You are on page 1of 14

Research Article

Hepatic and Biliary Cancer

Patient pIgR-enriched extracellular vesicles drive cancer


stemness, tumorigenesis and metastasis in
hepatocellular carcinoma

Graphical abstract Authors

Sze Keong Tey, Samuel Wan Ki Wong,


Janice Yuen Tung Chan, ., Yi Gao,
Hepatocellular Anti-pIgR antibody Jing Ping Yun, Judy Wai Ping Yam
carcinoma (HCC)

Correspondence
Cyto D judyyam@pathology.hku.hk
Self-renewal
EV-pIgR
(J.W.P. Yam).
P PDK1
Lay summary
P
Akt1
Liver Tumorigenesis The World Health Organization es-
β-catenin timates that more than 1 million
P GSK3β
patients will die from liver cancer,
mostly hepatocellular carcinoma
(HCC), in 2030. Understanding the
Metastasis
underlying mechanism by which
HCC acquires aggressive attributes
is crucial to improving the diag-
nosis and treatment of patients.
Highlights Herein, we demonstrated that
 Elevated pIgR levels are found in circulating extracellular vesicles nanometer-sized extracellular vesi-
from patients with liver cancer. cles released by tumors promote
cancer stemness and tumorigen-
 EV-pIgR promotes cancer stemness and cancerous phenotypes in
esis. Within these oncogenic vesi-
recipient liver cancer cells.
cles, we identified a key component
 EV-pIgR promotes liver cancer cell aggressiveness by activating the that functions as a potent modu-
PDK1/Akt/GSK3b/b-catenin signaling axis. lator of cancer aggressiveness. By
inhibiting this functional compo-
 Combined treatment using sorafenib and anti-pIgR antibody atten-
nent of EVs using a neutralizing
uates growth of patient-derived xenografts in mice.
antibody, tumor growth was pro-
foundly attenuated in mice. This
hints at a potentially effective
therapeutic alternative for patients
with cancer.

https://doi.org/10.1016/j.jhep.2021.12.005
© 2021 The Author(s). Published by Elsevier B.V. on behalf of European Association for the Study of the Liver. This is an open access article
under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). J. Hepatol. 2022, 76, 883–895
Research Article
Hepatic and Biliary Cancer

Patient pIgR-enriched extracellular vesicles drive cancer stemness,


tumorigenesis and metastasis in hepatocellular carcinoma
Sze Keong Tey1,†, Samuel Wan Ki Wong1,†, Janice Yuen Tung Chan1, Xiaowen Mao1,
Tung Him Ng1, Cherlie Lot Sum Yeung1, Zoe Leung1, Hiu Ling Fung1, Alexander Hin Ning Tang1,
Danny Ka Ho Wong2,3, Lung-Yi Mak2,3, Man-Fung Yuen2,3, Chun-Fung Sin1, Irene Oi-Lin Ng1,3,
Stephanie Kwai Yee Ma3,4, Terence Kin Wah Lee5, Peihua Cao6,7, Kebo Zhong7, Yi Gao7,
Jing Ping Yun8, Judy Wai Ping Yam1,3,*
1
Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; 2Department of Medicine, Li Ka Shing
Faculty of Medicine, The University of Hong Kong, Hong Kong; 3State Key Laboratory of Liver Research (The University of Hong Kong), Hong
Kong; 4School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; 5Department of Applied
Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong; 6Clinical Research Center, Zhujiang Hospital, Southern
Medical University, Guangzhou, Guangdong, PR China; 7Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical
University, Guangzhou, Guangdong, PR China; 8Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong,
PR China

See Editorial, pages 768–770

Background & Aims: Extracellular vesicles (EVs) play a pivotal catenin signaling axis. Furthermore, an anti-pIgR neutralizing
role in connecting tumor cells with their local and distant mi- antibody attenuated tumor growth in mice implanted with PDTXs.
croenvironments. Herein, we aimed to understand the role (on a Conclusions: This study illustrates a previously unknown role of
molecular basis) patient-derived EVs play in modulating cancer EV-pIgR in regulating cancer stemness and aggressiveness: EV-
stemness and tumorigenesis in the context of hepatocellular pIgR activates PDK1/Akt/GSK3b/b-catenin signaling cascades.
carcinoma (HCC). The blockade of the intercellular communication mediated by
Methods: EVs from patient sera were isolated, quantified and EV-pIgR in the tumor microenvironment may provide a new
characterized. The EVs were vigorously tested, both in vitro and therapeutic strategy for patients with cancer.
in vivo, through various functional assays. Proteomic analysis was Lay summary: The World Health Organization estimates that
performed to identify the functional components of EVs. The more than 1 million patients will die from liver cancer, mostly
presence and level of polymeric immunoglobulin receptor (pIgR) hepatocellular carcinoma (HCC), in 2030. Understanding the
in circulating EVs and tumor and non-tumorous tissues of pa- underlying mechanism by which HCC acquires aggressive attri-
tients with HCC were determined by ELISA, immunoblotting, butes is crucial to improving the diagnosis and treatment of
immunohistochemistry and quantitative PCR. The functional role patients. Herein, we demonstrated that nanometer-sized extra-
and underlying mechanism of EVs with enhanced pIgR expres- cellular vesicles released by tumors promote cancer stemness
sion were elucidated. Blockade of EV-pIgR with neutralizing and tumorigenesis. Within these oncogenic vesicles, we identi-
antibody was performed in nude mice implanted with patient- fied a key component that functions as a potent modulator of
derived tumor xenografts (PDTXs). cancer aggressiveness. By inhibiting this functional component
Results: Circulating EVs from patients with late-stage HCC (L-HCC) of EVs using a neutralizing antibody, tumor growth was pro-
had significantly elevated pIgR expression compared to the EVs foundly attenuated in mice. This hints at a potentially effective
released by control individuals. The augmenting effect of L–HCC– therapeutic alternative for patients with cancer.
EVs on cancer stemness and tumorigenesis was hindered by an © 2021 The Author(s). Published by Elsevier B.V. on behalf of Euro-
anti-pIgR antibody. EVs enriched with pIgR consistently promoted pean Association for the Study of the Liver. This is an open access
cancer stemness and cancerous phenotypes in recipient cells. article under the CC BY-NC-ND license (http://creativecommons.org/
Mechanistically, EV-pIgR-induced cancer aggressiveness was licenses/by-nc-nd/4.0/).
abrogated by Akt and b-catenin inhibitors, confirming that the role
of EV-pIgR depends on the activation of the PDK1/Akt/GSK3b/b- Introduction
Hepatocellular carcinoma (HCC) accounts for the majority of
Keywords: Liver cancer; Tumor microenvironment; Intercellular communication; primary liver cancers and is currently the fourth leading cause of
Proteomics; Neutralizing antibody. cancer-related death worldwide.1 HCC is often diagnosed at an
Received 24 April 2021; received in revised form 24 November 2021; accepted 7 advanced stage, thus precluding curative surgical resection.
December 2021; available online 16 December 2021
Therapeutic options for advanced HCC are limited in availability
* Corresponding author. Address: Department of Pathology, 7/F Block T, Queen Mary
Hospital, Pokfulam, Hong Kong, China; Tel.: 852-22552681, fax: 852-22185212. and efficacy. Therefore, understanding how HCC acquires
E-mail address: judyyam@pathology.hku.hk (J.W.P. Yam). aggressive traits is important to revamp the current state of early

Contributed equally
diagnosis and treatment.
https://doi.org/10.1016/j.jhep.2021.12.005

Journal of Hepatology 2022 vol. 76 j 883–895


Research Article Hepatic and Biliary Cancer

A Migration assay Invasion assay


Huh7 PLC/PRF/5 Huh7 PLC/PRF/5

No. of migrated cells ( 102)

No. of migrated cells ( 102)


PBS PBS PBS PBS

No. of invaded cells ( 102)

No. of invaded cells ( 10)


10 6 3 20
**
8 ** 5 ** **
Normal Normal ** Normal Normal 15 ***
-EV ** ** -EV 4 -EV 2 ** -EV * **
6 *
3 10
Cirrhosis Cirrhosis Cirrhosis Cirrhosis
4
-EV -EV 2 -EV 1 -EV
5
2 1
E-HCC E-HCC E-HCC E-HCC
-EV 0 -EV 0 -EV 0 -EV 0
S V V V V S V V V V S V V V V S V V V V
L-HCC PB al-Esis-E C-E C-E L-HCC PB al-Esis-E C-E C-E L-HCC PB al-Esis-E C-E C-E L-HCC PB al-Esis-E C-E C-E
r m ho HC HC r m ho HC HC r m ho HC HC r m ho HC HC
-EV NoCirr E- L- -EV NoCirr E- L- -EV NoCirr E- L- -EV NoCirr E- L-

B 8
Huh7
8
PLC/PRF/5 C
PBS PBS 3 weeks 6 weeks
No. of colonies ( 10)

No. of colonies ( 10)


7 7 **
6 6
5 5
EVs
Normal 4 *** Normal 4 n.s.
(one injection/week;
-EV 3 n.s. -EV 3 15 μg/dose)
2 2
1 1
L-HCC 0 L-HCC 0
S V V S V V Orthotopic Lung
-EV PB al-E C-E -EV PB al-E C-E liver implantation metastasis
r m HC r m HC (MHCC97L tumor seed)
No L- No L-
D E
Bioluminescence signal ( 108)
**

Bioluminescence signal ( 104)


Bioluminescence signal ( 108)

PBS Normal-EV L-HCC-EV Liver Lung **


20 *** 8 ** 10 **
***
6 8
15
6
10 4
4
* n.s.
5 2 **
2

0 0 0
S V V S V V S V V S V V S V V
PB al-E CC-E PB al-E C-E PB al-E CC-E PB al-E C-E PB al-E CC-E
r m H r m HC r m - H r m HC r m - H
No L- No L- No L No L- No L
F G I PBS Normal-EV

Relative vascular area (Dextran+)


Bioluminescence signal (x109)

6
**
PBS

EVs (10 g) ** 5 ***


+ p53-/-;Myc mouse
hepatoblasts (1x105) 4
4
L-HCC-EV Normal-EV

3
14 days n.s.
n.s.
2 L-HCC-EV LectinDextranDAPI 2

1
0
Lung S V V S V V 0
PB al-E CC-E PB al-E CC-E S V V
colonization
rm -H rm L-H PB al-E C-E
of hepatoblasts No L No rm C
No L-H
H PBS Normal-EV L-HCC-EV

Fig. 1. Circulating EVs from patients with HCC promote HCC cell aggressiveness. (A) Migration and invasion assays of HCC cells pretreated with the indicated
EVs. (B) Colony formation assay of EV-treated cells. (C) Schematic diagram of the EV education model. (D) Bioluminescence imaging of animals at the end of the
experiment (n = 5). (E) Ex vivo bioluminescence imaging of liver and lung tissues. (F) Schematic diagram of the experimental metastasis assay. (G) Biolumi-
nescence imaging of excised lungs (left). The intensity of signals in the lung were plotted (middle). Image of lungs after fixation (right). Tumor nodules are
indicated by arrows. (H) H&E staining of lung tissues. Scale bar, 200 lm. (I) Analysis of lung vessel leakiness. Arrowhead indicates areas with diffused dextran.
Scale bar: 20 lm. Data are presented as the mean ± SEM. Student’s t test was used for 2 groups. *p <0.05; **p <0.01; ***p <0.001; n.s., not significant. Cirrhosis-EV,
EV from patients with HBV-related cirrhosis; E–HCC–EV, EV from patients with early HCC; EVs, extracellular vesicles; HCC, hepatocellular carcinoma; L–HCC–EVs,
EVs from patients with late HCC; Normal-EV, EVs from healthy individuals.

884 Journal of Hepatology 2022 vol. 76 j 883–895


A Huh7 30
** ***
B Huh7
100 ** 5

Number of spheroids
PBS Normal-EV L-HCC-EV PBS Normal-EV L-HCC-EV
* ***
79.5% 84.1% 91.5% 80 4

CD133
20

CD133 (%)

CD90 (%)
P1 n.s. 60 3
n.s.
10 n.s. 40 2

1.54% 1.28% 3.80%


20 1

CD90
0
P2 S V V S V V 0 0
PB al-E C-E PB al-E C-E S V V
PB al-E C-E
S V V
PB al-E C-E
orm -HC orm -HC rm -HC rm -HC
N L N L o
N L o
N L
P1 P2
PLC/PF/5 60 PLC/PRF/5
25 5
Number of spheroids

PBS Normal-EV L-HCC-EV *** PBS Normal-EV


V L-HCC-EV ***
50 ** ***
20 4
13.6% 16.1% 23.9%

CD133
40

CD133 (%)

CD90 (%)
n.s. n.s.
P1 30 15 3
n.s.
20 n.s. 10 2
10 5 1
2.75% 2.57% 4.06%

CD90
0
P2 S V V S V V 0 0
PB al-E C-E PB al-E C-E S V V S V V
r m -HC r m HC PB al-E C-E PB al-E C-E
No L No L- r m HC r m HC
No L- No L -
P1 P2

C Huh7 40 **
n.s. PLC/PRF/5 60 n.s.
**
Apoptotic cells (%)

Apoptotic cells (%)


PBS Normal-EV L-HCC-EV PBS Normal-EV L-HCC-EV
30
40
20
20
10
PI

PI

34.5% 34.7% 25.9% 41.2% 38.2% 29.3%


Annexin V 0 Annexin V 0
S V V S V V
PB al-E C-E PB al-E C-E
orm - HC orm -HC
N L N L
D PBS
E Primary implantation Secondary implantation
1,000 cells

120 120
Tumor-free survival (%)

Tumor-free survival (%)

Normal-EV
100 100
L-HCC-EV
80 80
60 n.s. 60 n.s.
PBS 40 * 40
5,000 cells

n.s.
PBS PBS * **
Normal-EV 20 Normal-EV 20 Normal-EV
L- HCC-EV L-HCC-EV
L-HCC-EV 0 0
0 10 20 30 40 50 60 0 5 10 15 20
Day Day
Primary implantation Secondary implantation

Fig. 2. Circulating EVs from patients with late-stage HCC enhance the cancer stemness properties of HCC cells. (A) Hepatosphere formation and serial
passage of HCC cells pretreated with the indicated EVs before plating. Representative photographs of spheroids formed after incubation. The histogram indicates
the number of spheres formed per 1,000 cells. P1 and P2 represent passage 1 and 2, respectively. (B) Flow cytometry analysis of CD90 and CD133 expression in
HCC cells treated with PBS or EVs. (C) Apoptosis assay in which EVs and sorafenib-treated cells were stained with PI and annexin V-FITC. Flow cytometry was
conducted to determine the percentage of apoptotic cells. (D) Image of tumors formed by 1,000 and 5,000 PLC/PRF/5 cells treated with or without EVs before
subcutaneous injection into NOD/SCID mice (n = 8). (E) Tumor-free survival curves of mice injected with 5,000 cells in the primary (left) and secondary (right)
implantation are shown. Data are presented as the mean ± SEM. Student’s t test was used for 2 groups, and a log-rank (Mantel-Cox) test was used for survival
curves. *p <0.05; **p <0.01; ***p <0.001; n.s., not significant. EVs, extracellular vesicles; HCC, hepatocellular carcinoma; L–HCC–EVs, EVs from patients with late
HCC; Normal-EV, EVs from healthy individuals; PI, propidium iodide.

Growing evidence has revealed that intercellular communi- cancer cell-derived EVs are critical messengers that connect tu-
cation is mediated not only by direct cellular contact and soluble mor cells with their local and distant microenvironments,
factors, but also by extracellular vesicles (EVs). EVs are orchestrating multiple systemic pathophysiological processes to
membrane-derived nanometer-sized vesicles that function by facilitate cancer progression.
transferring donor cell-derived bioactive molecules, including Despite the multifarious effect of EVs in cancer development,
lipids, nucleic acids and proteins, into recipient cells. The tumor the role of EVs associated with cancer stem cells (CSCs) remains
microenvironment is a heterogeneous and composite milieu that uncertain, and the function of EVs in hepatic CSCs is scarcely
supports cancer cell growth, survival and metastasis. Evidently, reported. In a chemically induced HCC rat model, EVs derived

Journal of Hepatology 2022 vol. 76 j 883–895 885


Research Article Hepatic and Biliary Cancer

A Expression of proteins of L-HCC-EV B V V


C 1 10
-4 D 2 10
-5

EV -E V -5
compared to Normal-EV 1 10
al- -EV osis CC-ECC-E ***

EV-pIgR level ( g/ g)
EV-pIgR level ( g/ g)
-6
m 5 10
r V h
Upregulated Downregulated No HB Cirr E-H L-H -5 -6
1 10 3 10
-6
IGHG1 ADIPOQ C5 n.s. n.s. * 1 10
-7
IGHG3 F2 APOB -6
8 10
-7
pIgR C4BPA ITIH4 75 pIgR 1 10 7 10
-7
6 10
IGLC6 HRG C3 5 10
-7

LGALS3BP FCN3 MASP1 -7 4 10


-7

1 10 -7
CP THBS1 3 10
-7
VWF FN1 2 10
-7
GAPDH 1 10
IGHA1 C4B -8
37 1 10 0
IGHV5 -51 TTR is Before After
al V os C CC
SERPINA1 C1S rm HB Cirrh HC
Fold difference >2-fold No E- L-H

E Normal-EV HBV-EV Cirrhosis-EV E-HCC-EV L-HCC-EV

50 nm 50 nm 50 nm 50 nm 50 nm

CD63 pIgR

F G H
Copy number of pIgR/HPRT

6 10-5 3
F/5 7L M3
EV-pIgR level ( g/ g)

*** PR CC9 CCL


HA E h7 C/
4 10-5 2 ** MI HL Hu PL MH MH
***
pIgR
75
2 10-5 1
*
**
50 α-tubulin
0 0
HA HLE uh7 RF/5 97L LM3 HA HLE uh7 RF/5 97L LM3
MI H /P CC C MI H /P CC C
C H HC C H HC
PL M M PL M M
I J K
Copy number of pIgR/100 HPRT

Expression of pIgR, log2 (T/NT)

4 1 105
** ** 15
EV-pIgR level (w/w)

1 104
3 10
1 103

2 1 102
5

1 101 0
1
1 100
-5
0 1 10-1 Upregulation, 56% (28/50 cases)
0 1 2 3 4 5 NT T -10 Downregulation, 20% (10/50 cases)

L
Week
Non-tumourous tissues Tumor
Score 0 Score 1 Score 2 Score 3 Score 0 Score 1 Score 2 Score 3

Fig. 3. pIgR-enriched circulating EVs are found in patients with HCC. (A) Proteins modulated by >2-fold in L–HCC–EVs when compared to Normal-EVs
analyzed from proteomic profiling are listed. (B) Immunoblot showing pIgR expression in the indicated EVs, with GAPDH as the loading standard. (C) ELISA
of pIgR levels in circulating EVs from control individuals (Normal, n = 36) and patients with HBV (HBV, n = 20), HBV-related cirrhosis (Cirrhosis, n = 9), early HCC
(E-HCC, n = 49) or late HCC (L-HCC, n = 22). (D) Determination of EV-pIgR in patients with HCC before and after surgery (n = 25). Late-stage cases are indicated
in red and early-stage cases are marked in black. (E) Representative electron micrograph of EVs subjected to immunogold labeling using anti-CD63 and anti-
pIgR antibodies followed by secondary antibodies conjugated to 5 and 10 nm gold particles, respectively. Scale bar: 50 nm. (F) The expression of pIgR in EVs
obtained from conditioned medium of cells was analyzed via ELISA. The expression of pIgR in normal liver cell line (blue) and non-metastatic (orange) and
metastatic (red) HCC cell lines was determined using qPCR (G) and western blotting (H). (I) pIgR levels in circulating EVs collected from mice implanted with
MHCC97L tumor seeds in the liver (n = 4). (J) pIgR mRNA expression in 50 paired T and NT liver tissues was determined by qPCR. (K) Relative fold change in pIgR
mRNA expression between T and NT liver tissues (n = 50). A fold difference >2 was considered to indicate upregulation or downregulation. (L) Representative

886 Journal of Hepatology 2022 vol. 76 j 883–895


from hepatic CSCs augmented tumor growth and metastasis.2 In individuals (Normal-EVs), and from patients with chronic HBV
addition, EVs derived from CD90+ liver cancer cells modulated infection, HBV-related cirrhosis, early HCC (E–HCC–EVs) or late
endothelial cells to facilitate angiogenesis in HCC.3 The signifi- HCC (L–HCC–EVs) for functional analyses. Based on the Minimal
cant impact of CSCs and EVs in HCC underscores the need for a Information for Studies of Extracellular Vesicles (MISEV) guide-
better understanding of the link between EVs and liver CSCs. lines,7 the isolated EVs were validated by their expression of
Polymeric immunoglobulin receptor (pIgR) is widely small EV markers, size and morphology (Fig. S1A-S1C). Notably,
expressed in mucosal epithelial cells and regulates transcytosis the greatest amount of EVs was obtained from late-stage patients
of dimeric IgA and pentameric IgM, which are the first-line an- compared to other serum donors (Fig. S1D).
tibodies against initial infection. Expression of pIgR is upregu- EVs from patients with HCC but not Normal-EVs increased the
lated by proinflammatory cytokines upon viral or bacterial growth and motility of Huh7 and PLC/PRF/5 cells (Fig. S2A-S2D).
infection, thus bridging innate and adaptive immunity.4 Compared to Normal-, Cirrhosis- and E–HCC–EVs, L–HCC–EVs
Emerging findings have revealed the aberrant expression of exhibited the highest potency in promoting cell migration,
pIgR in cancerous tissues. However, the clinical relevance and invasiveness and colony formation (Fig. 1A-B) and in enhancing
functions of pIgR in tumor cells remain uncertain. Intriguingly, tumor development (Fig. S2E). In an orthotopic liver implanta-
studies have yet to report on the functional role of pIgR when tion model, repeated injection of L–HCC–EVs into mice resulted
expressed by tumor-derived EVs. For the first time, we identified in enhanced liver tumor formation and distant lung metastasis
pIgR as a key molecule that confers oncogenic effects on circu- (Fig. 1C-E). A consistent effect of L–HCC–EVs in promoting
lating EVs from patients with HCC. Herein, we uncover the role of metastasis was observed in an experimental metastasis assay
pIgR-enriched EVs in enhancing cancer stemness and aggres- involving coinjection of EVs and murine p53-/-;Myc-transduced
siveness, and provide insight into the clinical relevance of pIgR as hepatoblasts (Fig. 1F-H). L–HCC–EVs were shown to reduce
a promising biomarker and therapeutic target in HCC. expression of the tight junction protein VE-cadherin in human
umbilical vein endothelial cells (Fig. S3) and enhance endothelial
Materials and methods permeability in mice (Fig. 1I). These findings suggest that the
Human samples promotion of metastasis could be ascribed to leakiness in the
Serum samples were randomly collected from healthy donors pulmonary vasculature induced by L–HCC–EVs, thus facilitating
with non-liver disease backgrounds (as controls), individuals extravasation and colonization of tumor cells in the lungs.
with chronic HBV infection and individuals with liver disease
(cirrhosis, early HCC and late HCC) who had not received any HCC patient-derived EVs promote cancer stemness in
treatment. Information on serum donors is listed in Table S1. The HCC cells
collection of serum samples was executed at Queen Mary Hos- In addition to modulating the cancer phenotypes of tumor cells,
pital, Hong Kong and Zhujiang Hospital, Guangzhou, China with L–HCC–EVs increased the cancer stemness properties of HCC
informed consent from all donors. Procedure approval was ob- cells. HCC cells treated with L–HCC–EVs but not Normal-EVs
tained from the Institutional Review Board of The University of showed a marked increase in the ability to form tumorspheres
Hong Kong/Hospital Authority Hong Kong West Cluster (HKU/HA in a spheroid formation assay (Fig. 2A) and enhanced expression
HKW IRB) and Zhujiang Hospital of Southern Medical University. of the CSC markers CD90 and CD133 (Fig. 2B). L–HCC–EVs also
All experiments involving human samples were handled in increased the expression of CD24, CD47 and EpCAM in Huh7 cells
accordance with relevant ethical regulations. but not in PLC/PRF/5 cells (Fig. S4). In addition, L–HCC–EVs
reduced the sensitivity of cells to sorafenib (Fig. 2C). Limiting
Statistical analysis dilution analysis performed in NOD/SCID mice showed that HCC
The readings of all assays were calculated as the mean ± SEM. cells treated with L–HCC–EVs displayed a significant enhance-
Student’s t test, ANOVA and log-rank (Mantel-Cox) tests per- ment in tumorigenicity, with an increased tumor incidence,
formed using Prism software (version 8.0.1, GraphPad) were used shorter tumor latency and increased estimated tumor-initiating
for statistical analysis. A p value of less than 0.05 was considered cell frequency compared to cells treated with PBS or Normal-
statistically significant. EVs (Table S2). The self-renewal ability of L–HCC–EV-treated
For further details regarding the materials and methods used, cells was further expedited as revealed by serial transplantation
please refer to the CTAT table and supplementary information. of primary xenografts into secondary mouse recipients (Fig. 2D).
L–HCC–EV-treated cells showed significantly worse tumor-free
Results survival than cells treated with PBS or Normal-EVs after both
HCC patient-derived EVs promote aggressive phenotypes in primary and secondary implantation (Fig. 2E).
HCC cells
EVs derived from HCC cell lines have been shown to play pivotal pIgR is upregulated in circulating EVs of patients with HCC
roles in enhancing HCC aggressiveness5,6; however, the physio- To ascertain differences in biological activities between EVs of
logical relevance of their functional effect in patients remains healthy controls and those of patients, their proteomic profiles
ambiguous. Circulating EVs were collected from healthy were compared. Among candidates that were modulated by at

=
images of pIgR expression with intensity scores (0-3). Scale bar, 100 lm. (M) The number of cases with each intensity score (left) and the H-scores of pIgR
expression (right) were plotted (n = 143). (N) Representative cases with pIgR overexpression. Scale bar, 100 lm. Alteration of pIgR expression in paired samples are
depicted in the pie chart. Data are presented as the mean ± SEM. Student’s t test for 2 groups was used. *p <0.05; **p <0.01; ***p <0.001; n.s., not significant.
Cirrhosis-EV, EV from patients with HBV-related cirrhosis; E–HCC–EV, EV from patients with early HCC; EVs, extracellular vesicles; HCC, hepatocellular carci-
noma; L–HCC–EVs, EVs from patients with late HCC; Normal-EV, EVs from healthy individuals; NT, non-tumorous; pIgR, polymeric immunoglobulin receptor;
T, tumor.

Journal of Hepatology 2022 vol. 76 j 883–895 887


Research Article Hepatic and Biliary Cancer

M 150 400 N NT
Case # 595
T NT
Case # 611
T
*** 4
300
Number of cases

28

IHC H-score
100
200

100
50
111
0

0 -100
NT T NT T
Total = 143
Score 0
Score 1 T < NT

Score 2 T = NT

Score 3 T > NT

Fig. 3.

least 2-fold in L–HCC–EVs compared to Normal-EVs, pIgR – PLC/PRF/5 cells (Fig. 4G,H), as well as to reduce the sensitivity
which has been implicated in HCC8,9 – was chosen for further of HCC cells to sorafenib (Fig. 4I). The augmenting activity of L–
investigation (Fig. 3A). Upregulation of pIgR in E–HCC– and L– HCC–EVs was also observed in Huh7 cells and was neutralized by
HCC–EVs was corroborated by immunoblotting (Fig. 3B). A sig- anti-pIgR antibody (Fig. S5). These findings suggest that circu-
nificant increase in pIgR was consistently detected in a cohort of lating EVs from patients with HCC augment the cancer stemness
patients’ circulating EVs (Fig. 3C). The level of EV-pIgR was properties of cells through the functions of pIgR.
reduced in 72% (18/25) of patients after surgery (Fig. 3D). To identity the functional role of pIgR in EVs, pIgR was stably
Immunogold labeling revealed the presence of pIgR on the sur- expressed in Huh7 cells using CRISPR synergistic activation medi-
face of CD63+ circulating EVs (Fig. 3E). A higher level of pIgR was ators (SAM-pIgR) and an EV-targeting expression vector (XP-pIgR)
detected in EVs from metastatic HCC cells than in EVs from (Fig. S6A-S6B). EVs collected from the conditioned medium of SAM-
normal liver and non-metastatic cells (Fig. 3F). Similar pIgR pIgR and XP-pIgR cells showed upregulated pIgR compared to EVs
expression profiles were also observed at both the transcript collected from the respective control SAM-CTL and XP-CTL cells.
(Fig. 3G) and protein (Fig. 3H) level in HCC cell lines. In mice The purity and integrity of the isolated EVs were validated (Fig. S6A,
implanted with tumor seeds derived from metastatic S6C,D). As revealed by various in vitro functional assays, SAM-pIgR-
MHCC97L cells, the level of circulating EV-pIgR progressively EVs and XP-pIgR-EVs enhanced the cancer properties of PLC/PRF/5
increased during tumor development (Fig. 3I). These data indi- and Huh7 cells (Fig. 5A-C; Fig. S7A-S7C). This augmenting effect of
cate that tumors were likely the major source of EV-pIgR and SAM-pIgR-EVs and XP-pIgR-EVs in promoting Huh7 cell migration
that its level could be an indicator of tumor development. Similar and invasion was abolished by the addition of anti-pIgR antibody
to EV-pIgR, overall pIgR mRNA expression was significantly (Fig. S7D-S7E). XP-pIgR-EVs were capable of promoting the
upregulated in tumorous tissues compared to non-tumorous metastasis of murine p53-/-; Myc-transduced hepatoblasts (Fig. 5D-
tissues (Fig. 3J). Notably, the pIgR transcript was upregulated in F). Furthermore, XP-pIgR-EVs promoted in vitro self-renewal,
56% (28/50) of patients with HCC (Fig. 3K). In a tissue microarray increased the CD90-and CD133-expressing PLC/PRF/5 population
comprising 143 paired HCC samples, strong (score 3) and mod- (Fig. 5G-H) and suppressed the sensitivity of HCC cells to sorafenib
erate (score 2) positive staining of pIgR was detected in 91.6% (Fig. 5I). Serial transplantation assays showed that PLC/PRF/5 cells
(131/143) of tumorous tissues, whereas 62.9% (90/143) of non- treated with XP-pIgR-EVs displayed a significant enhancement in
tumorous tissues showed weak positive (score 1) or negative tumorigenicity, with an increased tumor incidence, shorter tumor
staining (score 0) (Fig. 3L-M). The H-score revealed a signifi- latency and increased estimated tumor-initiating cell frequency,
cantly higher immunoreactivity of pIgR in tumorous tissue compared to cells treated with PBS or XP-CTL-EVs (Table S3). The
(Fig. 3M). Frequent overexpression of pIgR was found in 77.6% self-renewal ability of XP-pIgR-EV-treated cells was further
(111/143) of cases (Fig. 3N). increased in secondary implants compared to primary xenografts
(Fig. 5J). XP-pIgR-EV-treated cells showed worse tumor-free sur-
vival than cells treated with PBS or XP-CTL-EVs (Fig. 5K). Taken
pIgR is functionally responsible for the tumor-promoting together, these findings demonstrate that the functional capacity of
effect of HCC patient-derived EVs EV-pIgR was consistent with the clinical results showing an upre-
Due to the presence of pIgR on the surface of EVs (Fig. 3E), we gulation of EV-pIgR in patients with HCC. Importantly, these data
examined whether neutralizing antibody against pIgR was able suggest a previously undiscovered role of EV-enriched pIgR in
to block the tumor-promoting effect of L–HCC–EVs. Indeed, promoting HCC aggressiveness, which prompted us to investigate
cotreatment with anti-pIgR antibody blocked the effect of L– the underlying molecular basis of this observation.
HCC–EVs on colony formation, migration and invasion of PLC/
PRF/5 cells (Fig. 4A,B). Furthermore, the metastasis of murine
p53-/-;Myc-transduced hepatoblasts induced by L–HCC–EVs was EV-pIgR activates b-catenin signaling through PDK1/Akt/
significantly hindered by coinjection with anti-pIgR antibody in GSK3b signaling
the experimental metastasis assay (Fig. 4C-F). Anti-pIgR antibody The expression of genes that are involved in cancer stemness and
also abrogated L–HCC–EVs’ ability to promote the in vitro self- drug resistance regulation was analyzed in PLC/PRF/5 and Huh7
renewal and the percentage of CD90-and CD133-expressing cells treated with Normal-EVs and L–HCC–EVs (Fig. 6A). L–HCC–

888 Journal of Hepatology 2022 vol. 76 j 883–895


A B

No. of migrated cells ( 10)

No. of invaded cells ( 10)


PBS + IgG
g 8 PBS + IgG 50 PBS + IgG 10

No. of colonies ( 10)


7 *** *** * ** *** **
40 8
6
5 30 6
4
L-HCC-EV L-HCC-EV 3 L-HCC-EV L-HCC-EV
20 L-HCC-EV L-HCC-EV
4
+ IgG + pIgR Ab 2 + IgG + pIgR Ab + IgG + pIgR Ab
10 2
1
0 0 0
b b b
IgG IgG A IgG IgG A IgG IgG A
S+ V+ IgR S+ V+ IgR S+ V+ IgR
PB C-E V+p PB C-E V+p PB C-E V+p
H C -E H C -E H C -E
L- CC L- C C L- CC
L-H L-H L- H
C D E

Bioluminescence signal (x1010)


Bioluminescence signal (x109)
PBS 2.0 * ** 2.0
L-HCC-EVs (10 g) +IgG
+ p53-/-;Myc mouse ** ***
hepatoblasts (1x105) 1.5 n.s. 1.5 n.s.
+ pIgR Ab (10 g) PBS
14 days +pIgR Ab 1.0 1.0

0.5 0.5
L-HCC-EV
+IgG
0 0
Lung IgG R A +Ig
b G Ab b b
IgG R Ab +IgG R A IgG R Ab +IgG R A
colonization S+ Ig V IgR S+ Ig V Ig S+ Ig V Ig
of hepatoblasts L-HCC-EV PB S+p C-E V+p PB S+p C-E V+p PB S+p C-E V+p
C -E C C
+pIgR Ab PB L - H PB L-H C-
E PB L-H C-
E
CC
L -H HC HC
L- L-
F PBS+IgG L-HCC-EV+IgG
G PBS L-HCC-EV L-HCC-EV 50
*** ***

Number of spheroids
+ IgG + IgG + pIgR Ab ** **
40

30
P1
20

PBS+pIgR Ab L-HCC-EV+pIgR Ab 10

0
b b
IgG IgG A IgG IgG A
P2 S+ V+ IgR S+ V+ IgR
PB C-E V+p PB C-E V+p
C -E C -E
L- H CC L-H CC
L- H L- H
P1 P2

H PBS
+ IgG
L-HCC-EV
+ IgG
L-HCC-EV
+ pIgR Ab
20 8 ** * I 40
Apoptotic cells (%)

11.0% 15.9% 8.0%


*** *** *** ** PBS L-HCC-EV L-HCC-EV
15 6 30 + IgG + IgG + pIgR Ab
CD133 (%)
CD133

CD90 (%)

10 4 20
30.38% 24.41% 29.73%
5 2 10
2.26% 5.86% 3.54%
PI
CD90

0 0 0
b b b
IgG IgG A IgG IgG A IgG IgG A Annexin V
S+ V+ IgR S+ V+ IgR S+ V+ IgR
PB C-E V+p PB C-E V+p PB C-E V+p
C -E C -E C -E
L-H CC L-H CC L-H CC
H H H
L- L- L-

Fig. 4. Anti-pIgR antibody hinders the ability of EVs from patients with HCC to enhance the cancer and cancer stemness properties of HCC cells. PLC/PRF/5
cells treated with L–HCC–EVs in the presence of control IgG or anti-pIgR antibody were subjected to colony formation (A), migration and invasion (B) assays. (C)
Schematic diagram of the experimental metastasis assay. (D) Bioluminescence imaging of animals at the end of experiment and quantification of the intensity of
signals (n = 5). (E) Ex vivo bioluminescence imaging of lungs. (F) Representative image of H&E staining of lung tissues. Scale bar, 200 lm. Cells treated with L–
HCC–EVs and with control IgG or anti-pIgR antibody were seeded for spheroid formation assays (G) and flow cytometry analysis to detect CD90 and CD133
expression (H). Histogram indicating the number of spheres formed per 1,000 cells. P1 and P2 represent passage 1 and 2, respectively. The percentage of
positively stained CD90 and CD133 cells was plotted. (I) Cells pretreated as described and treated with sorafenib were stained with PI and annexin V-FITC. Flow
cytometry analysis was conducted to determine the percentage of apoptotic cells. Data are presented as the mean ± SEM. Student’s t test for 2 groups was used. *p
<0.05; **p <0.01; ***p <0.001. EVs, extracellular vesicles; HCC, hepatocellular carcinoma; L–HCC–EVs, EVs from patients with late HCC; PI, propidium iodide; pIgR,
polymeric immunoglobulin receptor.

EVs exerted the highest potency in activating b-catenin expres- were further confirmed by immunofluorescence microscopy
sion, highlighting its potential in prompting malignant behav- which revealed a notable increase in nuclear translocation of b-
iors. The TOP/FOP reporter system demonstrated the activation catenin in HCC cells following treatment with EVs with enhanced
of b-catenin activity by L–HCC–EVs (Fig. 6B). These observations pIgR expression (Fig. 6C,D; Fig. S8A,B). Notably, the nuclear

Journal of Hepatology 2022 vol. 76 j 883–895 889


Research Article Hepatic and Biliary Cancer

A B

No. of migrated cells (x102)


No. of migrated cells (x102)
4 8 ** 2.5 ** 4
** *

colonies (x102)

No. of colonies (x10)


7 * **
PBS ** PBS n.s. PBS 2.0 n.s.
PBS
3 *
3 n.s. 6

No. of colonies
5 n.s.
1.5
2 4 2
SAM-CTL 1.0 XP-CTL
SAM-CTL
S XP -CTL 3 -EV -EV
-EV 1 -EV 2 1
0.5
1
N

0 0 0 0
SAM-pIgR XP-pIgR

XP TL S
-p -EV

V
SA -CT BS

XP TL S

SA -CT BS
V

V
-p EV

-p -EV

-p EV

-C PB

-E
-C PB
-E

-E

-E
SAM-pIgR
S X pIgR
XP- -EV -EV
M P

M P

R
M L-

M L-
R

Ig
-EV
EV Ig -EV
EV

Ig

Ig

XP
XP
SA

SA
C D

Bioluminescence signal ( 108)


*** XP-CTL XP-pIgR *
*
No. of invaded cells (x102)

No. of invaded cells (x10)


1.0 6 PBS -EV -EV 20 *
** *
PBS 0.8 PBS 5
15
4
0.6 ** n.s.
3 10
SAM-CTL 0.4 XP-CTL
-EV -EV 2
5 n.s.
0.2 1
0 0 0
SAM-pIgR XP-pIgR

S
SA -CT BS

XP TL S
-p EV

V
V

-p -EV

V
-C PB

XP PB

-E

-E
-E

-E
-EV -EV
M P
M L-

R
R

TL

Ig
Ig

Ig

-C

-p
XP

XP
SA

E F G
Bioluminescence signal ( 109)

XP-CTL XP-pIgR XP-CTL XP-pIgR **


8
*** PBS -EV -EV PBS -EV -EV 60 60 **
*** ** **

Number of spheroid

Number of spheroid
50 50

6 n.s.
40 40
P1 n.s.
30 30
4
n.s. 20 20

2 10 10

P2 0 0
0

XP TL S

XP TL S
V

V
-p -EV

-p -EV
-C PB

-C PB
-E

-E
S

S
V
V

V
PB

XP PB
-E
-E

R
XP L-E

-E

Ig

Ig
TL

R
T
Ig

Ig
C

-C

XP

XP
-p

-p
-
XP

XP

H
P1 P2
PBS XP-CTL-EV XP-pIgR-EV 4 *** 5 25
** ** **
1.96% 2.29% 3.22% *
I ***

Apoptotic cells (%)


4 20
CD90

3 n.s. PBS XP-CTL-EV XP-pIgR-EV


CD133 (%)
CD90 (%)

3 15
2
2 n.s. 10
1.11% 1.47% 3.72% 1 22.64% 19.90% 13.98%
1 5
PI
CD133

0 0 0
S V V S V V Annexin V
PB TL-E R-E PB TL-E R-E S V V
PB TL-E R-E
- C -pIg - C -pIg C - pIg
XP XP XP XP -
XP XP
J PBS K Primary implantation Secondary implantation
1,000 cells

120 120
Tumor-free survival (%)

Tumor-free survival (%)

XP-CTL-EV
100 100
XP-pIgR-EV
80 80
60 n.s. 60 n.s.
PBS 40 40
5,000 cells

PBS ** ** PBS * **
XP-CTL-EV 20 XP-CTL-EV 20 XP-CTL-EV
XP-pIgR-EV XP-pIgR-EV
XP-pIgR-EV 0 0
0 5 10 15 20 25 30 0 2 4 6 8 10 12
Day Day
Primary implantation Secondary implantation

Fig. 5. pIgR-enriched EVs exert enhanced promoting effects on HCC cancer and cancer stemness properties. Colony formation (A), migration (B) and invasion
(C) assays were performed on PLC/PRF/5 cells treated with the indicated EVs from Huh7 cells and corresponding control EVs. (D) Luciferase-labeled murine
p53-/-;Myc hepatoblasts were coinjected with the indicated EVs via the tail vein. Bioluminescence imaging of animals was performed 14 days post injection, and
the luciferase signals were quantified. (E) Bioluminescence imaging of excised lungs. (F) Representative image of H&E staining of lung tissues. Scale bar, 200 lm.
Cells treated with the indicated EVs were subjected to hepatosphere formation (G), flow cytometry analysis of the markers CD90 and CD133 (H) and apoptosis
assays (I). Histogram indicating the number of spheres formed per 1,000 cells. P1 and P2 represent passage 1 and 2, respectively. The percentages of positively
stained CD90+ and CD133+ cells and apoptotic cells were analyzed. (J) Image of tumors formed by PLC/PRF/5 cells pretreated with the indicated EVs in 2 rounds of
implantation (n = 8). (K) Tumor-free survival curves of mice injected with 5,000 cells in the primary and secondary implantation are shown. Data are presented as
the mean ± SEM. Student’s t test was used for 2 groups, and a log-rank (Mantel-Cox) test was used for the survival curve. *p <0.05; **p <0.01; ***p <0.001; n.s., not
significant. EVs, extracellular vesicles; HCC, hepatocellular carcinoma; pIgR, polymeric immunoglobulin receptor.

890 Journal of Hepatology 2022 vol. 76 j 883–895


A 2.5
PBS ***
2.5
PBS
***
Normal-EV Normal-EV
2.0 2.0
Late HCC-EV Late HCC-EV

Fold change
Fold change

1.5 * * * 1.5
* ** * * * ** * ***
1.0 1.0

0.5 0.5

0 0
B1 BCC1 BCG2 DHA1 tenin MI1 ANOG ESTIN TCH1 CT4 B1 C1 BCG2 DHA1 atenin MI1 ANOG ESTIN TCH1 CT4
ABC A A AL β-c
a B N N NO O ABC ABC A AL β-c B N N NO O

PLC/PRF/5 Huh7

B C 200 200
D PLC/PRF/5
150 150 PBS XP-CTL-EV
*

Mean fluorescent intensity


Mean fluorescent intensity
*** *** *** ***
***
Luciferase activity

Luciferase activity

n.s. 150 150


100 n.s.
100
100 100
n.s.
n.s.
50 50
50 50

0 0 0 0
S V V D S V V D
S V
PB al-E C-E
V S V
PB al-E C-E
V PB L-E R-E yto PB L-E R-E yto XP-pIgR-EV XP-pIgR-EV+CytoD
T C T C
rm C rm C -C P-pIg EV+ -C P-pIg EV+
No L-H No L-H P
X X R- P
X X R-
g g
PLC/PRF/5 Huh7 -p I -p I
XP XP
PLC/PRF/5 Huh7

E V V V +C
yto
D
V V V +C
yto
D

-E -E -E -E -E -E
TL pIgR pIgR TL pIgR pIgR
S -C - - S -C - -
PB XP XP XP PB XP XP XP

p-PDK1
50 Huh7
PBS XP-CTL-EV
PDK1
50

p-Akt
50

Akt
50
50
XP-pIgR-EV XP-pIgR-EV+CytoD
p-GSK3β

50
GSK3β

50
β-actin
37
PLC/PRF/5 Huh7

Fig. 6. pIgR-enriched EVs activate b-catenin signaling through the PDK1/Akt/GSK3b axis in HCC cells. (A) Expression of genes regulating cancer stemness and/
or drug resistance in HCC cells treated with the indicated EVs was quantified by qPCR. Expression levels are expressed as the fold change compared to the PBS
group. (B) The activity of b-catenin in HCC cells treated with the indicated EVs was analyzed using a TOP/FOP reporter assay. (C) HCC cells treated with EVs
derived from stable XP-CTL and XP-pIgR cells in the absence or presence of CytoD were stained with anti-b-catenin antibody and DAPI. Scale bar, 20 lm. His-
togram indicating the mean fluorescence intensity of the average nuclear b-catenin fluorescence captured by confocal microscopy (n = 10). (D) Representative
fluorescence images. (E) Immunoblotting of PDK1, p-PDK1 (S241), Akt, p-Akt (S473), GSK3b, and p-GSK3b (S9) expression in PLC/PRF/5 cells treated with the
indicated EVs; b-actin was used as the loading standard. Data are presented as the mean ± SEM. Student’s t test was used for 2 groups was used. *p <0.05; **p
<0.01; ***p <0.001; n.s., not significant. CytoD, cytochalasin D; EVs, extracellular vesicles; HCC, hepatocellular carcinoma; L–HCC–EVs, EVs from patients with late
HCC; Normal-EV, EVs from healthy individuals; XP, expression vector.

translocation of b-catenin activated by L–HCC–EVs was sup- was required. In PLC/PRF/5 cells, the phosphorylation of Akt and
pressed by treatment with an endocytosis inhibitor, cytochalasin GSK3b induced by XP-pIgR-EV was diminished by the Akt in-
D, indicating that EVs were taken up by cells via endocytosis. hibitor IV and the b-catenin inhibitor PRI-724 (Fig. 7A). Func-
Phosphorylation of PDK1 leads to activation of Akt, which tionally, treatment with these inhibitors abrogated the
subsequently phosphorylates GSK3b, leading to stabilization and promoting effect of XP-pIgR-EVs on HCC cell growth and motility
nuclear translocation of b-catenin. b-catenin signaling has been (Fig. 7B-D). Moreover, the effect of XP-pIgR-EVs on enhancing
reported to regulate tumorigenesis, cancer stemness and che- the in vitro self-renewal ability of PLC/PRF/5 cells, the percentage
moresistance.10 XP-pIgR-EVs increased PDK1, Akt and GSK3b of CD90+ and CD133+ cells (Fig. 7E-F) and the sensitivity of cells
phosphorylation (Fig. 6E; Fig. S8C), and this activation was to sorafenib (Fig. 7G) was diminished by cotreatment with the
blocked by cytochalasin D, suggesting that cellular uptake of EVs inhibitors. These findings demonstrate that pIgR-enriched EVs

Journal of Hepatology 2022 vol. 76 j 883–895 891


Research Article Hepatic and Biliary Cancer

V
h I 24
A +
SO MSO t In I-7
DM +D +Ak +PR
V -EV -EV -EV
B C
-E
TL gR gR gR
-C -pI -pI -pI
XP XP XP XP XP-CTL-EV XP-pIgR-EV XP-CTL-EV XP-pIgR-EV

Number of migrated cells ( 102)


+DMSO +DMSO 10 +DMSO +DMSO 5 **
DMSO
pAkt (S473)
73) * DMSO

Number of colonies ( 10)


Akt Inh IV * **
50
8 ** ** 4 Akt Inh IV
PRI -724
Akt PRI -724
50 6 3
50
pGSK3β (S9) XP-pIgR-EV XP-pIgR-EV 4 XP-pIgR-EV XP-pIgR-EV 2
+Akt Inh IV +PRI-724 +Akt Inh IV +PRI-724
50
2 1
GSK3β
50 0 0
β-actin XP-CTL XP-pIgR XP-CTL XP-pIgR
37 -EV -EV -EV -EV

D XP-CTL-EV XP-pIgR-EV E XP-CT


XP-CTL-EV XP-pIgR-EV XP-pIgR-EV XP-pIgR-EV 6 *** 8
+DMSO +DMSO * +DMS
+DMSO +DMSO +Akt Inh IV +PRI-724

Number of spheroid ( 10)

Number of spheroid ( 10)


Number of invaded cells ( 10)

8 ** ***
* * 5 **
6 *** ***
6 4
P1
3 4
4
XP-pIgR-EV XP-pIgR-EV 2
+Akt Inh IV +PRI-724 DMSO 2
2 1
Akt Inh IV
PRI-724 0 0
0 XP-CTL XP-pIgR XP-CTL XP-pIgR
XP-CTL XP-pIgR P2 -EV -EV -EV -EV
-EV -EV P1 P2
DMSO Akt Inh IV PRI -724

F XP-CTL-EV
+DMSO
XP-pIgR-EV
+DMSO
XP-pIgR-EV
+Akt Inh IV
XP-pIgR-EV
+PRI-724
6
** 6
***
4.17% 5.57% 4.46% 3.35% ** ** *** ***
CD90

5 5

CD133 (%)
4 4
CD90 (%)

3 3

2 2 DMSO
2.53% 5.40% 3.19% 2.75%
CD133

1 1 Akt Inh IV
PRI -724
0 0
XP-CTL XP-pIgR XP-CTL XP-pIgR
-EV -EV -EV -EV

50 ***
G XP-CTL-EV XP-pIgR-EV pg
XP-pIgR-EV XP-pIgR-EV ** ***
Apoptotic cells (%)

40
+DMSO +DMSO +Akt Inh IV +PRI-724
30

20
DMSO
40.4% 36.8% 41.8% 45.8% 10 Akt Inh IV
PI

PRI -724
0
XP-CTL XP-pIgR
Annexin
Anne in V -EV -EV

Fig. 7. Inhibitors of Akt and b-catenin obstruct the promoting effect of pIgR-enriched EVs on cancer and cancer stemness properties. (A) Immunoblot
showing Akt, phospho-Akt (S473), GSK3b, and p-GSK3b (S9) expression in PLC/PRF/5 cells treated with XP-CTL-EV and XP-pIgR-EV and cotreated with either
vehicle, Akt inhibitor IV (Akt Inh IV) or b-catenin inhibitor PRI-724. Anti-b-actin was used as the loading standard. Treated cells were subjected to colony for-
mation (B), migration (C) and invasion (D) assays. (E) Spheroid formation assays of the cells treated as described above were performed for 2 passages, P1 and P2,
each with 1,000 cells. Flow cytometry analysis of CD90+ and CD133+ cells (F) and apoptotic cells (G) was performed. The percentages of CD90+ and CD133+ cells
and apoptotic cells were plotted. Data are presented as the mean ± SEM. Student’s t test was used for 2 groups. *p <0.05; **p <0.01; ***p <0.001. EV, extracellular
vesicle; XP, expression vector.

promote cancer and cancer stemness properties through the (Fig. 8B). Remarkably, cotreatment with sorafenib and anti-pIgR
PDK1/Akt/GSK3b/b-catenin axis. antibody exerted the most potent inhibitory effect on tumor
formation. We did not observe signs of distress or significant
changes in the body weights of mice in any treatment group
Targeting EV-pIgR with a pIgR neutralizing antibody is a
(Fig. 8C). The weight and dimensions of dissected tumors were
potential therapeutic option for HCC
significantly reduced in mice subjected to combined treatment
The therapeutic effect of the anti-pIgR antibody and sorafenib
with sorafenib and anti-pIgR antibody (Fig. 8D). Immunohisto-
was tested in mice implanted with PDTXs that expressed pIgR
chemistry analysis revealed no change in cellular pIgR expres-
(Fig. 8A). Administration of anti-pIgR antibody or sorafenib
sion in tumors from different treatment groups, suggesting that
suppressed tumor growth compared to vehicle treatment

892 Journal of Hepatology 2022 vol. 76 j 883–895


A B 1,200 PBS/IgG C 25
Sorafenib
1,000

Tumor volume (mm3)


Subcutaneous pIgR Ab

Body weight (g)


injection Sorafenib+pIgR Ab
800 20
* ** ***
600
0 3 6 9 12 15 Days
400 * 15 PBS/IgG
Tumor size Sorafenib Sorafenib
reached 200 pIgR Ab
about 90 mm3 pIgR Ab
Sorafenib+pIgR Ab
Sorafenib+pIgR Ab 0 10
0 3 6 9 12 15 0 3 6 9 12 15
Day Day

D 0.8 ** 20 ***

Max. tumor dimension (mm)


n.s. *
* *** n.s.

Tumor weight (g)


PBS/IgG 0.6 15 n.s.
*
Sorafenib *
0.4 n.s. 10
**
pIgR Ab
0.2 5
Sorafenib/pIgR Ab

0 0
IgG nib Ab Ab IgG nib Ab Ab
BS/
or afe pI gR pI gR BS/
or afe pI gR pI gR
P S ib/ P S ib/
en en
oraf oraf
S S
Fig. 8. Treatment with sorafenib and pIgR neutralizing antibody effectively inhibited the development of patient-derived tumor xenografts. (A) Schematic
diagram of the treatment regimen applied to mice subcutaneously implanted with patient-derived tumor xenografts. Mice were administered vehicle (DMSO in
PBS and IgG), sorafenib, anti-pIgR neutralizing antibody or a combination of sorafenib and anti-pIgR neutralizing antibody. DMSO/PBS and sorafenib were
administered daily via oral gavage. Control IgG and anti-pIgR neutralizing antibody were injected peritoneally every 3 days (n = 8). (B) Tumor growth was
monitored and recorded every 3 days until the end of the experiment. Tumor volume was estimated and plotted against time. (C) The body weight of the mice
was examined every 3 days until the experimental endpoint. The tumors were excised from the mice. (D) The weight and maximum dimension of tumors were
measured and plotted. Data are presented as the mean ± SEM. Two-way ANOVA with Turkey’s test was used for multiple groups, or Student’s t test was used for 2
groups. *p <0.05; **p <0.01; ***p <0.001; n.s., not significant. Ab, antibody; pIgR, polymeric immunoglobulin receptor.

the pIgR neutralizing antibody acts by targeting EV-pIgR but not that pIgR is highly expressed in hepatocarcinoma cells and plays
endogenous pIgR (data not shown). important oncogenic functions in HCC. More than 30 years ago,
the detection of high levels of secretory components, which
Discussion result from proteolytic cleavage of pIgR, in the sera of patients
Dysregulation of pIgR was initially reported 45 years ago in with HCC was reported.27 High levels of secretory pIgR have also
malignantly transformed epithelial cells.11–13 Since then, studies been detected in the sera of patients with bladder carcinoma,
have reported contrasting expression of pIgR in different human lung carcinoma and colonic carcinoma with liver metastasis.29–31
carcinomas,14–23 and its role in prognostication of carcinomas Previously, the existence of pIgR within EVs has been reported in
remains inconclusive. pIgR was previously implicated in metas- both CCA and HCC.32 In the present study, our data underscore
tasis by a study demonstrating its positive expression in hepatic the importance of EV-pIgR in hepatocarcinogenesis and the po-
metastatic tissues of colon carcinoma.24 pIgR has been reported tential of EV-pIgR as a diagnostic and prognostic marker in HCC.
to be mostly expressed in cholangiocytes, contributing to an Although alterations in pIgR expression have been detected in
increased secretory pIgR level in the serum.25–27 Immunohisto- different cancer types, the understanding of the mechanism
chemistry of pIgR revealed low to medium staining in hepato- leading to dysregulation of pIgR is limited. In HBV-related HCC,
cytes but no staining in cholangiocytes based on data retrieved pIgR interacts with and activates the Smad2/3 complex leading
from the Human Protein Atlas. Comparison of tissue expression to epithelial-mesenchymal transition (EMT), providing a missing
levels in cholangiocarcinoma (CCA) and HCC in the Cancer link between chronic inflammation and HCC metastasis.8 Of
Genome Atlas and the Genotype-Tissue Expression databases note, it is possible that HBV-related chronic inflammation might
revealed pIgR expression in both carcinoma types, although contribute to changes in the microenvironment, which result in
there was no significant difference between tumorous and non- increased pIgR expression and promote HCC metastasis. Addi-
tumorous tissue. In line with the reported overexpression of pIgR tionally, a previous study suggested EV-pIgR is a non-specific
in HCC,8,9,28 we observed increased transcript and protein levels biomarker related to inflammation in CCA and HCC.32 However,
of pIgR in tumorous tissue compared with their counterparts, as we did not observe an increased level of EV-pIgR in either pa-
well as in metastatic HCC cells compared with non-metastatic tients with HBV or cirrhosis. Thus, the involvement of EV-pIgR in
HCC cells and immortalized liver cells. Our findings suggest persistent inflammation and viral hepatitis requires further in-

Journal of Hepatology 2022 vol. 76 j 883–895 893


Research Article Hepatic and Biliary Cancer

depth investigation. The functional link between pIgR and EMT is epithelial cell adhesion molecular; EVs, extracellular vesicles;
also substantiated by its inverse correlation with E-cadherin GSK3b; glycogen synthase kinase 3b; HBV, hepatitis B virus; HCC,
expression in pancreatic cancer.33 In addition, pIgR activates the hepatocellular carcinoma; L–HCC–EVs, EVs from patients with
Rac1/CDC42-MEK/ERK cascade to promote HCC tumor growth.9 late HCC; PBS, phosphate-buffered saline; PDK1, 3-
Another study revealed that bufalin inhibits HCC cell growth phosphoinositide dependent kinase 1; PDTX, patient-derived
and motility by suppressing APOBEC3 and pIgR.34 pIgR has been tumor xenograft; pIgR, polymeric immunoglobulin receptor;
shown to exert oncogenic functions by activating ribosomal qPCR, quantitative PCR; SAM, synergistic activation mediators;
proteins in HCC.28 However, these studies were either conducted SEM, standard error of mean; XP, XPack expression vector.
in a physiologically irrelevant canine kidney cell line MDCK or in
SMMC7721, BEL7404 and SKHEP1 cells which have been re- Financial support
ported to be either HeLa cell contaminated or of endothelial The work was supported by in part by the funding of Research
origin.35 Therefore, it is not surprising that neither endogenous Grants Council Theme-Based Research Project on Liver Cancer
pIgR nor EV-pIgR activated Smad2/3 signaling and induced EMT Stemness (Project No. T12-704/16-R); The University of Hong
in HCC cells (data not shown).8 Here, the EV-pIgR-induced PDK1/ Kong Seed Funding for Strategic Interdisciplinary Research
Akt/GSK3b/b-catenin axis was observed in different HCC cell Scheme (Project no.: 102009863 and 007000142).
lines, regardless of their b-catenin mutation or activation sta-
tus.36 This mechanistic finding demonstrates how an immuno- Conflict of interest
globulin can be associated with tumorigenesis induced by EVs. The authors have no conflict of interest to declare.
Intriguingly, during our establishment of stable clones with Please refer to the accompanying ICMJE disclosure forms for
enhanced pIgR expression, we noticed the discrete molecular further details.
size of endogenous pIgR and EV-pIgR. Treatment of cells with the
N-glycosylation inhibitor tunicamycin blocks the glycosylation of Author’s contributions
pIgR, revealing 2 distinct sizes of 110 kDa and 83 kDa.17,37 SKT, SWKW contributed equally to this work. Conceptualization:
Functionally, the glycosylation of pIgR aids in transcytosis and SKT, JWPY; Methodology: SKT, JWPY; Investigation: SKT, SWKW,
mediates the attachment of bacteria.37,38 To date, the functional JYTC, XM, THN, CLSY, ZL, HLF, AHNT, SKYM, TKWL; Resources:
implications of non-glycosylated pIgR remain elusive. Our find- DKHW, LM, MY, CS, ION, PC, KZ, YG, JPY; Manuscript writing: SKT,
ings suggest that pIgR residing within EVs is in a non- JWPY; Funding Acquisition: JWPY.
glycosylated form. Importantly, our observation reveals an un-
reported oncogenic function of non-glycosylated EV-pIgR. Data availability statement
However, the detailed regulatory mechanism of deglycosylation We declare that all data involved in this study are available in the
of pIgR during its packaging into EVs remains unanswered and article along with the supplementary materials.
requires further investigation.
Sorafenib is the first-line systemic therapy for patients with
inoperable advanced HCC; unfortunately, the effect of sorafenib Acknowledgment
is modest, and most patients are highly resistant to sorafenib.39 We acknowledge the assistance of The University of Hong Kong,
Other multikinase inhibitors, such as lenvatinib, regorafenib Li Ka Shing Faculty of Medicine, Centre for PanorOmic Sciences
and carbozantinib, are not superior to sorafenib in terms of Imaging and Flow Cytometry Core for providing equipment
survival. In 2020, bevacizumab (anti-VEGF antibody) plus ate- needed for animal imaging and confocal microscopy. Raw data
zolizumab (anti-PD-L1 antibody) treatment was approved as a were acquired using Mass spectrometer maintained by CPOS
first-line treatment for unresectable HCC.40 The combination Proteomics and Metabolomics Core. We also thank Centre for
strategy, which has a substantially better outcome than sorafenib Comparative Medicine Research for providing animals and fa-
alone, acentuates the advantages of combination treatment over cility for animal experimentation and the Electron Microscope
monotherapy. Herein, our findings suggest that blockade of EV- Unit for providing service and support needed for experiments
pIgR with neutralizing antibodies either alone or in combina- involving electron microscope.
tion with sorafenib or other therapeutic agents may serve as a
therapeutic option beyond the current limited treatments Supplementary data
for HCC. Supplementary data to this article can be found online at https://
In conclusion, our study provides compelling evidence doi.org/10.1016/j.jhep.2021.12.005.
implicating patient-derived EVs with enhanced pIgR expression
as key mediators of HCC cancer stemness and tumorigenesis. We References
also demonstrate that EV-pIgR exerts oncogenic functions via Author names in bold designate shared co-first authorship
activation of b-catenin and that blockade of EV-pIgR with a
[1] Villanueva A. Hepatocellular carcinoma. N Engl J Med
neutralizing antibody is a potential treatment alternative.
2019;380:1450–1462.
[2] Alzahrani FA, El-Magd MA, Abdelfattah-Hassan A, Saleh AA, Saadeldin IM,
Abbreviations El-Shetry ES, et al. Potential effect of exosomes derived from cancer stem
Akt, protein kinase B; ANOVA, analysis of variance; CCA, chol- cells and MSCs on progression of DEN-induced HCC in rats. Stem Cells
Int 2018;2018:8058979.
angiocarcinoma; CSC, cancer stem cell; E–HCC–EVs, EVs from [3] Conigliaro A, Costa V, Lo Dico A, Saieva L, Buccheri S, Dieli F, et al. CD90+
patients with early HCC; ELISA, enzyme-linked immunosorbent liver cancer cells modulate endothelial cell phenotype through the release
assay; EMT, epithelial-mesenchymal transition; EpCAM, of exosomes containing H19 lncRNA. Mol Cancer 2015;14:155.

894 Journal of Hepatology 2022 vol. 76 j 883–895


[4] Kaetzel CS. The polymeric immunoglobulin receptor: bridging innate and [23] Krajci P, Meling GI, Andersen SN, Hofstad B, Vatn MH, Rognum TO, et al.
adaptive immune responses at mucosal surfaces. Immunol Rev Secretory component mRNA and protein expression in colorectal ade-
2005;206:83–99. nomas and carcinomas. Br J Cancer 1996;73:1503–1510.
[5] Mao X, Tey SK, Yeung CLS, Kwong EML, Fung YME, Chung CYS, et al. [24] Liu F, Ye P, Bi T, Teng L, Xiang C, Wang H, et al. COLORECTAL polymeric
Nidogen 1-enriched extracellular vesicles facilitate extrahepatic metas- immunoglobulin receptor expression is correlated with hepatic metas-
tasis of liver cancer by activating pulmonary fibroblasts to secrete tumor tasis and poor prognosis in colon carcinoma patients with hepatic
necrosis factor receptor 1. Adv Sci (Weinh) 2020;7:2002157. metastasis. Hepatogastroenterology 2014;61:652–659.
[6] Mao X, Zhou L, Tey SK, Ma APY, Yeung CLS, Ng TH, et al. Tumour extra- [25] Delacroix DL, Courtoy PJ, Rahier J, Reynaert M, Vaerman JP, Dive C.
cellular vesicle-derived Complement Factor H promotes tumorigenesis Localization and serum concentration of secretory component during
and metastasis by inhibiting complement-dependent cytotoxicity of massive necrosis of human liver. Gastroenterology 1984;86:521–531.
tumour cells. J Extracell Vesicles 2020;10:e12031. [26] Tomana M, Kulhavy R, Mestecky J. Receptor-mediated binding and uptake
[7] Thery C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, of immunoglobulin A by human liver. Gastroenterology
Andriantsitohaina R, et al. Minimal information for studies of extracellular 1988;94:762–770.
vesicles 2018 (MISEV2018): a position statement of the International [27] Rossel M, Seilles E, Voigt JJ, Vuitton D, Legait N, Revillard JP. Polymeric Ig
Society for Extracellular Vesicles and update of the MISEV2014 guidelines. receptor expression in hepatocellular carcinoma. Eur J Cancer
J Extracell Vesicles 2018;7:1535750. 1992;28A:1120–1124.
[8] Ai J, Tang Q, Wu Y, Xu Y, Feng T, Zhou R, et al. The role of polymeric [28] Zhang Y, Zhang J, Chen X, Yang Z. Polymeric immunoglobulin receptor
immunoglobulin receptor in inflammation-induced tumor metastasis of (PIGR) exerts oncogenic functions via activating ribosome pathway in
human hepatocellular carcinoma. J Natl Cancer Inst 2011;103:1696–1712. hepatocellular carcinoma. Int J Med Sci 2021;18:364–371.
[9] Yue X, Ai J, Xu Y, Chen Y, Huang M, Yang X, et al. Polymeric immuno- [29] Rossel M, Billerey C, Bittard H, Ksiazek P, Alber D, Revillard JP, et al. Al-
globulin receptor promotes tumor growth in hepatocellular carcinoma. terations in polymeric immunoglobulin receptor expression and secretory
Hepatology 2017;65:1948–1962. component levels in bladder carcinoma. Urol Res 1991;19:361–366.
[10] MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, [30] Kvale D, Norstein J, Meling GI, Bormer OP, Brandtzaeg P, Langmark F, et al.
mechanisms, and diseases. Dev Cell 2009;17:9–26. Circulating secretory component in relation to early diagnosis and
[11] Harris JP, Caleb MH, South MA. Secretory component in human mammary treatment of liver metastasis from colorectal carcinomas. J Clin Pathol
carcinoma. Cancer Res 1975;35:1861–1864. 1992;45:568–571.
[12] Poger ME, Hirsch BR, Lamm ME. Synthesis of secretory component by [31] Rossel M, Brambilla E, Billaud M, Vuitton DA, Blanc-Jouvan F, Biichle S,
colonic neoplasms. Am J Pathol 1976;82:327–338. et al. Nonspecific increased serum levels of secretory component in lung
[13] Harris JP, South MA. Secretory component: a glandular epithelial cell tumors: relationship to the gene expression of the transmembrane re-
marker. Am J Pathol 1981;105:47–53. ceptor form. Am J Respir Cell Mol Biol 1993;9:341–346.
[14] Zhou M, Liu C, Cao G, Gao H, Zhang Z. Expression of polymeric immu- [32] Arbelaiz A, Azkargorta M, Krawczyk M, Santos-Laso A, Lapitz A,
noglobulin receptor and its biological function in endometrial adenocar- Perugorria MJ, et al. Serum extracellular vesicles contain protein bio-
cinoma. J Cancer Res Ther 2019;15:420–425. markers for primary sclerosing cholangitis and cholangiocarcinoma.
[15] Wang X, Du J, Gu P, Jin R, Lin X. Polymeric immunoglobulin receptor Hepatology 2017;66:1125–1143.
expression is correlated with poor prognosis in patients with osteosar- [33] Kadaba R, Birke H, Wang J, Hooper S, Andl CD, Di Maggio F, et al.
coma. Mol Med Rep 2014;9:2105–2110. Imbalance of desmoplastic stromal cell numbers drives aggressive cancer
[16] Niu H, Wang K, Wang Y. Polymeric immunoglobulin receptor expression processes. J Pathol 2013;230:107–117.
is predictive of poor prognosis in glioma patients. Int J Clin Exp Med [34] Yang Z, Tao Y, Xu X, Cai F, Yu Y, Ma L. Bufalin inhibits cell proliferation and
2014;7:2185–2190. migration of hepatocellular carcinoma cells via APOBEC3F induced in-
[17] Arumugam P, Bhattacharya S, Chin-Aleong J, Capasso M, Kocher HM. testinal immune network for IgA production signaling pathway. Biochem
Expression of polymeric immunoglobulin receptor and stromal activity in Biophys Res Commun 2018;503:2124–2131.
pancreatic ductal adenocarcinoma. Pancreatology 2017;17:295–302. [35] Rebouissou S, Zucman-Rossi J, Moreau R, Qiu Z, Hui L. Note of caution:
[18] Ohkuma R, Yada E, Ishikawa S, Komura D, Kubota Y, Hamada K, et al. High contaminations of hepatocellular cell lines. J Hepatol 2017;67:896–897.
expression levels of polymeric immunoglobulin receptor are correlated [36] Ding Z, Shi C, Jiang L, Tolstykh T, Cao H, Bangari DS, et al. Oncogenic
with chemoresistance and poor prognosis in pancreatic cancer. Oncol Rep dependency on beta-catenin in liver cancer cell lines correlates with
2020;44:252–262. pathway activation. Oncotarget 2017;8:114526–114539.
[19] Berntsson J, Lundgren S, Nodin B, Uhlen M, Gaber A, Jirstrom K. [37] Matsumoto N, Asano M, Ogura Y, Takenouchi-Ohkubo N, Chihaya H,
Expression and prognostic significance of the polymeric immunoglobulin Chung-Hsing W, et al. Release of non-glycosylated polymeric immuno-
receptor in epithelial ovarian cancer. J Ovarian Res 2014;7:26. globulin receptor protein. Scand J Immunol 2003;58:471–476.
[20] Qi X, Li X, Sun X. Reduced expression of polymeric immunoglobulin re- [38] Mathias A, Corthesy B. Recognition of gram-positive intestinal bacteria by
ceptor (pIgR) in nasopharyngeal carcinoma and its correlation with hybridoma- and colostrum-derived secretory immunoglobulin A is
prognosis. Tumour Biol 2016;37:11099–11104. mediated by carbohydrates. J Biol Chem 2011;286:17239–17247.
[21] Fristedt R, Gaber A, Hedner C, Nodin B, Uhlen M, Eberhard J, et al. [39] Niu L, Liu L, Yang S, Ren J, Lai PBS, Chen GG. New insights into sorafenib
Expression and prognostic significance of the polymeric immunoglobulin resistance in hepatocellular carcinoma: responsible mechanisms and
receptor in esophageal and gastric adenocarcinoma. J Transl promising strategies. Biochim Biophys Acta Rev Cancer
Med 2014;12:83. 2017;1868:564–570.
[22] Khattar NH, Lele SM, Kaetzel CS. Down-regulation of the polymeric [40] Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim T-Y, et al. Atezolizumab
immunoglobulin receptor in non-small cell lung carcinoma: correlation plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med
with dysregulated expression of the transcription factors USF and AP2. 2020;382(20):1894–1905.
J Biomed Sci 2005;12:65–77.

Journal of Hepatology 2022 vol. 76 j 883–895 895

You might also like