total
fluorophore that generates an activity-based, plasma membrane- number of PEVs (with and without proteasomes) was significantly
permeable inhibitor that can identify the proteasome in cells.48,49 reduced in the presence of actin inhibitors (cytochalasins B, D, and
Using LWA300, we detected and quantified active proteasome- E and latrunculin A) but not by the tubulin polymerization inhibitor
containing PEVs directly in the platelet secretome.48,49 hs-FCM nocodazole (supplemental Figure 2B). Proteasome release in PEVs
confirmed PEV heterogeneity after platelet activation by thrombin was not unique to thrombin stimulation, as ADP, cross-linked
(Figure 1H). Approximately 16.6% 6 6.5% of the larger (ie, 500- collagen–related peptide (CRP-XL), and heat-aggregated IgG
900 nm) PEVs50 contained proteasome, whereas the smaller (HA-IgG) also triggered release of proteasome-containing PEVs
vesicles (ie, ,500 nm) had no detectable proteasome (Figure 1H; (supplemental Figure 2C).
supplemental Figure 1). The detection specificity of proteasome-
containing PEVs by hs-FCM was confirmed by using a combina- Identification of proteasome-containing PEVs
tion of controls. We confirmed efficient competition of the under physiological and pathological conditions
LWA300 probe by unlabeled epoxomicin, and we determined The presence of proteasome-containing PEVs was assessed under
the particulate nature and membrane moiety of proteasome- conditions conducive to platelet activation and PEV release. A
containing PEVs, as they were respectively pelleted by ultracen- mean of 1.82 3 106 (range, 1.13 3 105 to 8.11 3 106; n 5 6)
trifugation and sensitive to detergent treatment (Figure 1I-J). proteasome-containing PEVs per mL were detected by hs-FCM in
Confocal microscopic visualization of platelets as positive controls, the blood of healthy individuals, which corresponded to 2.6% 6
and PEVs from thrombin-activated platelets labeled with LWA300 1.8% of the total PEVs in blood. PEVs were quantified in platelet
revealed that both platelets and a subpopulation of PEVs concentrates (PCs) known to have caused ATRs and compared
contained active proteasome (Figure 1K). with control PCs that did not induce ATRs. Given the reported
increase in mitochondria-containing PEVs in ATRs,36,37 we also
hs-FCM was further used to characterize proteasome-containing determined their levels. High levels of proteasome-containing
PEVs in terms of surface markers and mitochondrial content. PEVs were found in all tested PCs (Figure 2A) but the
Approximately half of the proteasome-containing PEVs exposed concentrations of proteasome-containing PEVs (with or without
phosphatidylserine, whereas most expressed surface P-selectin mitochondria) were not significantly elevated in PCs that induced
(supplemental Figure 2A). Furthermore, 68.3% 6 7.8% of the ATRs (Figure 2A). In contrast, compared with the controls, the
proteasome-containing PEVs also contained mitochondria (sup- concentrations of mitochondria-containing PEVs were increased in
plemental Figure 2A). Investigation of the mechanisms underlying ATR-associated PCs, consistent with prior findings.36,37

2610 blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 MARCOUX et al


A Resting Thrombin B Platelets

40 ****
Platelets

SSC

(% of CD41+ events)
30
CD41+ CD41+

MHC I positives
CD41

20

10

SSC
MHC I+ MHC I+
0
MHC I Resting Thrombin
SSC

Basal Thrombin
PEVs
20 ****
SSC

FSC
PEVs

(% of CD41+ events)
15

MHC I positives
CD41 CD41+ CD41+

10

5
SSC

MHC I+ MHC I+ 0
MHC I Basal Thrombin

C CD41+ activated platelets D Activated E Activated


platelets platelets
250 **** 150 ****
CD41+ CD62p+ events/µL

CD41+ CD62p+ events/µL


200
25D1.16 positives

25D1.16 positives
CD62P

25D1.16+
100
150
Unpulsed
25D1.16 100
50
50
25D1.16+
0 0
SIINFEKL – + OVA – +
Pulsed
CD41+ PEVs PEVs PEVs
250 **** 150 ****
CD41+ CD62p+ events/µL

CD41+ CD62p+ events/µL

200
25D1.16 positives

25D1.16 positives
CD62P

25D1.16+ 100
150

Unpulsed
100
25D1.16 50
50

25D1.16+ 0 0
SIINFEKL – + OVA – +
Pulsed

Figure 3. Platelets and PEVs load and process OVA onto MHC-I. (A-B) Thrombin (0.1 U/mL)–activated murine platelets and their PEVs express MHC-I (detected by
hs-FCM; n 5 19). Data are the mean 6 standard error of the mean (SEM). ****P , .0001 (Mann-Whitney U test). Activated platelets and their PEVs loaded the SIINFEKL
peptide (C-D) or processed and loaded OVA (C-E) onto MHC-I (n 5 19). Data are the mean 6 SEM. ****P , .0001, pulsed (1) vs unpulsed (2) (Kruskal-Wallis test).

ACTIVE PROTEASOME IN EXTRACELLULAR VESCICLES blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 2611
2612
B

C
A

D
PEV aggregates Individual PEVs 60 min 15 min Vehicle
CMFDA+ PEVs
(number/mm2) (number/mm2)
SSC-H

2×10
8×10
PB

0
5
10
15
20
0
100
200
300
400
0
7
4×107
6×107
7
1×108

10

101
102
3
104
105
S-
60
PE m 2
V- in m
15 in

**
**
PE m
V- in

Spleen
Spleen
*

60 15
m m
in in
****

EVs
60

FSC-PMT-H
m
PEV aggregates Individual PEVs in
(number/mm2) (number/mm2)

101 102 103 104 105


PB CMFDA+ platelets

0
50
100
150
200
250

0
5
10
15
20
SSC-H S-

10
60 (% of CD41+ platelets)

101
102
3
104
105
PE m
in
0.0
0.2
0.4
0.6
0.8
1.0

V-
15

**

**
PE m

PLN
PLN

V- i 2
60 n m
m in
in
15
**

CD41
in
**

PEV aggregates Individual PEVs


(number/mm2) (number/mm2) 60
m

CD41+
in

101 102 103 104 105


PB

0
10
20
30
0
100
200
300
400
S-
60
MHC I PE m

10
in

blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25


CMFDA+ leukocytes

101
102
3
104
105
V-

**

**
15

ILN
ILN

PE m (% of CD45+)
V- i
0
10
20
30
40

60 n

*
m
in
2
m
in
PEV aggregates Individual PEVs
*

Figure 4.
Labelled
15
(number/mm2)

Proteasome
(number/mm2) m
in
**

PB

0
2
4
6
8
10
0
20
40
60
80
100
60

101 102 103 104 105


S-
60 m
PE m in
MHC I V- in

**

**
15

10
PE m

101
102
3
104
105
V- i
60 n CMFDA+ neutrophils

*
m

Bone marrow
Bone marrow

in (% of Ly6G+ cells)
0
20
40
60
80

Individual PEVs
PEV aggregates 2
(number/mm2) m
in
(number/mm2)

Proteasome
Unlabelled
PB

0
5
10
15
20

0
2
4
6
15
S-
60 m
in
**

101 102 103 104 105


PE
V- in
15 60

**
m m
Lung
Lung

PE
V- i in

E
% of CD41+ PEVs 60 n
m
in

0
10
20
30
50
100
M CMFDA+ lymphocytes
H
C Individual PEVs (% of CD3+)
I+ PEV aggregates
0
1
2
3

(number/mm2)
+ (number/mm2)
/+ PB 2

0
100
200
300

0
1
2
3
4
5
S- m
60 in
LW PE m
V- in 15
A+ m
**

15 **
Liver
Liver

PE m in
V- in
– 60 60
/– m m
in in

MARCOUX et al
Transfusion-related acute lung injury (TRALI) is a potentially lethal molecules (Figure 3C-D). Native OVA was also efficiently
adverse reaction that can result from transfusion of PCs.51 Thus, processed by platelets, and the SIINFEKL peptide was loaded
we quantified proteasome-containing PEVs in murine bronchoal- into MHC-I (Figure 3C-E), consistent with prior work.26 We found
veolar lavages in an inducible TRALI model.52,53 Proteasome- that an average of 2.53% 6 0.74% of CD411 PEVs pulsed with the
containing PEVs were detected in bronchoalveolar lavages from peptide and 1.83% 6 0.24% of CD411 PEVs pulsed with OVA (n
both TRALI and control mice (Figure 2B); however, no significant 5 18) were positive for 25D.1.16. Of interest, incubation of native
difference was observed between the 2 groups (Figure 2B), which OVA with PEVs resulted in proteolysis of the former and retrieval
suggests that proteasome-containing PEVs are not increased of the SIINFEKL peptide from MHC-I molecules expressed by the
during lung inflammation in this model and therefore may not PEVs. Taken together, the data show that PEVs can process native
participate in the acute inflammation that characterizes the proteins into smaller peptides thereby enabling antigen presen-
pathogenesis. tation through MHC-I.

Our in vitro investigations pointed to the high potency of immune Proteasome-containing PEVs can reach lymphoid
complexes (HA-IgG) in generating proteasome-containing PEVs organs and circulate through the lymphatic system
(supplemental Figure 2C). Although mice lack FcgRIIA, that IV injected PEVs have a limited circulation time in human blood,
receptor is the only Fcg receptor expressed by human platelets ranging from 10 minutes to hours, depending on the study.60,61 It
that is capable of responding to immune complexes.54 Recent is unclear, however, whether they can reach lymphoid organs.
findings indicate that circulating immune complexes stimulate the Fluorescently labeled PEVs generated from activated mouse
release of mitochondria-containing PEVs in mice expressing the platelets were IV injected into mice, and their presence in blood
FcgRIIA transgene.55,56 Compared with the diluent-injected con- and different organs was monitored. We identified free PEVs
trol mice, the mice with immune-complex challenge showed (unbound to cells) for up to 2 minutes in blood (Figure 4A;
significantly elevated levels of proteasome-containing PEVs in supplemental Figure 4A-C). PEVs in blood were also found bound
plasma (Figure 2C). These findings confirmed that proteasome- to platelets and to leukocytes, mainly Ly6G1 neutrophils, and to a
containing PEVs are present under various physiological and lesser extent, lymphocytes, but were mostly undetectable by 60
pathological conditions. minutes (Figure 4A). Screening of individual PEVs in whole tissue
sections in different organs identified spleen and lymph nodes
Protein processing by proteasome- (popliteal and inguinal) as primary targets, followed by liver, bone
containing PEVs marrow, lungs, and kidneys, whereas none were found in brain
To study proteasome function in PEVs, we investigated its ability (Figure 4B-C; supplemental Figure 4D-E). Moreover, aggregates
to process proteins into smaller peptides by assessing their of PEVs (ie, larger than 1 mm2 and up to 541 mm2) were mainly
successful loading into the antigen-binding groove of MHC-I observed in spleen (mean size, 2.84 6 0.16 mm2) and popliteal
molecules. We confirmed the expression of MHC-I on resting and (4.16 6 0.40 mm2) and inguinal (4.08 6 0.30 mm2) lymph nodes,
thrombin-activated murine platelets and verified whether MHC-I is followed by bone marrow (2.75 6 0.15 mm2), lung (33.51 6 7.15
maintained on PEVs present in the platelet secretome. We found mm2), and liver (3.40 6 0.21 mm2). This distribution may reflect their
that washed resting platelets did not express MHC-I on their accumulation in smaller vessels or the internalization of numerous
surface (Figure 3A-B); however, thrombin activation led to a PEVs within single cellular recipients in these organs (Figure 4B-C;
significant increase in surface MHC-I expression (Figure 3A-B), supplemental Figure 4D-E).
consistent with the reported presence of this molecule in
a-granules and its release upon activation.26,27,57,58 A small PEVs can circulate through the lymphatic system, and the levels of
proportion (0.93% 6 0.13%) of the spontaneously released PEVs in lymph are increased in mouse models of atherosclerosis
PEVs expressed MHC-I, but this proportion significantly increased and autoimmune inflammatory arthritis.34,41,42 Using the lymph
upon platelet activation with thrombin (mean, 4.64% 6 0.98%). from mice, we assessed to determine whether PEVs are associated
with proteasome and MHC-I molecules. We found that a fraction
To determine whether PEV MHC-I can indeed load small of the PEVs in lymph expressed MHC-I (11.2% 6 2.2%) and
peptides, we pulsed PEVs present in the platelet secretome with contained an active proteasome (12.0% 6 3.9%). Remarkably, a
the OVA peptide SIINFEKL and monitored its association with detectable proportion (1.6% 6 0.7%) of the lymph PEVs
MHC-I molecules using the 25D1.16 monoclonal antibody, which contained both proteasome and MHC-I molecules (Figure 4D-
specifically recognizes MHC-I/SIINFEKL complexes.59 Similar to E), significant given the substantial number of PEVs in lymph
platelets, PEVs loaded the SIINFEKL peptide onto their MHC-I (mean, 2.5 3 107/mL in mice41).

Figure 4. PEVs in blood circulation can reach lymphoid organs and circulate in lymph. (A-C) Fluorescently labeled PEVs generated from activated mouse platelets were
IV injected into mice and their presence in blood (A) and different organs (B-C) was monitored after 2, 15, and 60 minutes. Free PEVs (unbound to cells) were identified by
flow cytometry for up to 2 minutes in blood, as well as PEVs bound to platelets and to leukocytes (mainly Ly6G1 neutrophils and a few lymphocytes), but were mostly
undetectable by 60 minutes. Dashed lines represent the mean of vehicle (n 5 9-13); n 5 11 (2 minutes), n 5 5 (15 minutes) and n 5 6 (60 minutes). Data are the mean 6
standard error of the mean (SEM). *P , .05, **P , .01, ***P , .001 (Kruskal-Wallis). (B) Representative images of CMFDA-labeled (green) individual PEVs (arrowhead) and
PEV aggregates (asterisk) in whole tissue sections (spleen, popliteal LN [PLN], inguinal LN [ILN], bone marrow, lungs, and liver) at 15 and 60 minutes by confocal microscopy;
nuclei (Hoechst 33342 ) are blue. Results are representative of observations made in 5 and 6 mice per group. (C) PEVs and aggregates were quantified using 5 different
sections for lymph nodes (PLN and ILN; representing a total surface of at least 1.5 mm2), 8 zones of 500 000 mm2 each, randomly assigned on 2 different sections in femurs
(total surface, 4 mm2), and 10 zones of 500 000 mm2 each, randomly assigned on 2 sections each of lungs, spleen, kidneys, and brain and 1 section of liver (total surface, 5
mm2) using Zen 3.3 software (n 5 6 [phosphate buffered saline, 60 minutes], 5 [15 minutes], and 5-6 [60 minutes]). Data are the mean 6 SEM. *P , .05, **P , .01 (Kruskal-
Wallis). (D-E) PEVs in lymph were detected by hs-FCM. (D) Gating strategy to analyze expression of MHC-I and proteasome (LWA300) on CD411 EVs in lymph and
representative dot plot of labeled and unlabeled (CD41 only) lymph. (E) Expression of MHC-I and proteasome (LWA300) on CD411 EVs in lymph was determined. 1/1,
double positive, and 2/2, double negative for MHC-I and proteasome (n 5 6). Data are the mean 6 SEM.

ACTIVE PROTEASOME IN EXTRACELLULAR VESCICLES blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 2613
A B C D
PLT proteasome 25D1.16– EVT proteasome 25D1.16–
2500 * 25D1.16+ 250 * 25D1.16+
2000 200

LWA 300 MFI


LWA 300 MFI

Count

Count
1500 150
1000 100
500 50
101 102 103 104 105 101 102 103 104 105
0 Proteasome 0 Proteasome
25D1.16 – 25D1.16+ 25D1.16 – 25D1.16+
PLT Annexin V EV Annexin V
25000 * 2500 NS

Annexin V MFI
Annexin V MFI

20000 2000

Count

Count
15000 1500
10000 1000
5000 1 2 3 4 5 500
10 10 10 10 10 101 102 103 104 105
0 Annexin V 0 Annexin V
25D1.16 – 25D1.16+ 25D1.16 – 25D1.16+
PLT CD40L EV CD40L
2000 * 2000 *

1500 1500

CD40L MFI
CD40L MFI

Count

Count
1000 1000

500 500
101 102 103 104 105 101 102 103 104 105
0 CD40L 0 CD40L
25D1.16 – 25D1.16+ 25D1.16 – 25D1.16+
PLT P-selectin EV P-selectin
8000 * 2500 *
P-selectin MFI
P-selectin MFI

6000 2000
Count

Count
1500
4000
1000
2000 500
101 102 103 104 105 101 102 103 104 105
0 P-selectin 0 P-selectin
25D1.16 – 25D1.16+ 25D1.16 – 25D1.16+
PLT CD40 EV CD40
20000 * 8000 *

15000 6000
Count

Count
CD40 MFI
CD40 MFI

10000 4000

5000 10 1
10 2
10 3
10 4
10 5 2000 101 102 103 104 105
CD40 CD40
0 0
25D1.16 – 25D1.16+ 25D1.16 – 25D1.16+
PLT OX40L EV OX40L
3000 * 6000 *
Count

Count
OX40L MFI
OX40L MFI

2000 4000

1000 2000
101 102 103 104 105 101 102 103 104 105
OX40L OX40L
0 0
25D1.16 – 25D1.16+ 25D1.16 – 25D1.16+

Figure 5. Platelets and PEVs loaded with OVA peptide express activation and costimulatory molecules. (A-B) Activated platelets and (C-D) PEVs loaded with OVA
peptide (25D1.161) express higher levels of proteasome (LWA300), activation (annexin V, P-selectin), and costimulatory molecules (CD40, CD40L, and OX40L). (A,C) Mean
fluorescence intensity (MFI) of the different markers assessed by hs-FCM (n 5 7). Data are mean 6 standard error of the mean (SEM). *P , .05 (Student t test). NS
nonsignificant. (B,D) Representative MFI histograms of the 25D1.162 and 25D1.161 populations for each marker shown on CD411proteasome1 events.

Proteasome-containing PEVs express lymphocyte known PEV markers displayed by CD411proteasome1 EVs.
costimulatory molecules Compared with PEVs that had undetectable SIINFEKL loading,
Efficient stimulation of adaptive immunity requires both recogni- both platelets and PEVs present in the platelet secretome loaded
tion of the antigen–MHC-I complexes by the T-cell receptor and with SIINFEKL (25D1.161) expressed higher levels of proteasome
the activity of costimulatory molecules. We examined to find (Figure 5). Moreover, in contrast to thrombin-activated platelets,
whether platelets or proteasome-containing PEVs loaded with where phosphatidylserine expression is increased when loaded
SIINFEKL express costimulatory molecules in addition to other with SIINFEKL, both PEVs bearing SIINFEKL and those negative

2614 blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 MARCOUX et al


A NS

SIINFEKL
C57BL/4J mice Isolate Harvest OT-1 mice
Harvest Isolate DC O/N
lymphocytes spleen
spleen
NS
Harvest SIINFEKL 30
ICS
blood
Thrombin staining
OVA 20
activation
and Activated
platelets NS 10
Isolate centrifugation
platelets SIINFEKL 0
Surface markers and cytokines
PEVs OVA

IFN-Y CD40
B 30 C 40
**
42

42
(% of CD3+ CD8+ cells)

(% of CD3+ CD8+ cells)


07

07
NS ** ** NS
0.

0.
30 *

CD40 positives
IFN-y positives

42
20

07
NS * NS * NS

0.
20
10
10

0 0
Pulsed – + – + OVA – + OVA Pulsed – + – + OVA – + OVA
DC Activated PLTs PEVs DC Activated PLTs PEVs

OX40 IL-2
D 10 E 2.5
(% of CD3+ CD8+ cells)

(% of CD3+ CD8+ cells)

8 2.0
NS
OX40 positives

IL-2 positives

6 NS NS NS NS NS NS 1.5 NS
NS NS NS NS NS NS
4 1.0

2 0.5

0 0.0
Pulsed – + – + OVA – + OVA Pulsed – + – + OVA – + OVA
DC Activated PLTs PEVs DC Activated PLTs PEVs

Figure 6. PEVs induce antigen-specific T-cell activation and cytokine production through antigen presentation. (A) The experimental plan. Cells and PEVs used for the
stimulation of lymphocytes assessed by intracellular cytokines staining (ICS). NS, unpulsed; O/N, overnight. (B-E) Expression of receptors or cytokines by CD31CD81 T cells
coincubated with DCs, activated platelets (PLTs), or PEVs left unpulsed or pulsed with SIINFEKL (PP), or OVA. IFN-g production (B), CD40 expression (C), OX40 expression
(D), and IL-2 production (E). Dashed lines are unstimulated conditions (n 5 6, 7, or 9). Data are the mean 6 SEM. *P , .05, **P , .01 vs unpulsed (Wilcoxon).

for SIINFEKL expressed similar levels of phosphatidylserine washed, and the expression of CD40, OX40, IL-2, and IFN-g was
(Figure 5). Furthermore, both platelets and SIINFEKL-bearing recorded to assess T-cell activation.
PEVs expressed higher levels of P-selectin, and the costimulatory
molecules CD40L, CD40, and OX40L (Figure 5). Thus, among the Compared with DCs and platelets, PEVs induced a significant
different subtypes of PEVs, those with a higher density of antigen- release of IFN-g when pulsed with the OVA peptide, whereas
MHC-I complexes show more abundant expression of lymphocyte native OVA led to an increase in IFN-g that did not reach statistical
costimulatory molecules and bear a higher content of active significance (Figure 6B). Moreover, DCs and, to a lesser extent,
proteasome. platelets and PEVs were capable of inducing only significant CD40
expression by T lymphocytes previously pulsed with the OVA
peptide (Figure 6C). In contrast, OX40 and IL-2 expression was not
Proteasome-containing PEVs can support antigen- induced by DCs, platelets, or PEVs under these experimental
specific T-cell activation conditions (Figure 6D-E).
T cells isolated from OT-1 mice were coincubated for 18 hours
with PEVs present in the platelet secretome that were either Whether PEVs could stimulate T-cell proliferation, a hallmark
pulsed or not with the SIINFEKL peptide or native OVA. DCs and response by the lymphocyte antigen-MHC-I complex was
platelets were treated similarly as positive controls and for explored. T cells from OT-1 mice were labeled with carboxy-
comparison (Figure 6A). The T cells (CD31CD81) were then fluorescein diacetate succinimidyl ester (CFSE) to monitor cellular

ACTIVE PROTEASOME IN EXTRACELLULAR VESCICLES blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 2615
A NS

SIINFEKL
CFSE Isolate Harvest OT-1 mice
C57BL/4J mice Harvest Isolate DC 7 days labelling lymphocytes spleen
spleen
NS
Harvest SIINFEKL
blood Staining
Thrombin

Events
OVA
activation
Activated
and
platelets NS
Isolate centrifugation
CFSE
platelets SIINFEKL
OVA
Lymphoproliferation
PEVs

B Unpulsed DC+PP PLT+PP PLT+OVA

300 0 300 300 300

3
200 200 2 200 200 3

100 100 100 4 100 2 0


4
1 0
1 43 10 5 1
2 2
1 102 103 104 105 101 102 103 104 105 101 102 103 104 105 101 102 103 104 105
10

PEV+PP PEV+OVA
0
300 0 300
Count

200 200

1
100 100 2
43
1 5
CFSE-FITC 5 4 32
1
10 102 103 104 105 101 102 103 104 105

C D E
90 **** *** ** *** ** 90 ** ** ** 90 ** 90
S

S
N

N
% of lymphoproliferative cells

% of lymphoproliferative cells

% of lymphoproliferative cells
(based on CD3+ CD8+ cells)

(based on CD3+ CD8+ cells)

(based on CD3+ CD8+ cells)

75 75 75 75

60 60 60 60

45 45 45 45

30 30 30 30

15 15 15 15

0 0 0 0
Pulsed – + – + OVA – + OVA Pulsed + – + OVA VA VA PP PP
O O + +
DC Activated PLTs PEVs DC PEV depleted sup + )+ C o)
Vs xo
D ox
PE o ep
s( ep C(
V D
PE

Figure 7. PEVs loaded with native OVA process and present OVA peptide to induce antigen-specific T-cell lymphoproliferation (A) The experimental plan. NS, unpulsed.
(B) Histogram showing CFSE fluorescence shift of CD31CD81 T-cell populations when coincubated with DCs, activated platelets (PLTs), or PEVs left unpulsed or pulsed with
SIINFEKL peptide (PP) or OVA for 7 days. (C) Percentage of CD31CD81 lymphoproliferative cells after coincubation with DCs, PLTs, or PEVs, unpulsed or pulsed with PP or OVA for
7 days. Data are the mean 6 standard error of the mean (SEM); n 5 14). **P , .01; ***P , .001; ****P , .0001 vs unpulsed (Friedman test followed by Dunn’s post hoc test for
multiple comparisons). NS, nonsignificant. (D) Percentage of CD31CD81 lymphoproliferative cells after a 7-day coincubation with PP pulsed DCs or supernatant (surn), depleted of
PEVs by ultracentrifugation, left unpulsed or pulsed with PP or OVA. Data are the mean 6 SEM (n 5 5). **P , .01 vs unpulsed (Mann-Whitney). (E) Proportion of CD31CD81
lymphoproliferative cells after a 7-day coincubation with OVA-pulsed PEVs, treated or not with epoxomicin (epoxo) for 2 hours, and PP-pulsed DCs (DC1PP), treated or not with
epoxomicin (epoxo). Dashed lines are unstimulated conditions. Data are the mean 6 SEM (n 5 9 for PEVs and n 5 3 for DC). **P , .01 (Wilcoxon). NS, nonsignificant.

2616 blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 MARCOUX et al


division and were coincubated for 5 days with DCs, activated core is present and active within EVs derived from apoptotic
platelets, or PEVs, which were either pulsed or not with SIINFEKL endothelial cells and regulates the formation of tertiary
or native OVA (Figure 7A). Lymphoproliferation is represented by lymphoid structure, production of autoantibodies, and graft
a decrease in the mean fluorescence intensity histogram (ie, a rejection after transplantation.12 Although some evidence
dilution of CFSE fluorescence; Figure 7B). supports that a circulating extracellular proteasome may be
transported by EVs, we showed that EVs of platelet origin,
As expected, we found that the proportion of lymphoproliferative among the most abundant EVs in blood, do contain the
cells was significantly higher when OT-1 T lymphocytes were proteasome. We further suggest, based on our characterization
incubated with peptide- or native OVA-pulsed DCs or activated of these EVs, that platelet microvesicles, not exosomes, contain
platelets (Figure 7B-C). Of particular note, PEVs also supported the proteasome. Consistent with this, mass spectrometry
T-cell proliferation when pulsed with either the SIINFEKL or native analysis of the human PEV proteome identified numerous
OVA (Figure 7B-C). In addition, when PEVs present in the platelet proteasomal subunits.67-69 These include subunits of the 20S
secretome were removed from pulsed conditions by ultracentri- catalytic core and immunoproteasome subunit (PSMB8),
fugation, no proliferation was observed, confirming that the subunits of the 11S and 19S regulator, and the 26S protea-
pulsed proteins alone, or the platelet secretome devoid of PEVs, some.67-69 Moreover, with calnexin, calreticulin, ERP57, and
cannot support proliferation (Figure 7D). Furthermore, inhibition ERP29, other members of the ubiquitin-proteasome pathway
of the PEV proteasome by epoxomicin before pulsing with native were identified in PEVs, such as members of the E1 and E2
OVA inhibited the ability of PEVs to induce T-cell proliferation ubiquitin-conjugating enzyme family.67,69 The presence of
(Figure 7E, left). The effect was directed toward the PEV these proteins and the fact that intact OVA must be
proteasome, as addition of epoxomicin before peptide pulsing ubiquitinated for degradation by the proteasome70 point to
at the same concentration used on DCs did not inhibit prolifer- the occurrence of functional ubiquitination in PEVs. To our
ation (Figure 7E, right). Thus, PEVs are capable of proteosome- knowledge, there is no evidence of protein TAP-1 and -2
dependent processing of native proteins, thereby enabling (related to TAP transporter) presence in PEVs. Further inves-
peptide loading onto MHC-I. PEVs express costimulatory tigations are needed to see whether the TAP transporter is
molecules, and their interaction with T lymphocytes promotes present in PEVs and/or whether the processing pathway of the
lymphocyte cytokine production and proliferation. antigen differs in extracellular vesicles, as there is no reported
endoplasmic reticulum in PEVs. Thus, although proteomic data
point to ubiquitin-proteasome system proteins in PEVs, the
Discussion present work unequivocally demonstrates its presence and
Megakaryocytes and platelets are emerging as active players in documents that the extracellular proteasome in PEVs is func-
innate and adaptive immunity.7,30,31 The platelet’s role in immu- tional and can contribute to antigen processing.
nity is mainly confined to the blood circulation, whereas
megakaryocytes are localized in bone marrow and lungs. The We used complementary approaches and developed an
latter location potentially provides the megakaryocyte with more hs-FCM–based assay to detect active proteasome at the single
direct access to airborne pathogens and allergens.32,62,63 In EV level, thereby permitting quantification and assessment of
contrast, PEVs can disseminate into organs and tissues, and this other molecules expressed by the EVs. In particular, the
dissemination may be due to their small dimensions and the proteasome-containing PEVs also expressed MHC-I and cos-
presence of unique surface molecules. In this study, we found that timulatory molecules, which enabled lymphocyte activation/
the proteasome and the necessary machinery to process and proliferation and cytokine generation. These findings demon-
present antigens to CD81 T cells are packaged into PEVs by strate a novel and potentially important role for PEVs in adaptive
platelets. Thus, PEVs may extend the immune functions played by immunity. Although our work suggests that PEVs are involved in
platelets and megakaryocytes outside the confines of the blood. adaptive immunity through antigen presentation, the findings
do not necessarily exclude that other cells release proteasome-
PEVs are heterogeneous in terms of surface molecules and their containing EVs capable of playing this role. Indeed, EVs derived
platelet-derived content. The presence of mitochondria within from DCs, B and T lymphocytes, macrophages, and NK cells can
PEVs is well documented,36,37,64 but it was unknown whether perform crosspresentation, suggesting that they also contain
other organelles were also transferred from the platelet. The the necessary antigen-processing machinery.71-76 Further stud-
proteasome is much more abundant than mitochondria, at ies are needed to determine the impact and the importance of
800 000 copies per cell,65 in contrast to 3 to 7 mitochondria PEVs as antigen-presenting elements.
per platelet.37 Further investigation is necessary to determine
whether the presence of multiple organelles within a single We identified proteasome-containing PEVs in the blood of healthy
vesicle is the result of a specific sorting mechanism or the large donors. As most PEVs in blood under healthy conditions are
size and storage capacity of the vesicles. Nonetheless, we suggested to originate from megakaryocytes,77,78 the latter may
observed that the release of proteasome-containing PEVs also constitutively release proteasome-containing EVs. Moreover,
requires cytoskeleton remodeling via intact actin microfilament we found that numerous stimuli of human platelets, as well as in
dynamics and that a broad array of platelet agonists induce the vivo stimulation of mouse platelets, induce release of protea-
release of proteasome-containing PEVs.37 somes in PEVs, suggesting that proteasome release is at least
conserved in both humans and mice and takes place via platelet
The presence of an extracellular proteasome has already been activation. Furthermore, platelets can actively induce immunity
documented in healthy human blood, and elevated levels have against the Plasmodium berghei parasite,26 and megakaryocytes
been found in patients who have autoimmune diseases, develop can be infected by dengue virus79 and can also phagocytose
sepsis, or experience trauma.66 Moreover, the 20S proteasome Escherichia coli.32 Given their small size, intact microorganisms

ACTIVE PROTEASOME IN EXTRACELLULAR VESCICLES blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 2617
may not be present inside PEVs, but PEVs may process cytosolic Acknowledgments
microbial proteins derived from intact platelets/megakaryocytes The authors thank the blood donors and patients who participated in
that lack the ability to enter the lymphatic system. Thus, PEVs may the study; Nicolas Tessandier and Carolanne G elinas for general
be implicated in immune surveillance and may contribute to technical assistance; and Julie-Christine L
evesque from the Cytometry
presentation of microbial antigens within lymph tissues. Further and Microscopy platform (Centre Hospitalier Universitaire [CHU] de
studies are needed to determine whether exposure to PEVs Quebec), and Richard Janvier from the Microscopy platform (Universit
e
Laval) for microscopy assistance.
suffices to establish immunity in vivo, such as to less immunodo-
minant antigens than OVA, or whether costimulation by inflam- This work was supported by the Natural Sciences and Engineering
mation or infection is needed to establish sustained immune Research Council (E.B.). E.B. is the recipient of a senior award from the
response. Fonds de Recherche du Qu ebec-Sant e (FRQS). This work was also

supported by the Etablissement Français du Sang (EFS), INSERM, and
Self-antigens such as those from mitochondria were identified Universit
e Paris-Est (B.V.) and the Swedish Research Council (Veten-
in a proportion of the proteasome-containing PEVs. Although skapsrådet, 2017-01779) (J.W.S.). G.M. received an award from the
Canadian Blood Services and from Mitacs and a postdoctoral fellow-
prior studies showed that mitochondria-containing PEVs are
ship from FRQS. A.Z. was the recipient of a postdoctoral fellowship
rare in lymph (0.41% 6 0.25% [n 5 4] of the PEVs in mouse from Swiss National Science Foundation. A.L. has an award from the
lymph contain mitochondria)41 in comparison to proteasome- fonds Pierre Borgeat and from The Arthritis Society (TAS). M.B. is the
containing PEVs 13.61% 6 4.27% (n 5 6), these proportions recipient of a postdoctoral fellowship from TAS.
may be augmented in certain diseases. It would be interesting
to determine whether PEVs contribute to the formation of
mitochondrial autoantibodies that are described in autoim-
mune diseases, such as systemic lupus erythematosus.80 Authorship
Furthermore, the presence of proteasome-containing PEVs in Contribution: E.B., B.V., G.M., and A.Z., conceived and designed the study
and wrote the manuscript; G.M., A.L., S.H., M.B., M.M., I.A., M.T., and B.V.
platelet concentrates was not associated with increased risks of
performed the experiments, analyzed the data, and participated in
ATR or TRALI in a mouse model. It remains to be verified manuscript preparation; T.L., J.R., A.K.-R., J.T., H.H.-C., F.C., R.K., J.W.S.,
whether the presentation of platelet antigens (eg, CD41 or M.-J.H., S.G.B., F.P., H.S.O., B.I.F., and M.D. performed experiments,
CD61) by PEVs from PCs contribute to generation of contributed critical biospecimens and analyzed the data; and all authors
antiplatelet immunity in transfused recipients, although this read and approved the manuscript.
has been shown with megakaryocytes.56 It also cannot be
excluded that PEVs participate in other immune responses, Conflict-of-interest disclosure: The authors declare no competing financial
interests.
such as autoantibody production or in tissue remodeling,
or that they could be used as a platform for cell-based
The current affiliation for G.M. is Division of Hematology and Transfusion
vaccines.81-83 The crosspresentation of PEVs presented herein Medicine, Lund University, Lund, Sweden.
may allow for new therapeutic possibilities, such as in antitumor
or antiviral immunity or inducing cytotoxic immunity by vacci- ORCID profiles: S.H., 0000-0003-0859-3990; I.A., 0000-0003-4697-
nation.84 For example, PEVs have already been proposed as 046X; J.R., 0000-0003-1673-0684; A.K.-R., 0000-0002-4499-089X;
antigen carriers for vaccination,85,86 and our results suggest S.G.B., 0000-0001-9779-0368; H.H.-C., 0000-0003-1462-3893; F.C.,
that these types of PEVs are also endowed with cross-priming 0000-0001-8041-928X; R.K., 0000-0002-1608-876X; J.W.S., 0000-0002-
properties that offer new prophylactic or therapeutic 1510-0077; B.I.F., 0000-0001-7114-2266; M.D., 0000-0002-9300-4232;
E.B., 0000-0001-6319-6432.
vaccination.
Correspondence: Eric Boilard, Centre de Recherche du Centre Hospitalier
Human platelets injected into WT mice circulate ,2 hours, in Universitaire de Qu
ebec, Faculte de Medecine de l’Universite Laval, 2705
contrast to mouse platelets transfused into mice that can Laurier Blvd, Room T1-49, Qu ebec, QC G1V 4G2, Canada; e-mail: eric.
circulate for several days.87,88 We thus used mouse PEVs in our 
boilard@crchudequebec.ulaval.ca; and Beno^ıt Vingert, Etablissement
transfusion experiments, and yet most were undetectable from Français du Sang, Institut Mondor de Recherche Biom edicale, INSERM
the blood circulation after 15 minutes, pointing to their rapid U955, 5 rue Gustave Eiffel, 94017 Creteil, France; e-mail: benoit.vingert@
efs.sante.fr.
uptake in surrounding tissues. In blood, the main absolute
cellular target was the platelets, mostly because platelets
outnumber leukocytes, which may suggest that PEVs recycle
molecules back to platelets. PEVs were also found in bone
Footnotes
Submitted 12 November 2020; accepted 12 July 2021; prepublished
marrow, consistent with recent findings that pointed to their
online on Blood First Edition 22 July 2021. DOI 10.1182/
role in the stimulation of megakaryocyte biogenesis.43 The blood.2020009957.
main organs that were targeted were the lymphoid organs. Our
findings in mouse lymph revealed that proteasome-containing Original data are available by e-mail request to either corresponding
PEVs can circulate in the lymphatic system, potentially explain- author.
ing their accumulation in lymphoid organs after IV injection.
This access to the lymphatic system by proteasome-containing The online version of this article contains a data supplement.
PEVs may reveal a new immune route for PEVs to reach
There is a Blood Commentary on this article in this issue.
lymphoid organs or infected tissues. Our study highlights the
diversity of PEVs and supports the concept that different The publication costs of this article were defrayed in part by page charge
subtypes of PEVs may play different roles, depending on their payment. Therefore, and solely to indicate this fact, this article is hereby
cargo and tissue distribution. marked “advertisement” in accordance with 18 USC section 1734.

2618 blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 MARCOUX et al


REFERENCES 17. Nayak MK, Kumar K, Dash D. Regulation of 32. Pariser DN, Hilt ZT, Ture SK, et al. Lung
1. Davı G, Patrono C. Platelet activation and proteasome activity in activated human megakaryocytes are immune modulatory
atherothrombosis. N Engl J Med. 2007; platelets. Cell Calcium. 2011;49(4):226-232. cells. J Clin Invest. 2021;131(1):137377.
357(24):2482-2494.
18. Grundler Groterhorst K, Mannell H, Pircher J, 33. Melki I, Tessandier N, Zufferey A, Boilard E.
2. Ribeiro LS, Migliari Branco L, Franklin BS. Kraemer BF. Platelet Proteasome Activity and Platelet microvesicles in health and disease.
Regulation of Innate Immune Responses by Metabolism Is Upregulated during Bacterial Platelets. 2017;28(3):214-221.
Platelets. Front Immunol. 2019;10:1320. Sepsis. Int J Mol Sci. 2019;20(23):E5961.
34. Puhm F, Boilard E, Machlus KR. Platelet
3. Rayes J, Bourne JH, Brill A, Watson SP. The 19. Gupta N, Li W, Willard B, Silverstein RL, Extracellular Vesicles: Beyond the Blood.
dual role of platelet-innate immune cell McIntyre TM. Proteasome proteolysis Arterioscler Thromb Vasc Biol. 2021;41(1)
interactions in thrombo-inflammation. Res supports stimulated platelet function and 87-96.
Pract Thromb Haemost. 2019;4(1):23-35. thrombosis. Arterioscler Thromb Vasc Biol.
2014;34(1):160-168. 35. Siljander PR. Platelet-derived microparticles -
4. Semple JW, Italiano JE Jr, Freedman J. an updated perspective. Thromb Res. 2011;
20. Colberg L, Cammann C, Greinacher A, Seifert 127(suppl 2):S30-S33.
Platelets and the immune continuum. Nat Rev
Immunol. 2011;11(4):264-274. U. Structure and function of the ubiquitin-
proteasome system in platelets. J Thromb 36. Marcoux G, Magron A, Sut C, et al. Platelet-
derived extracellular vesicles convey
5. Morrell CN, Aggrey AA, Chapman LM, Haemost. 2020;18(4):771-780.
mitochondrial DAMPs in platelet
Modjeski KL. Emerging roles for platelets
21. Yukawa M, Sakon M, Kambayashi J, et al. concentrates and their levels are associated
as immune and inflammatory cells. with adverse reactions. Transfusion. 2019;
Purification and characterization of
Blood. 2014;123(18): 59(7):2403-2414.
endogenous protein activator of human
2759-2767.
platelet proteasome. J Biochem. 1993;114(3):
37. Boudreau LH, Duchez AC, Cloutier N, et al.
6. Kapur R, Zufferey A, Boilard E, Semple JW. 317-323.
Platelets release mitochondria serving as
Nouvelle cuisine: platelets served with substrate for bactericidal group IIA-secreted
22. Yukawa M, Sakon M, Kambayashi J, et al.
inflammation. J Immunol. 2015;194(12): Proteasome and its novel endogeneous phospholipase A2 to promote inflammation.
5579-5587. activator in human platelets. Biochem Blood. 2014;124(14):2173-2183.
Biophys Res Commun. 1991;178(1):256-262.
7. Cunin P, Nigrovic PA. Megakaryocytes as 38. Burnouf T, Chou ML, Goubran H, Cognasse F,
immune cells. J Leukoc Biol. 2019;105(6): 23. Boegel S, L€
ower M, Bukur T, Sorn P, Castle Garraud O, Seghatchian J. An overview of the
1111-1121. JC, Sahin U. HLA and proteasome expression role of microparticles/microvesicles in blood
components: Are they clinically beneficial or
body map. BMC Med Genomics. 2018;11(1):
8. Garraud O, Cognasse F. Are Platelets Cells? harmful? Transfus Apheresis Sci. 2015;53(2):
36.
And if Yes, are They Immune Cells? Front 137-145.
Immunol. 2015;6:70. 24. Semple JW, Speck ER, Milev YP, Blanchette V,
39. Boilard E, Nigrovic PA, Larabee K, et al.
Freedman J. Indirect allorecognition of
9. Opperman CM, Sishi BJ. Tumor necrosis Platelets amplify inflammation in arthritis via
platelets by T helper cells during platelet
factor alpha stimulates p62 accumulation and collagen-dependent microparticle produc-
transfusions correlates with anti-major histo-
enhances proteasome activity independently tion. Science. 2010;327(5965):580-583.
compatibility complex antibody and cytotoxic
of ROS. Cell Biol Toxicol. 2015;31(2):83-94. T lymphocyte formation. Blood. 1995;86(2): 40. Gy€orgy B, Szabo TG, Turi
ak L, et al. Improved
805-812. flow cytometric assessment reveals distinct
10. Tanaka K. The proteasome: overview of
structure and functions. Proc Jpn Acad, Ser B, microvesicle (cell-derived microparticle)
25. Zufferey A, Speck ER, Machlus KR, et al.
signatures in joint diseases. PLoS One. 2012;
Phys Biol Sci. 2009;85(1):12-36. Mature murine megakaryocytes present
7(11):e49726.
antigen-MHC class I molecules to T cells and
11. Klockenbusch C, Walsh GM, Brown LM, et al. transfer them to platelets. Blood Adv. 2017; 41. Tessandier N, Melki I, Cloutier N, et al.
Global proteome analysis identifies active 1(20):1773-1785. Platelets Disseminate Extracellular Vesicles in
immunoproteasome subunits in human Lymph in Rheumatoid Arthritis. Arterioscler
platelets. Mol Cell Proteomics. 2014;13(12): 26. Chapman LM, Aggrey AA, Field DJ, et al. Thromb Vasc Biol. 2020;40(4):929-942.
3308-3319. Platelets present antigen in the context of
MHC class I. J Immunol. 2012;189(2):916-923. 42. Milasan A, Tessandier N, Tan S, Brisson A,
12. Dieude M, Bell C, Turgeon J, et al. The 20S Boilard E, Martel C. Extracellular vesicles are
proteasome core, active within apoptotic 27. Zufferey A, Schvartz D, Nolli S, Reny JL, present in mouse lymph and their level differs
exosome-like vesicles, induces autoantibody Sanchez JC, Fontana P. Characterization in atherosclerosis. J Extracell Vesicles. 2016;
production and accelerates rejection. Sci of the platelet granule proteome: 5(1):31427.
Transl Med. 2015;7(318):318ra200. evidence of the presence of MHC1 in
alpha-granules. J Proteomics. 2014;101: 43. French SL, Butov KR, Allaeys I, et al. Platelet-
13. Shi DS, Smith MC, Campbell RA, et al. 130-140. derived extracellular vesicles infiltrate and
Proteasome function is required for platelet modify the bone marrow during
production. J Clin Invest. 2014;124(9): 28. Iannacone M, Sitia G, Isogawa M, et al. inflammation. Blood Adv. 2020;4(13):3011-
Platelets mediate cytotoxic T lymphocyte- 3023.
3757-3766.
induced liver damage. Nat Med. 2005;11(11):
14. Murai K, Kowata S, Shimoyama T, et al. 1167-1169. 44. Sprague DL, Elzey BD, Crist SA, Waldschmidt
Bortezomib induces thrombocytopenia by the TJ, Jensen RJ, Ratliff TL. Platelet-mediated
29. Verschoor A, Neuenhahn M, Navarini AA, et modulation of adaptive immunity: unique
inhibition of proplatelet formation of
al. A platelet-mediated system for shuttling delivery of CD154 signal by platelet-derived
megakaryocytes. Eur J Haematol. 2014;93(4):
blood-borne bacteria to CD8a1 dendritic membrane vesicles. Blood. 2008;111(10):
290-296. cells depends on glycoprotein GPIb and 5028-5036.
complement C3. Nat Immunol. 2011;12(12):
15. Nayak MK, Kulkarni PP, Dash D. Regulatory
1194-1201. 45. Yari F, Motefaker M, Nikougoftar M, Khayati
role of proteasome in determination of Z. Interaction of Platelet-Derived Micropar-
platelet life span. J Biol Chem. 2013;288(10): 30. Maouia A, Rebetz J, Kapur R, Semple JW. The ticles with a Human B-Lymphoblast Cell Line:
6826-6834. Immune Nature of Platelets Revisited. A Clue for the Immunologic Function of the
Transfus Med Rev. 2020;34(4):209-220. Microparticles. Transfus Med Hemother.
16. Grundler K, Rotter R, Tilley S, et al. The
2018;45(1):55-61.
proteasome regulates collagen-induced 31. Marcoux G, Laroche A, Espinoza Romero J,
platelet aggregation via nuclear-factor- Boilard E. Role of platelets and 46. Sadallah S, Amicarella F, Eken C, Iezzi G,
kappa-B (NFŒB) activation. Thromb Res. 2016; megakaryocytes in adaptive immunity. Schifferli JA. Ectosomes released by platelets
148:15-22. Platelets. 2021;32:340-351. induce differentiation of CD41T cells into T

ACTIVE PROTEASOME IN EXTRACELLULAR VESCICLES blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 2619
regulatory cells. Thromb Haemost. 2014; Immune Sensors and Interactors. Front
60. Rand ML, Wang H, Bang KW, Packham MA,
112(6):1219-1229. Immunol. 2020;11:262.
Freedman J. Rapid clearance of procoagulant
47. Dinkla S, van Cranenbroek B, van der Heijden platelet-derived microparticles from the cir- 75. Zeng F, Morelli AE. Extracellular vesicle-
WA, et al. Platelet microparticles inhibit IL-17 culation of rabbits. J Thromb Haemost. 2006; mediated MHC cross-dressing in immune
production by regulatory T cells through P- 4(7):1621-1623. homeostasis, transplantation, infectious dis-
selectin. Blood. 2016;127(16):1976-1986. eases, and cancer. Semin Immunopathol.
61. Rank A, Nieuwland R, Crispin A, et al.
2018;40(5):477-490.
48. Verdoes M, Florea BI, Menendez-Benito V, et Clearance of platelet microparticles in vivo.
Platelets. 2011;22(2):111-116. 76. Noulsri E. Effects of Cell-Derived Micropar-
al. A fluorescent broad-spectrum proteasome
inhibitor for labeling proteasomes in vitro and ticles on Immune Cells and Potential Impli-
62. Lefrançais E, Ortiz-Mu~noz G, Caudrillier A,
in vivo. Chem Biol. 2006;13(11):1217-1226. cations in Clinical Medicine. Lab Med. 2021;
et al. The lung is a site of platelet biogenesis 52(2):122-135.
and a reservoir for haematopoietic progenitors.
49. Raz V, Raz Y, Paniagua-Soriano G, et al.
Nature. 2017;544(7648):105-109. 77. Flaumenhaft R, Dilks JR, Richardson J, et al.
Proteasomal activity-based probes mark pro-
tein homeostasis in muscles. J Cachexia Megakaryocyte-derived microparticles: direct
63. Campbell RA, Schwertz H, Hottz ED, et al. visualization and distinction from platelet-
Sarcopenia Muscle. 2017;8(5):798-807. Human megakaryocytes possess intrinsic derived microparticles. Blood. 2009;113(5):
antiviral immunity through regulated 1112-1121.
50. Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ,
induction of IFITM3. Blood. 2019;133(19):
Sixma JJ. Activated platelets release two
2013-2026. 78. Gitz E, Pollitt AY, Gitz-Francois JJ, et al.
types of membrane vesicles: microvesicles
CLEC-2 expression is maintained on activated
by surface shedding and exosomes derived 64. Marcoux G, Duchez AC, Rousseau M, et al. platelets and on platelet microparticles.
from exocytosis of multivesicular bodies and Microparticle and mitochondrial release Blood. 2014;124(14):2262-2270.
alpha-granules. Blood. 1999;94(11): during extended storage of different types of
3791-3799. platelet concentrates. Platelets. 2017;28(3): 79. Vogt MB, Lahon A, Arya RP, Spencer Clinton
272-280. JL, Rico-Hesse R. Dengue viruses infect
51. Semple JW, Rebetz J, Kapur R. Transfusion- human megakaryocytes, with probable
associated circulatory overload and 65. Princiotta MF, Finzi D, Qian SB, et al. clinical consequences. PLoS Negl Trop Dis.
transfusion-related acute lung injury. Blood. Quantitating protein synthesis, degradation, 2019;13(11):e0007837.
2019;133(17):1840-1853. and endogenous antigen processing.
Immunity. 2003;18(3):343-354. 80. Becker Y, Marcoux G, Allaeys I, et al.
52. Kapur R, Kim M, Rebetz J, et al. Autoantibodies in Systemic Lupus
Gastrointestinal microbiota contributes to the 66. Sixt SU, Dahlmann B. Extracellular, circulating Erythematosus Target Mitochondrial RNA.
development of murine transfusion-related proteasomes and ubiquitin - incidence and Front Immunol. 2019;10:1026.
acute lung injury. Blood Adv. 2018;2(13): relevance. Biochim Biophys Acta. 2008;
1651-1663. 1782(12):817-823. 81. Słomka A, Urban SK, Lukacs-Kornek V,
_
Zekanowska E, Kornek M. Large Extracellular
53. Kapur R, Kim M, Aslam R, et al. T regulatory 67. Garcia BA, Smalley DM, Cho H, Shabanowitz Vesicles: Have We Found the Holy Grail of
cells and dendritic cells protect against J, Ley K, Hunt DF. The platelet microparticle Inflammation? Front Immunol. 2018;9:2723.
transfusion-related acute lung injury via IL-10. proteome. J Proteome Res. 2005;4(5):
Blood. 2017;129(18):2557-2569. 1516-1521. 82. de Jong OG, Kooijmans SAA, Murphy DE, et
al. Drug Delivery with Extracellular Vesicles:
54. McKenzie SE, Taylor SM, Malladi P, et al. The 68. Dean WL, Lee MJ, Cummins TD, Schultz DJ, From Imagination to Innovation. Acc Chem
role of the human Fc receptor Fc gamma RIIA Powell DW. Proteomic and functional Res. 2019;52(7):1761-1770.
in the immune clearance of platelets: a characterisation of platelet microparticle size
transgenic mouse model. J Immunol. 1999; classes. Thromb Haemost. 2009;102(4): 83. Urbanelli L, Buratta S, Tancini B, et al. The
162(7):4311-4318. 711-718. Role of Extracellular Vesicles in Viral Infection
and Transmission. Vaccines (Basel). 2019;7(3):
55. Cloutier N, Allaeys I, Marcoux G, et al. 69. Capriotti AL, Caruso G, Cavaliere C, 102.
Platelets release pathogenic serotonin and Piovesana S, Samperi R, Lagana A. Proteomic
return to circulation after immune complex- characterization of human platelet-derived 84. Jhunjhunwala S, Hammer C, Delamarre L.
mediated sequestration. Proc Natl Acad Sci microparticles. Anal Chim Acta. 2013;776: Antigen presentation in cancer: insights into
57-63. tumour immunogenicity and immune evasion.
USA. 2018;115(7):E1550-E1559.
Nat Rev Cancer. 2021;21(5):298-312.
56. Melki I, Allaeys I, Tessandier N, et al. Platelets 70. Benaroudj N, Tarcsa E, Cascio P, Goldberg
AL. The unfolding of substrates and ubiquitin- 85. Sabanovic B, Piva F, Cecati M, Giulietti M.
release mitochondrial antigens in systemic
independent protein degradation by protea- Promising Extracellular Vesicle-Based Vac-
lupus erythematosus. Sci Transl Med. 2021;
somes. Biochimie. 2001;83(3-4):311-318. cines against Viruses, Including SARS-CoV-2.
13(581):eaav5928.
Biology (Basel). 2021;10(2):94.
71. Chen Z, Larregina AT, Morelli AE. Impact of
57. Angenieux C, Dupuis A, Gachet C, de la Salle
extracellular vesicles on innate immunity. Curr 86. Bliss CM, Parsons AJ, Nachbagauer R, et al.
H, Ma^ıtre B. Cell surface expression of HLA I
Opin Organ Transplant. 2019;24(6):670-678. Targeting Antigen to the Surface of EVs
molecules as a marker of young platelets. J Improves the In Vivo Immunogenicity of
Thromb Haemost. 2019;17(9):1511-1521. 72. Lindenbergh MFS, Stoorvogel W. Antigen Human and Non-human Adenoviral Vaccines
Presentation by Extracellular Vesicles from in Mice. Mol Ther Methods Clin Dev. 2020;16:
58. N ~ ez-Avellaneda D, Mosso-Pani MA,
un
Professional Antigen-Presenting Cells. Annu 108-125.
S
anchez-Torres LE, Castro-Mussot ME,
Rev Immunol. 2018;36(1):435-459.
Corona-de la Pe~na NA, Salazar MI. Dengue 87. Baker GR, Sullam PM, Levin J. A simple,
Virus Induces the Release of sCD40L and 73. Lindenbergh MFS, Wubbolts R, Borg EGF, fluorescent method to internally label
Changes in Levels of Membranal CD42b and van ’t Veld EM, Boes M, Stoorvogel W. platelets suitable for physiological
CD40L Molecules in Human Platelets. Viruses. Dendritic cells release exosomes together measurements. Am J Hematol. 1997;56(1):
2018;10(7):357. with phagocytosed pathogen; potential 17-25.
implications for the role of exosomes in
59. Porgador A, Yewdell JW, Deng Y, Bennink JR, antigen presentation. J Extracell Vesicles. 88. Blessinger SA, Tran JQ, Jackman RP, et al.
Germain RN. Localization, quantitation, and in 2020;9(1):1798606. Immunodeficient mice are better for
situ detection of specific peptide-MHC class I modeling the transfusion of human blood
complexes using a monoclonal antibody. 74. Federici C, Shahaj E, Cecchetti S, et al. components than wild-type mice. PLoS One.
Immunity. 1997;6(6):715-726. Natural-Killer-Derived Extracellular Vesicles: 2020;15(7):e0237106.

2620 blood® 23 DECEMBER 2021 | VOLUME 138, NUMBER 25 MARCOUX et al

View publication stats

You might also like