• No results found

University of Groningen Decoding therapeutic roles of adipose tissue-derived stromal cells and their extracellular vesicles in liver disease Afsharzadeh, Danial

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Decoding therapeutic roles of adipose tissue-derived stromal cells and their extracellular vesicles in liver disease Afsharzadeh, Danial"

Copied!
51
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Decoding therapeutic roles of adipose tissue-derived stromal cells and their extracellular

vesicles in liver disease

Afsharzadeh, Danial

DOI:

10.33612/diss.121499227

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Afsharzadeh, D. (2020). Decoding therapeutic roles of adipose tissue-derived stromal cells and their

extracellular vesicles in liver disease. University of Groningen. https://doi.org/10.33612/diss.121499227

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

CHAPTER

3

Adipose tissue-derived

stromal cells suppress

liver fibrosis in an

extracellular

vesicle-mediated fashion

Danial Afsharzadeh

1

Svenja Sydor

2

Emilia Gore

3

Julian Friedrich

4

Guido Krenning

4

Patrick Van Rijn

5

Ali Canbay

2

Peter Olinga

3

Lars P. Bechmann

2

Martin C. Harmsen

4

Klaas Nico Faber

1,6

Departments of 1Hepatology and Gastroenterology,

4Pathology and Medical Biology, 3Pharmaceutical

Technology and Biopharmacy, Biomedical Engineering5,

and 6Laboratory Medicine, Center for Liver, Digestive and

Metabolic Disease, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.

2Department of Gastroenterology, Hepatology and

Infectious Diseases, Otto von Guericke University Magdeburg, Magdeburg, Germany.

(3)

ABSTRACT

Background & Aim: Liver fibrosis develops in response to chronic liver injury

and is mainly driven by activated hepatic stellate cells and portal myofibroblasts

that produce excessive extracellular matrix proteins. Human adipose

tissue-derived stromal cells (hASC) are able to suppress fibrogenesis, but the underlying

mechanisms are largely unknown. Here, we investigated the anti-fibrotic potential

of hASC-derived extracellular vesicles (EVs) in in vitro, ex vivo and in vivo models

of hepatic fibrosis.

Methods: Primary rat hepatic stellate cells (rHSC) and portal myofibroblasts

(rPMF) were transwell-cocultured with isolated primary hASC, cultured in

hASC-conditioned medium (hASC-CM) or treated with hASC-derived EVs.

CCl

4

-treated (4 weeks) mice were treated (i.v.) with hASC-derived EVs. Human

precision-cut liver slices (hPCLS) were exposed to hASC-derived EVs. Cultured

cells, mouse livers and hPCLS were analysed for markers of fibrosis. hASC-derived

EVs were analysed for their miRNA content.

Results: hASC, hASC-CM and hASC-derived EVs suppressed proliferation and

Collagen I and alpha-smooth muscle actin (αSMA) expression of activated rHSC

and rPMF, while the EV-free fraction of hASC-CM did not. hASC-derived EVs

contained more than 1,000 types of miRNAs, the most abundant of which are

known to mediate fibroblast function and fibrosis.

Administration of

hASC-derived EVs to CCl

4

-treated mice reduced serum markers of liver injury (ALT and

AST), as well as hepatic expression of Col1a1, Acta2, Tgf-β1, Timp1 and Timp2.

Accordingly, EV-treatment reduced hepatic αSMA protein levels and collagen

deposition. Ex vivo, hASC-derived EVs suppressed the expression of COL1A1,

ACTA2 and PAI1 in hPCLS.

Conclusion: hASC-derived EVs show potent anti-fibrotic potential in vitro, ex

vivo and in vivo and are therefore promising entities for the treatment of liver

(4)

3

3.1. INTRODUCTION

Hepatic fibrogenesis is a wound-healing response, which represents a ubiquitous

reaction of the liver to one of many causes of chronic injury, such as alcoholic,

non-alcoholic, viral and autoimmune hepatitis, as well as exposure to toxins or drugs

1-3

. Chronic liver injury leads to the activation of hepatic fibroblasts, in particular

hepatic stellate cells (HSC) and portal myofibroblasts (PMF), which produce

excessive amounts of extracellular matrix (ECM) proteins, such as collagens and

fibronectins

4-6.

In response to liver injury, HSC undergo a phenotypic switch

from a quiescent, vitamin A-storing cell type residing in the healthy liver, into a

migratory and proliferative, α-smooth muscle actin-positive and ECM-producing,

myofibroblast-like cell

7

. Liver fibrosis may progress to liver cirrhosis, in which

liver vasculature and architecture are largely destroyed, which predisposes to liver

cancer and end-stage liver disease

6,8

. Emerging data from animal and human

studies support the notion that liver fibrogenesis is a dynamic and bidirectional

process and reversal of fibrosis is therefore an important therapeutic target

9-12

.

Nevertheless, drugs to reverse fibrosis in patients are not available yet and liver

transplantation remains the only cure for patients with advanced cirrhosis

13

.

Encouragingly, therapeutic application of mesenchymal stem cells (MSC) have

shown promising results in treatment of fibrosis in murine models

14-17

, as well

as in patients with advanced fibrosis

18,19

. MSC are adult stem cells that reside

in virtually all organs of the body, except for neural tissue and cartilage. MSC

from bone marrow, umbilical cord blood, trabecular bone, synovial membrane

and adipose tissue have already been used in MSC-based therapies

20-22

. Human

adipose tissue-derived stem cells (hASC) are being considered for MSC-based

therapies to treat chronic liver diseases, as they lack human leukocyte

antigen-DR expression, suppress proliferation of activated allogenic lymphocytes and

inhibit inflammatory cytokine production

23

. Moreover, the high abundance of

hASC in adipose tissue and the relative ease of obtaining significant quantities

compared to other tissue-specific MSC make hASC an attractive and practical

source for cell therapy

24

. hASC have already been shown to reverse toxin-

25,26

and

diet-

16

induced liver fibrosis in rodent models. MSC-related therapeutic effects

are considered to be based on paracrine signaling

27-32

, but the identification and

characterization of the relevant factors remain to be determined. Various trophic

molecules have been considered as the main anti-inflammatory, anti-apoptotic

and anti-fibrotic factors in the secretome of cultured MSC

33-36

, while the influence

of MSC-derived EVs has not been thoroughly investigated yet. EVs are carriers

of a variety of RNAs and proteins and are able to deliver this cargo to target cells.

Cargo of EVs may modulate key regulatory processes, such as transcription, post

(5)

transcriptional modulation and signal transduction in the recipient cells

37-43

.

Particular

microRNAs (miRNAs) have been shown to be enriched in EVs and

may strongly affect the expression of target genes in recipient cells

44,45

. Here,

we

investigated the anti-fibrotic potential of hASC-derived EVs in in vitro, ex vivo

and in vivo models of liver fibrosis.

3.2. MATERIAL AND METHODS

3.2.1. Human liver

This study was approved by the Medical Ethical Committee of University Medical

Centre Groningen, according to the Dutch legislation and the Code of Conduct

for dealing responsibly with human material in the context of health research

(https://www.federa.org/codes-conduct), refraining the need of written consent

for the ‘further use’ of coded-anonymous human tissue. Clinically healthy

liver tissue was obtained from surgical excess material acquired from donors

undergoing partial hepatectomy or organ donation. The liver tissue was stored in

ice-cold tissue preservation solution (University of Wisconsin) between 3 to 5 h.

3.2.2. Preparation of human precision-cut liver slices (hPCLS)

hPCLS (5 mm diameter, 250-300 μm thickness and 4-5 mg wet weight) were

prepared as previously described 46. Slices were obtained with a Krumdieck slicer

(Alabama Research and Development, Munford, AL, USA) in 4°C Krebs-Henseleit

buffer supplemented with 25 mM D-glucose (Merck, Darmstadt, Germany), 25

mM NaHCO3 (Merck), 10 mM HEPES (MP Biomedicals, Aurora, OH, USA)

saturated with carbogen (95% O2/5% CO2) at pH 7.42. Slices were incubated

in Williams’ medium E (with L-glutamine, Fisher Scientific, Landsmeer, The

Netherlands) supplemented with 25 mM D-glucose and, 50 μg/ml gentamycin

(Invitrogen). hPCLS were incubated for 48 h at 37°C in an 80% O2/ and 5% CO2

atmosphere, while horizontally shaken at 90 rpm. hPCLS were treated with a final

concentration of 1x106 hASC-derived EVs per ml of culture medium.

3.2.3. Human Adipose tissue–derived Stromal Cell (hASC) isolation and

culture

Human subcutaneous adipose tissue was obtained under informed consent from

healthy donors with BMI below 30 undergoing liposuction (Bergman Clinics,

The Netherlands). Adipose tissue was stored at 4°C and processed within 24 h

post-surgery. hASC were isolated as described 47 and seeded in culture flasks

at 4x104 /cm2, expanded by passing three times and used for experiments. All

(6)

3

experiments were performed using a pool of hASC from three donors. The use of

adipose tissue as the source of hASC was approved by the local Ethics Committee

of University Medical Center Groningen, given the fact that it was considered the

use of anonymized waste material.

3.2.4. Preparation of hASC-CM

hASC in passage 3-7 were cultured without serum for 24 h. hASC-CM was

collected and centrifuged at 500 xg for 10 min to remove cell debris and stored at

-80°C until use.

3.2.5. Isolation and identification of hASC-derived extracellular vesicles

(EVs)

hASC-derived extracellular vesicles (EVs) were isolated by the gold standard

differential centrifugation method, essentially as previously described 48. Briefly,

serum-free hASC-CM was centrifuged at 10,000 xg for 20 min to remove apoptotic

bodies. The supernatant was collected and subjected to 100,000 xg for 60 min

(optimal-XPN; Beckman Coulter). The EV-enriched pellet was washed in PBS and

subjected to an additional round of ultracentrifugation at 100,000 xg for 90 min.

EVs were resuspended in PBS and, together with the corresponding EV-depleted

hASC-CM, stored at -80°C. hASC-derived EVs were analyzed by transmission

electron microscopy (FEI Tecnai 12; Philips) and with a nanoparticle tracking

analyzer (NanoSight NS500, Malvern, Worcestershire, UK).

3.2.6. CCl4-induced mouse model of liver fibrosis and injection of

hASC-derived EVs

Inbred C57Bl/6 mice were housed in the animal facility of the University

Hospital Essen (ZTL), University of Duisburg-Essen, Germany according to the

recommendations of the Federation of European Laboratory Animal Science

Association (FELASA). All procedures were approved by the Landesamt für

Natur, Umwelt und Verbraucherschutz Nordrhein-Westfalen (LANUV NRW). To

induce liver fibrosis, mice (four-to-six week-old) were injected intraperitoneally

with 0.6 ml CCl4 per kg (Sigma-Aldrich) in corn oil, twice per week for four weeks

as described earlier 49. 2x108 ASC-derived EVs were resuspended in 100 µl PBS

and injected intravenously once per week (3 hours post-CCl4 injection) during

the last two weeks of the CCl4 treatment (CCl4+EV group) (n =8). The control

groups were administered with CCl4 and PBS (CCl4+PBS group) (n=8) or corn

oil and PBS/EVs (oil+PBS and oil+EV groups) (n= 6). The two-weeks control

mice were subjected to either CCl4 or corn oil for the duration of two weeks

(n=6). At the end of the experiment, blood samples were taken, clotted at room

(7)

temperature, and the isolated serum was stored at -80°C for further analysis. Liver

samples were snap-frozen in liquid nitrogen and stored at -80°C until isolation

of RNA or fixed in 4.5% formalin, paraffin-embedded and sectioned following

standard procedures 50.

3.2.7. Isolation, culture and treatment of rat hepatic stellate cells (rHSC) and

portal myofibroblasts (rPMF)

Specified pathogen-free male Wistar rats (350 – 400 g; Charles River Laboratories

Inc., Wilmington, MA, USA) were kept under standard laboratory conditions

with free access to standard laboratory chow and water. All experiments were

performed according to Dutch law on welfare of laboratory animals and

guidelines of the ethics committee of University of Groningen for the care and

use of laboratory animals. Primary rHSC were obtained as described before 51

by perfusion of the liver with pronase (Merck, Amsterdam, the Netherlands)

and collagenase-P (Roche, Almere, the Netherlands) and further purified by

Nycodenz (Axis-ShieldPOC, Oslo, Norway) gradient centrifugation. rPMF were

obtained by outgrowth from intrahepatic bile duct segments 52. rHSC and rPMF

were cultured in Iscove’s Modified Dulbecco’s Medium (IMDM) with Glutamax

(Invitrogen, Brenda, the Netherlands) supplemented with 20% heat-inactivated

fetal calf serum (Invitrogen), 1 mmol/l sodium pyruvate (Invitrogen), 1x

nonessential amino acids (Invitrogen), 50 µg/ml gentamicin (Invitrogen), 100 U/

ml penicillin (Lonza, Vervier, Belgium), 10 µg/ml streptomycin (Lonza) and 250

ng/ml fungizone (Lonza) in a humidified incubator at 37°C with 5% CO2. rHSC

and rPMF were cultured to a density of ~1.5x104 cells per cm2 and transwell

co-cultured with an equal number of hASC or co-cultured in hASC-CM (see “Real-time

monitoring of cell proliferation” below). rHSC and rPMF were treated with a final

concentration of 1x106 hASC-derived EVs per ml of culture medium or cultured

in the EV-depleted hASC-CM, which was supplemented with serum.

3.2.8. RNA isolation and qPCR

RNA was isolated using TRI reagent (Sigma-Aldrich‎) according to the

manufacturer’s instructions. Reverse transcription was performed on 2.5 µg

total RNA using random nanomers (Sigma-Aldrich) in a final volume of 50 µl.

Real-time semi-quantitative PCR (qPCR) was performed on the 7900HT Fast

Real-TimePCR system (Applied Biosystems Europe, The Netherlands) using the

TaqMan or SYBR Green protocol 53. mRNA levels were normalized to ACTB/

Actb and further normalized to the mean expression level of the control group.

qPCR primers and probes are shown in Supplementary Tables S1 and S2.

(8)

3

3.2.9. miRNA isolation, library preparation and sequencing

miRNAs were isolated from hASC and hASC-derived EVs according to the

mirVana PARIS kit manual (Ambion, Carlsbad, CA, USA). Next generation

sequencing libraries were constructed using the TruSeq Small RNA Sample Prep

Kit (Illumina, San Diego, CA, USA) according to the manufacturer’s instructions.

Libraries were subsequently sequenced on a Nextseq550 sequencer (Illumina, San

Diego, CA, USA).

3.2.10. Real-time monitoring of cell proliferation

Proliferation of rHSC and rPMF was monitored using the xCELLigence system

(RTCA DP; ACEA Biosciences, Inc., San Diego, CA, USA). rHSC and rPMF

were seeded in E-plates with interdigitated gold microelectrodes to constantly

record the cell proliferation (Δ Cell index/ h-1), according to manufacturer’s

instructions54. rHSC and rPMF were cultured and treated with isolated

hASC-derived EVs as it was described (see “Isolation, culture and treatment of rHSC and

rPMF” above). Results were recorded and analyzed by the xCELLigence software.

3.2.11. Immunofluorescence microscopy

rHSC and rPMF were cultured on coverslips, fixed with 4% paraformaldehyde in

PBS (Merck Millipore) and permeabilized in 1% Triton-X100. Coverslips were

incubated with primary antibodies against rat collagen type 1 (1:400; Southern

Biotech, Birmingham, AL, USA) and rat αSMA (1:400; Sigma-Aldrich, St. Louis,

MO, USA) according to the manufacturer’s instructions. Further, coverslips were

labeled using the GFP-tagged secondary antibody for collagen type 1 (1: 2,000

polyclonal Rabbit Anti-Goat Immunoglobulins, Dako, Denmark) and the

GFP-tagged secondary antibody for αSMA (1: 2,000 polyclonal Rabbit Anti-mouse

Immunoglobulins, Dako, Denmark). Later, all coverslips were mounted in the

fluorescence mounting medium S3023 (DAKO, Heverlee, Belgium). Images were

captured using a Leica DMI6000 and analyzed by ImageJ image analysis software

(ImageJ; National Institutes of Health (NIH), Bethesda, MD, USA) and Adobe

Photoshop CS6.

3.2.12. Histopathological staining

Liver tissues were processed for paraffin embedding and 4-µm-thick sections were

prepared. The sections were stained with picrosirius red and counter-stained with

hematoxylin and eosin according to standard protocols. Picrosirius red staining

was quantified using ImageJ image analysis software (ImageJ; NIH).

(9)

3.2.13. Immunohistochemistry

αSMA immunostaining on 4-µm liver sections was carried out using the MOM

kit (Vector labs, USA), according to the manufacturer’s protocol. Briefly, tissue

sections were antigen retrieved by the overnight incubation of liver sections

with the Tris buffer (0.1 M, pH 9) at 80°C. Further, sections were blocked with

MOM mouse Ig blocking reagent. The primary antibody was detected using the

biotin-conjugated anti-mouse IgG antibody, and then incubated with Vectastain

and Novared. The complex was visualized with 3,3-diaminobenzidine reagent for

microscopical examination. Absence of the primary antibody from the procedure

served as negative control. The quantification of αSMA positive areas was

performed using Image-J image analysis software (ImageJ; NIH).

3.2.14. Serum liver damage marker assays

Serum levels of lactate dehydrogenase (LDH), aspartate transaminase (AST) and

alanine aminotransferase (ALT) levels were quantified using the LDH, AST and

ALT Activity Assay Kits (all Abcam), respectively.

3.2.15. Pathway enrichment analysis

To identify pathways enriched with targets of abundant miRNAs in EVs, miRNAs

with a proportion of at least 0.5 % in hASC-derived EVs were selected. For each

miRNA, predicted targets were obtained using miRNAtap and KEGG pathway

enrichment analysis was performed using clusterProfiler55, with a

Bonferroni-Holm adjusted cut-off set to P < 0.1. KEGG pathways with significantly enriched

targets of at least 6 miRNAs were displayed using ggplot2 56. All analysis was

conducted in R (R Foundation for Statistical Computing, Vienna, Austria). The

bigger the size of the circle, the more counts of predicted targets of a certain

miRNA in the pathway. The darker a circle, the lower the adjusted p-value of the

enrichment analysis of a miRNA.

3.2.16. Statistical analysis

Data are expressed as average with standard deviation. Statistical significance was

determined using Student’s t-test or by One-way ANOVA (with Tukey’s

post-hoc test for individual experimental conditions). All tests were performed with

GraphPad Prism (v. 5.0; GraphPad Software, La Jolla, CA, USA). Differences were

considered significant at P < 0.05.

(10)

3

3.3. RESULTS

3.3.1. ASC suppress proliferation and activation of HSC and PMF in a

paracrine manner.

Culture-activated primary rat HSC (Figure 1A) or PMF (Figure 1B) were

co-cultured with human ASC (hASC) for 48 h in a transwell system allowing only

paracrine signaling between the two cell types and cell proliferation of HSC/PMF

was monitored using a real-time cell analyzer (xCELLigence). Monocultures of

HSC or PMF served as controls. Proliferation of rHSC and rPMF was significantly

suppressed in the presence of hASC as monitored by a delayed increase in cell

index during the full 48 h (grey lines representing a single experiment and grey

bars for n=4). Especially the proliferation of rPMF appeared highly sensitive to

co-culturing with hASC. The hASC-mediated inhibition of HSC/PMF proliferation

was accompanied by a significant reduction in Col1a1 and Acta2 expression in

rHSC and rPMF, as compared to the rHSC and rPMF monocultures (Figure

1A,B). Next, rHSC and rPMF were cultured for 48 h in 24 h-conditioned medium

produced by hASC (hASC-CM; Figure 1 C,D). Similar as observed for the

co-culture experiments, hASC-CM strongly suppressed proliferation of rHSC and

rPMF, as well as Col1a1 and Acta2 expression, compared to these cells grown

in normal (DMEM) media. In line with the effect on the gene expression, both

co-culture with hASC and exposure to hASC-CM strongly reduced protein

expression of collagen type-1 and αSMA in rHSC and rPMF as analyzed by

immunofluorescence microscopy (Figure 1E). These results show that hASC

secrete factors that suppress activation and proliferation of hepatic fibroblasts.

3.3.2. hASC-derived EVs suppress the proliferation and activation of rHSC

and rPMF.

Next, we analyzed the nature of the hASC-derived factors carrying the

anti-fibrotic properties. The hASC-CM was cleared from (apoptotic) cell remnants

followed by ultracentrifugation (90’@100,000xg) to obtain a pellet fraction with

particle-bound/encapsulated factors, in particular extracellular vesicles (EV), and

a supernatant fraction with soluble factors. The EV-fraction showed high potential

to suppress rHSC and rPMF proliferation (Figure 2A), and reduced Col1a1 and

Acta2 mRNA (Figure 2C) and corresponding collagen type-1 and αSMA protein

levels (Figure 2E). In sharp contrast, soluble factors in hASC-CM did not affect

rHSC and rPMF proliferation (Figure 2B), nor the mRNA (Figure 2D) and

protein levels (Figure 2E) of collagen type-1 and αSMA. These data indicate that

anti-fibrotic factors secreted by hASC are contained in the pelletable entities.

(11)

A

B

C

D

Fig

ur

e 1. H

uma

n A

di

pos

e t

iss

ue-d

eriv

ed s

tr

oma

l c

el

ls (hA

SC) s

up

pr

ess p

ro

lif

er

at

io

n a

nd ac

tiva

tio

n o

f r

at HSC a

nd PMF

.

C

on

tin

ue

d o

n n

ext p

ag

e. L

eg

en

d o

n n

ext p

ag

e.

(12)

3

3.3.3. Identification and miRNA content of hASC-derived EVs.

Morphological characterization of the pelletable fraction of hASC-CM by

TEM, revealed sphere-shaped vesicles of rather homogeneous size (Figure 3A).

Nanoparticle analysis showed that approximately 85% of vesicles were sized

between 55-220 nm in diameter, with distinguishable peaks at 55, 115 and 165

nm (Figure 3B). This size distribution suggests that most of the purified

derived EVs were exosomes. Isolation and sequencing of miRNA from the

hASC-derived EVs revealed that they contain over 1,000 unique miRNAs (Supplementary

Table S3), but that they were mostly enriched for 27 dominant miRNAs that each

contribute for at least 0.5% to the total exosomal miRNA content (Supplementary

Table S3). Pathway enrichment analysis showed that these miRNAs are predicted

to control biological pathways involved in fibroblast proliferation and activation,

such as MAPK-, FoxO-, PI3K-Akt- and p53-signaling, as well as cellular senescence

and plasticity pathways (Figure 3C). Interestingly, most of these miRNAs were

enriched in the ASC-derived EVs, when compared to the cellular miRNA content

of hASC (Supplementary Table S3). These results suggest that miRNAs in

hASC-derived EVs may contribute to the anti-fibrotic actions on in vitro-cultured rHSC

and rPMF.

Figure 1. Human Adipose tissue-derived stromal cells (hASC) suppress proliferation

and activation of rat HSC and PMF. Cell proliferation and Col1a1 and Acta2 gene

expression in culture-activated rHSC (A) and rPMF (B) after transwell co-culturing with

hASC for 48 h (A,B) and after culturing in hASC-conditioned medium (hASC-CM) (C,D);

(E) Immunofluorescence microscopy images of collagen type I (green, top panels,

counterstained with DAPI blue) and αSMA (green, bottom panels) in culture-activated

rHSC (a-c, g-i) and rPMF (d-f, j-l) after transwell co-culturing with hASC (b,h and e,k) or

culturing in hASC-CM (c,I and f,l) for 72 h compared to control-grown rHSC (a,g) and

rPMF (d,j) (scale bar, 40 µm). Data are presented as mean value ± SD of 4 experiments.

*p ≤0.05, **p ≤0.01, ***p ≤0.001, Student’s t test.

(13)

A

B

C

D

Fig

ur

e 2. hA

SC-d

eriv

ed e

xt

rac

el

lu

la

r v

es

icl

es (EVs) s

up

pr

ess p

ro

lif

er

at

io

n a

nd ac

tiva

tio

n o

f c

ul

tur

e-ac

tiva

te

d rHSC a

nd rPMF

.

C

on

tin

ue

d o

n n

ext p

ag

e. L

eg

en

d o

n n

ext p

ag

e.

(14)

3

3.3.4. Administration of hASC-derived EVs alleviates liver fibrosis in

CCl4-induced mice.

To test the anti-fibrotic activity of hASC-derived EVs in vivo, mice were exposed

to CCl4 for 4 weeks with 2 therapeutic tail vein injections of hASC-EVs in the

final 2 weeks before sacrifice. As expected, serum levels of liver injury markers

(ALT, AST and LDH) were increased in mice after four weeks of CCl4-exposure

(Figure 4A-C). Administration of hASC-derived EVs did not change serum AST

or LDH levels, but did significantly suppress serum ALT levels (416.5±105.9 U/L

compared to 1256.1± 194.6 U/L in CCl4-treated mice), the latter being considered

the more specific marker for liver injury. Serum liver damage markers were not

affected in mice receiving hASC-derived EVs only (Figure 4). PicroSirius Red

staining revealed that CCl4 exposure induced collagen deposition in mouse

livers when compared to controls (Figure 5Aa1-b1), which was strongly

suppressed by hASC-EV treatment (Figure 5A-c1, quantification in right panel).

No hepatic collagen deposition was observed in EV-only-treated mice (Figure

5A-d1). Similar results were obtained when analyzing αSMA protein levels by

immunohistochemistry; e.g. CCl4 treatment induced αSMA levels, particularly

in the periportal areas, which was strongly suppressed by hASC-EV treatment

(Figure 5Aa2-d2, including quantification in the right panel). Hepatic levels of

Col1a1, Acta2, Timp1 and Timp2 mRNA, all markers of fibrosis, were increased

Figure 2. hASC-derived extracellular vesicles (EVs) suppress proliferation and activation

of culture-activated rHSC and rPMF. Cell proliferation of activated rHSC and rPMF during

exposure to hASC-derived EVs (A) or EV-depleted hASC-CM (B) monitored for 48 h; Col1a1

and Acta2 gene expression in rHSC and rPMF after culturing with hASC-derived EVs (C)

or EV-depleted hASC-CM (D) for 48 h; (E) Immunofluorescence microscopy images of

collagen type I (green, top panels, counterstained with DAPI blue) and αSMA (green,

bottom panels) in culture-activated rHSC (a-c, g-i) and rPMF (d-f, j-l) after 72 h exposure

to hASC-derived EVs (b,h and e,k) or EV-depleted hASC-CM (c,I and f,l) compared to

control rHSC (a,g) and rPMF (d,j) (scale bar, 40 µm). Data are presented as mean value ±

SD of 4 experiments. *p ≤0.05, **p ≤0.01, ***p ≤0.001, Student’s t test.

(15)

in CCl4-treated mice, when compared to sham-treated controls (Figure 5B),

which were all suppressed when the CCl4-exposed mice were treated with

hASC-derived EVs. In mice receiving only EVs, hepatic expression of Col1a1, Acta2,

Figure 3. Morphology and miRNA content of hASC-derived EVs. (A) Transmission electron

micrograph of hASC-derived EVs, scale bar = 200 µm; (B) Size distribution histogram

of hASC-derived EVs measured by a nanoparticle tracking analyzer (NTA). (C) Pathway

enrichment analysis of the 27 most dominant miRNA (each with a relative abundance of

at least at least 0.5%) in hASC-derived EVs. KEGG pathways with significantly enriched

targets of at least 6 miRNAs are displayed. The bigger the size of the circle, the more

counts of predicted targets of a certain miRNA in the pathway. The darker a circle, the

lower the adjusted p-value of the enrichment analysis of a miRNA.

C

(16)

3

Tgfb1, Timp1 and Timp2 were indistinguishable from sham-treated controls

(Figure 5B).

hASC-derived EVs suppress early onset of fibrosis in human precision-cut

liver slices.

To determine the relevance of our findings for the development of fibrosis in

human liver, we next performed experiments with human precision-cut liver

slices (hPCLS) that were shown earlier to be a relevant model to study the early

onset of liver fibrosis ex vivo 57,58. hPCLS generated from viable non-fibrotic

human liver were therefore cultivated in the absence and presence of hASC for 48

h. As reported before, expression of COL1A1, ACTA2 and PAI1 increased during

the 48 h ex vivo culture period, which were all significantly suppressed when the

hPCLS were co-treated with hASC-derived EVs (Figure 6).

3.4. DISCUSSION

In this study, we show that hASC-derived EVs suppress fibrotic features of hepatic

stellate cells and portal myofibroblasts in vitro, both of which are pivotal in the

development of liver fibrosis. Moreover, hASC-derived EVs suppressed fibrosis

in the in vivo murine CCl4 model of liver fibrosis and suppress the early onset of

fibrosis ex vivo in human precision-cut liver slices. Remarkably, the anti-fibrotic

properties in the hASC-secretome appeared (almost) solely attributable to the EV

fraction, while such features were not detected in the EV-free fraction containing

Figure. 4. hASC-derived EVs ameliorate CCl

4

-induced liver injury in mice. Serum levels

of (A) alanine aminotransferase (ALT), (B) Aspartate aminotransferase (AST), and (C)

lactate dehydrogenase (LDH) of mice (n=8 per group) that were exposed to 4-week CCl

4

treatment with or without co-treatment with hASC-derived EVs in the last 2 weeks. Data

shows mean value ± SD. *p ≤0.05, **p ≤0.01, ***p ≤0.001, ANOVA (with Tukey’s

post-hoc test for individual experimental conditions).

(17)

trophic factors. The hASC-derived EVs carry a great variety of miRNA, the most

dominant of which have been shown earlier to act on fibrosis-related pathways.

Mesenchymal stem cells (MSC) have shown potent therapeutic effects in a vast

variety of diseases, including liver fibrosis. Still, it remains largely unknown what

Figur

e 5.

hASC-d

eriv

ed

EV

s ame

lior

at

e

CCl

4

-induced

liv

er

fibr

osis

in

mice

. (A)

Repr

esen

ta

tiv

e

sirius

red

st

aining

(a1-d1;

sc

ale

bar=1

mm)

and

αSMA

immunohis

tochemis

tr

y

(a2-d2;

sc

ale

bar

=

500

µm)

of

liv

er

sections

from

vehicle-tr

ea

ted

(a),

CCl

4

-tr

ea

ted

(b),

CCl

4

+EV

s

cotr

ea

ted

(c)

and

EV

-only

tr

ea

ted

(d)

mice.

Quan

tific

ation

of

the

st

aining

in

tensity

for

all

animals

(n=8

per

gr

oup)

is

pr

esen

ted

in

the

panels

to

the

righ

t.

(B)

Corr

esponding

hepa

tic

gene

expr

ession

analy

sis

of

Col1a1,

Act

a2,

Tgfb1,

Timp1

and

Timp2

. Da

ta

sho

w

s mean

value

±

SD

. *p

≤0.05,

**p

≤0.01,

***p

≤0.001,

ANO

VA

(with

Tuk

ey

’s pos

t-hoc t

es

t f

or individual e

xperimen

tal c

onditions).

A

B

Upper (g reen) panel Lo w er (br own) panel

(18)

3

the exact mechanisms are that underlie the therapeutic effect in liver fibrosis,

which may involve 1) stem cell-to-liver cell transdifferentiation, 2) juxtacrine, 3)

paracrine or 4) endocrine signaling. We demonstrate that hASC produce EVs that

suppress the proliferation and activation, e.g. ECM production and cytoskeletal

changes, of culture-activated rHSC and rPMF, thus acting through paracrine/

endocrine signaling. This finding is in line with the current believe that the

secretome of MSC is primary responsible for therapeutic effects of these stem

cells31,59,60. An earlier study reported that conditioned medium produced by

human amniotic epithelial cells suppresses collagen production and proliferation

in human hepatic stellate cells and additionally induced HSC apoptosis61. In line,

others have shown that conditioned medium of MSC suppresses CCl4-induced

liver fibrosis and inflammation in mice62. In part, the anti-fibrotic effect of human

umbilical cord MSC has been attributed through the cytokines that they secrete

rather than by differentiation to hepatocytes63. Remarkably, our results show that

the anti-fibrotic effect of the hASC secretome on in vitro-cultured HSC and PMF

is predominantly, if not solely, dependent on the EVs and is absent in the EV-free

conditioned medium after the 100,000xg centrifugation step. EVs are released by

virtually all cell types, including MSC 64. The 100,000xg pellet fraction of

hASC-CM revealed vesicle sizes ranging from ~55-220 nm, which indicates that most of

them have the size typical for exosomes (30-150 nm). However, a distinct peak of

165 nm vesicles was also detected, which suggest that also other types of EVs are

produced by the hASC. EVs carry a great variety of different proteins and various

types of RNA, such as miRNAs, which may induce functional and phenotypic

Figure 6. hASC-derived EVs suppress early onset of fibrosis in human precision-cut

liver slices (hPCLS). hPLCS were exposed to hASC-derived EVs for 48 h, followed by

gene expression analysis for COL1A1, ACTA2 and PAI1. Spontaneous induction of fibrosis

markers is observed during 48 h of hPLCS culturing (compare T=0 and T=48 h), which

is suppressed by co-treatment with hASC-derived EVs. Data shows mean value ± SD.

*p ≤0.05, **p ≤0.01, ***p ≤0.001, ANOVA (with Tukey’s post-hoc test for individual

experimental conditions).

(19)

changes in recipient cells65-68,69,70. Our initial analysis of the hASC-derived

EVs was focused on the miRNA content and revealed that they contain over 1,000

unique types of miRNAs (see Supplementary Table S3). However, most of the

miRNA only appear at very low levels in ASC-derived EVs, where only the top 27

most dominant ones exceeded a relative contribution of 0.5% of the total miRNA

content. Interestingly, ~50% of these abundant miRNAs were enriched in EVs

when compared to the miRNA content of the corresponding hASC, including

21, 181a, 142, 150, 155, 22, 26a, 26b,

27a, 342, 92a, 146b, 16, let-7g, let-7f, let-7a, 30e,

miR-186 and miR-29a. Pathway enrichment analysis indicated various associations

between these miRNAs and pathways regulating fibroblast function and fibrosis,

such as MAP Kinase and FoxO signaling pathways. Mitogen-activated protein

kinases (MAPKs) play central roles in HSC activation and liver fibrosis 71-73 and

MAPK inhibitors alleviate fibrosis in the murine models of liver fibrosis 74-76. In

particular, the various miR-29s have been shown to control MAPK signaling77,78.

miR-29a is the most abundant miR-29 in hASC-derived EVs and suppresses

(cardiac) fibroblast proliferation via modulation of the MAPK pathway79.

Moreover, several EV-associated miRNAs modulate the FoxO signaling pathway,

including let-7a, let-7f, let-7g and miR-29a. As liver inflammation and fibrosis are

augmented by FoxO180,81, it suggests that aforementioned miRNA might have

regulatory roles in anti-fibrotic characteristics of hASC-derived EVs as well. It is

thus likely that the hASC-derived EVs do not act through one unique pathway to

suppress liver fibrosis. The miRNA content would allow modulation of various

pathways controlling hepatic fibroblast proliferation and activation, while other

protein and/or lipid factors contained in the EVs may also contribute to their

overall anti-fibrotic properties.

In the CCl4-treated mice, EVs were administered in the peripheral circulation

while the beneficial effects were apparent in the liver. The biodistribution in vivo

of EVs is similar to that of synthetic membranous nanoparticles, i.e. liposomes,

as they are similar in size and structure of the lipophilic outer layer82-85.

Liposomes (50-200 nm) are able to pass the hepatic endothelium cells because the

sinusoidal endothelial cells are extensively fenestrated with pore sizes of ~50-200

nm86,87.The fact that most of i.v. injected liposomes are captured by the liver and

spleen rather than by other organs88,89, might predict that hASC-derived EVs

accumulate in the mouse liver, even when it is chronically injured by repeated

CCl4 administration. Indeed, Wiklander et al. showed that EVs distribute

predominantly to organs of the mononuclear phagocyte system with highest

accumulation in the liver, followed by spleen, GI-tract and lungs90. Our data show

that hASC-derived EVs ameliorate the early stages of liver fibrosis in mice and

(20)

3

reverse it to a nearly normal condition. As the treatment with EVs also suppressed

CCl4-induced serum ALT levels, the effect on fibrosis could come –in part- also

as a secondary effect due to a protective effect on hepatocytes. However, in pilot

experiments preceding the 4-week CCl4 treatment protocol we first analyzed the

effect of hASC-derived EVs on acute –single dosing- CCl4 exposure. This showed

that EVs suppressed serum ALT levels only when given 1 h before CCl4 treatment

and did not do so when given 3 h after the CCl4 injection (Supplementary Figure

S4). In the 4-week CCl4 protocol, we therefore applied the EV treatment 3 h after

the CCl4-injection in the 3rd and 4th week to determine the anti-fibrotic action of

EVs independent of potential effects on hepatocyte viability. Still, we cannot fully

rule out the possibility that the EV treatment may have an additional hepatocyte

protective effect on CCl4 exposure applied 3-4 days later as performed in the

4-week protocol. Importantly, the anti-fibrotic potential of ASC-derived EVs

was also confirmed in the human ex-vivo approach using hPLCS, where all cell

types of the liver are present in their original context. Hepatocytes, Kupffer cells,

endothelial cells, and hepatic stellate cells remain viable and functional in cultured

hPCLS for at least 48 h57,58. Interestingly, early onset of fibrosis occurs during

PCLS culture as demonstrated by increased expression of COL1A1, ACTA2 and

PAI158. As a prelude to future clinical studies on liver fibrosis, we applied EVs

on hPCLS and found that also in this experimental model, the hASC-derived

EVs suppress transcriptional activation of markers of fibrosis, e.g. COL1A1 and

ACTA2.

Our findings are promising for the future use of hASC-derived EVs as treatment

for liver fibrosis. In conclusion, hASC-derived EVs are promising therapeutic

entities for the treatment of liver fibrosis. Clinical application of hASC-derived

EVs is expected to provide a novel alternative therapy for patients with liver

fibrosis.

Acknowledgements

We are thankful to Sebo Withoff, Rutger Modderman and Ineke Tan from the

department of Genetics, University Medical Center Groningen for their technical

support in miRNA analysis. We also thank Johannes Haybaeck and Kirsten

Herrmanns from the department of Pathology, Otto-von-Guericke-university

hospital Magdeburg, for their time and laboratory technical support.

Funding

This study was funded by EASL Entry-Level fellowship, Short-term training

fellowship Andrew K. Burroughs, De Stichting De Cock-Hadders and

Wilhelm-Laupitz foundation.

(21)

REFERENCES

1. Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: Immune regulation of wound healing in a solid organ. Nat Rev Immunol. 2014;14(3):181-194.

2. Okura H, Soeda M, Morita M, et al. Therapeutic potential of human adipose tissue-derived multi-lineage progenitor cells in liver fibrosis. Biochem Biophys Res Commun. 2015;456(4):860-865.

3. Mormone E, George J, Nieto N. Molecular pathogenesis of hepatic fibrosis and current therapeutic approaches. Chem Biol Interact. 2011;193(3):225-231.

4. Mallat A, Lotersztajn S. Cellular mechanisms of tissue fibrosis. 5. novel insights into liver fibrosis. Am J Physiol Cell Physiol. 2013;305(8):C789-99.

5. Friedman SL. Mechanisms of disease: Mechanisms of hepatic fibrosis and therapeutic implications. Nat Clin Pract Gastroenterol Hepatol. 2004;1(2):98-105.

6. Xu R, Zhang Z, Wang FS. Liver fibrosis: Mechanisms of immune-mediated liver injury. Cell

Mol Immunol. 2012;9(4):296-301.

7. Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol

Hepatol. 2017;14(7):397-411.

8. Ellis EL, Mann DA. Clinical evidence for the regression of liver fibrosis. J Hepatol. 2012;56(5):1171-1180.

9. Friedman SL. Liver fibrosis -- from bench to bedside. J Hepatol. 2003;38 Suppl 1:S38-53. 10. Pellicoro A, Ramachandran P, Iredale JP. Reversibility of liver fibrosis. Fibrogenesis Tissue

Repair. 2012;5(Suppl 1):S26.

11. Iredale JP, Benyon RC, Pickering J, et al. Mechanisms of spontaneous resolution of rat liver fibrosis. hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 1998;102(3):538-549.

12. Issa R, Williams E, Trim N, et al. Apoptosis of hepatic stellate cells: Involvement in resolution of biliary fibrosis and regulation by soluble growth factors. Gut. 2001;48(4):548-557.

13. Kamath PS, Kim WR, Advanced Liver Disease Study Group. The model for end-stage liver disease (MELD). Hepatology. 2007;45(3):797-805.

14. Okura H, Soeda M, Morita M, et al. Therapeutic potential of human adipose tissue-derived multi-lineage progenitor cells in liver fibrosis. Biochem Biophys Res Commun. 2015;456(4):860-865.

15. Jang YO, Kim MY, Cho MY, Baik SK, Cho YZ, Kwon SO. Effect of bone marrow-derived mesenchymal stem cells on hepatic fibrosis in a thioacetamide-induced cirrhotic rat model.

BMC Gastroenterol. 2014;14:198-014-0198-6.

16. Seki A, Sakai Y, Komura T, et al. Adipose tissue-derived stem cells as a regenerative therapy for a mouse steatohepatitis-induced cirrhosis model. Hepatology. 2013;58(3):1133-1142.

17. Meier RP, Mahou R, Morel P, et al. Microencapsulated human mesenchymal stem cells decrease liver fibrosis in mice. J Hepatol. 2015;62(3):634-641.

18. Kantarcioglu M, Demirci H, Avcu F, et al. Efficacy of autologous mesenchymal stem cell transplantation in patients with liver cirrhosis. Turk J Gastroenterol. 2015;26(3):244-250. 19. Ma XR, Tang YL, Xuan M, Chang Z, Wang XY, Liang XH. Transplantation of autologous

mesenchymal stem cells for end-stage liver cirrhosis: A meta-analysis based on seven controlled trials. Gastroenterol Res Pract. 2015;2015:908275.

20. Horwitz EM, Le Blanc K, Dominici M, et al. Clarification of the nomenclature for MSC: The international society for cellular therapy position statement. Cytotherapy. 2005;7(5):393-395. 21. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev

Immunol. 2008;8(9):726-736.

22. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: An update. Cell

(22)

3

23. Lin PC, Chiou TW, Lin ZS, et al. A proposed novel stem cell therapy protocol for liver cirrhosis.

Cell Transplant. 2015;24(3):533-540.

24. Gimble JM, Bunnell BA, Frazier T, et al. Adipose-derived stromal/stem cells: A primer.

Organogenesis. 2013;9(1):3-10.

25. Wang Y, Lian F, Li J, et al. Adipose derived mesenchymal stem cells transplantation via portal vein improves microcirculation and ameliorates liver fibrosis induced by CCl4 in rats. J Transl

Med. 2012;10:133-5876-10-133.

26. Okura H, Soeda M, Morita M, et al. Therapeutic potential of human adipose tissue-derived multi-lineage progenitor cells in liver fibrosis. Biochem Biophys Res Commun. 2015;456(4):860-865.

27. Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R. Mesenchymal stem cell secretome: Toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci. 2017;18(9):10.3390/ijms18091852.

28. Banas A, Teratani T, Yamamoto Y, et al. IFATS collection: In vivo therapeutic potential of human adipose tissue mesenchymal stem cells after transplantation into mice with liver injury.

Stem Cells. 2008;26(10):2705-2712.

29. Kapur SK, Katz AJ. Review of the adipose derived stem cell secretome. Biochimie. 2013;95(12):2222-2228.

30. Gimble JM, Bunnell BA, Guilak F. Human adipose-derived cells: An update on the transition to clinical translation. Regen Med. 2012;7(2):225-235.

31. Togel F, Weiss K, Yang Y, Hu Z, Zhang P, Westenfelder C. Vasculotropic, paracrine actions of infused mesenchymal stem cells are important to the recovery from acute kidney injury. Am J

Physiol Renal Physiol. 2007;292(5):F1626-35.

32. Camussi G, Deregibus MC, Bruno S, Cantaluppi V, Biancone L. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 2010;78(9):838-848.

33. Kupcova Skalnikova H. Proteomic techniques for characterisation of mesenchymal stem cell secretome. Biochimie. 2013;95(12):2196-2211.

34. Parekkadan B, van Poll D, Megeed Z, et al. Immunomodulation of activated hepatic stellate cells by mesenchymal stem cells. Biochem Biophys Res Commun. 2007;363(2):247-252. 35. Wang J, Bian C, Liao L, et al. Inhibition of hepatic stellate cells proliferation by mesenchymal

stem cells and the possible mechanisms. Hepatol Res. 2009;39(12):1219-1228.

36. Lin N, Hu K, Chen S, et al. Nerve growth factor-mediated paracrine regulation of hepatic stellate cells by multipotent mesenchymal stromal cells. Life Sci. 2009;85(7-8):291-295. 37. Li T, Yan Y, Wang B, et al. Exosomes derived from human umbilical cord mesenchymal stem

cells alleviate liver fibrosis. Stem Cells Dev. 2013;22(6):845-854.

38. Hyun J, Wang S, Kim J, Kim GJ, Jung Y. MicroRNA125b-mediated hedgehog signaling influences liver regeneration by chorionic plate-derived mesenchymal stem cells. Sci Rep. 2015;5:14135.

39. Li J, Ghazwani M, Zhang Y, et al. miR-122 regulates collagen production via targeting hepatic stellate cells and suppressing P4HA1 expression. J Hepatol. 2013;58(3):522-528.

40. Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med. 2017;49(6):e346.

41. Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for parkinson’s disease therapy. J Control Release. 2015;207:18-30.

42. Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18(9):1606-1614.

43. Johnsen KB, Gudbergsson JM, Skov MN, Pilgaard L, Moos T, Duroux M. A comprehensive overview of exosomes as drug delivery vehicles - endogenous nanocarriers for targeted cancer therapy. Biochim Biophys Acta. 2014;1846(1):75-87.

(23)

44. van Balkom BW, Eisele AS, Pegtel DM, Bervoets S, Verhaar MC. Quantitative and qualitative analysis of small RNAs in human endothelial cells and exosomes provides insights into localized RNA processing, degradation and sorting. J Extracell Vesicles. 2015;4:26760.

45. Zhou W, Fong MY, Min Y, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25(4):501-515.

46. de Graaf IA, Olinga P, de Jager MH, et al. Preparation and incubation of precision-cut liver and intestinal slices for application in drug metabolism and toxicity studies. Nat Protoc. 2010;5(9):1540-1551.

47. Przybyt E, Krenning G, Brinker MG, Harmsen MC. Adipose stromal cells primed with hypoxia and inflammation enhance cardiomyocyte proliferation rate in vitro through STAT3 and Erk1/2. J Transl Med. 2013;11:39-5876-11-39.

48. Momen-Heravi F, Balaj L, Alian S, et al. Current methods for the isolation of extracellular vesicles. Biol Chem. 2013;394(10):1253-1262.

49. Scholten D, Trebicka J, Liedtke C, Weiskirchen R. The carbon tetrachloride model in mice. Lab

Anim. 2015;49(1 Suppl):4-11.

50. Bush VJ, Moyer TP, Batts KP, Parisi JE. Essential and toxic element concentrations in fresh and formalin-fixed human autopsy tissues. Clin Chem. 1995;41(2):284-294.

51. Moshage H, Casini A, Lieber CS. Acetaldehyde selectively stimulates collagen production in cultured rat liver fat-storing cells but not in hepatocytes. Hepatology. 1990;12(3 Pt 1):511-518. 52. Kinnman N, Francoz C, Barbu V, et al. The myofibroblastic conversion of peribiliary fibrogenic cells distinct from hepatic stellate cells is stimulated by platelet-derived growth factor during liver fibrogenesis. Lab Invest. 2003;83(2):163-173.

53. Shajari S, Laliena A, Heegsma J, Tunon MJ, Moshage H, Faber KN. Melatonin suppresses activation of hepatic stellate cells through RORalpha-mediated inhibition of 5-lipoxygenase. J

Pineal Res. 2015;59(3):391-401.

54. Urcan E, Haertel U, Styllou M, Hickel R, Scherthan H, Reichl FX. Real-time xCELLigence impedance analysis of the cytotoxicity of dental composite components on human gingival fibroblasts. Dent Mater. 2010;26(1):51-58.

55. Yu G, Wang LG, Han Y, He QY. clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 2012;16(5):284-287.

56. Ito K, Murphy D. Application of ggplot2 to pharmacometric graphics. CPT Pharmacometrics

Syst Pharmacol. 2013;2:e79.

57. Graaf IA, Groothuis GM, Olinga P. Precision-cut tissue slices as a tool to predict metabolism of novel drugs. Expert Opin Drug Metab Toxicol. 2007;3(6):879-898.

58. Westra IM, Oosterhuis D, Groothuis GM, Olinga P. Precision-cut liver slices as a model for the early onset of liver fibrosis to test antifibrotic drugs. Toxicol Appl Pharmacol. 2014;274(2):328-338.

59. Qin ZH, Xu JF, Qu JM, et al. Intrapleural delivery of MSCs attenuates acute lung injury by paracrine/endocrine mechanism. J Cell Mol Med. 2012;16(11):2745-2753.

60. Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev. 2009;20(5-6):419-427. 61. Hodge A, Lourensz D, Vaghjiani V, et al. Soluble factors derived from human amniotic

epithelial cells suppress collagen production in human hepatic stellate cells. Cytotherapy. 2014;16(8):1132-1144.

62. Huang B, Cheng X, Wang H, et al. Mesenchymal stem cells and their secreted molecules predominantly ameliorate fulminant hepatic failure and chronic liver fibrosis in mice respectively. J Transl Med. 2016;14:45-016-0792-1.

63. Tsai PC, Fu TW, Chen YM, et al. The therapeutic potential of human umbilical mesenchymal stem cells from wharton’s jelly in the treatment of rat liver fibrosis. Liver Transpl. 2009;15(5):484-495.

(24)

3

64. Thery C, Zitvogel L, Amigorena S. Exosomes: Composition, biogenesis and function. Nat Rev

Immunol. 2002;2(8):569-579.

65. Taraboletti G, D’Ascenzo S, Giusti I, et al. Bioavailability of VEGF in tumor-shed vesicles depends on vesicle burst induced by acidic pH. Neoplasia. 2006;8(2):96-103.

66. Taraboletti G, D’Ascenzo S, Borsotti P, Giavazzi R, Pavan A, Dolo V. Shedding of the matrix metalloproteinases MMP-2, MMP-9, and MT1-MMP as membrane vesicle-associated components by endothelial cells. Am J Pathol. 2002;160(2):673-680.

67. Sidhu SS, Mengistab AT, Tauscher AN, LaVail J, Basbaum C. The microvesicle as a vehicle for EMMPRIN in tumor-stromal interactions. Oncogene. 2004;23(4):956-963.

68. Parekkadan B, van Poll D, Suganuma K, et al. Mesenchymal stem cell-derived molecules reverse fulminant hepatic failure. PLoS One. 2007;2(9):e941.

69. Herrera MB, Fonsato V, Gatti S, et al. Human liver stem cell-derived microvesicles accelerate hepatic regeneration in hepatectomized rats. J Cell Mol Med. 2010;14(6B):1605-1618. 70. Bruno S, Grange C, Deregibus MC, et al. Mesenchymal stem cell-derived microvesicles protect

against acute tubular injury. J Am Soc Nephrol. 2009;20(5):1053-1067.

71. Pinzani M, Marra F, Carloni V. Signal transduction in hepatic stellate cells. Liver. 1998;18(1):2-13.

72. Marshall CJ. Specificity of receptor tyrosine kinase signaling: Transient versus sustained extracellular signal-regulated kinase activation. Cell. 1995;80(2):179-185.

73. Gentilini A, Marra F, Gentilini P, Pinzani M. Phosphatidylinositol-3 kinase and extracellular signal-regulated kinase mediate the chemotactic and mitogenic effects of insulin-like growth factor-I in human hepatic stellate cells. J Hepatol. 2000;32(2):227-234.

74. Shu M, Huang DD, Hung ZA, Hu XR, Zhang S. Inhibition of MAPK and NF-kappaB signaling pathways alleviate carbon tetrachloride (CCl4)-induced liver fibrosis in toll-like receptor 5 (TLR5) deficiency mice. Biochem Biophys Res Commun. 2016;471(1):233-239.

75. Liu Q, Wang CY, Liu Z, et al. Hydroxysafflor yellow A suppresses liver fibrosis induced by carbon tetrachloride with high-fat diet by regulating PPAR-gamma/p38 MAPK signaling.

Pharm Biol. 2014;52(9):1085-1093.

76. Yu Z, Jv Y, Cai L, et al. Gambogic acid attenuates liver fibrosis by inhibiting the PI3K/AKT and MAPK signaling pathways via inhibiting HSP90. Toxicol Appl Pharmacol. 2019;371:63-73. 77. Zhang Y, Huang XR, Wei LH, Chung AC, Yu CM, Lan HY. miR-29b as a therapeutic agent

for angiotensin II-induced cardiac fibrosis by targeting TGF-beta/Smad3 signaling. Mol Ther. 2014;22(5):974-985.

78. Machova Polakova K, Lopotova T, Klamova H, et al. Expression patterns of microRNAs associated with CML phases and their disease related targets. Mol Cancer. 2011;10:41-4598-10-41.

79. Tao H, Chen ZW, Yang JJ, Shi KH. MicroRNA-29a suppresses cardiac fibroblasts proliferation via targeting VEGF-A/MAPK signal pathway. Int J Biol Macromol. 2016;88:414-423.

80. Xiao J, Ho CT, Liong EC, et al. Epigallocatechin gallate attenuates fibrosis, oxidative stress, and inflammation in non-alcoholic fatty liver disease rat model through TGF/SMAD, PI3 K/akt/ FoxO1, and NF-kappa B pathways. Eur J Nutr. 2014;53(1):187-199.

81. Adachi M, Osawa Y, Uchinami H, Kitamura T, Accili D, Brenner DA. The forkhead transcription factor FoxO1 regulates proliferation and transdifferentiation of hepatic stellate cells. Gastroenterology. 2007;132(4):1434-1446.

82. Lopez-Berestein G, Kasi L, Rosenblum MG, et al. Clinical pharmacology of 99mTc-labeled liposomes in patients with cancer. Cancer Res. 1984;44(1):375-378.

83. Rank A, Nieuwland R, Crispin A, et al. Clearance of platelet microparticles in vivo. Platelets. 2011;22(2):111-116.

84. Liu D, Mori A, Huang L. Role of liposome size and RES blockade in controlling biodistribution and tumor uptake of GM1-containing liposomes. Biochim Biophys Acta. 1992;1104(1):95-101.

(25)

85. Ostro MJ, Cullis PR. Use of liposomes as injectable-drug delivery systems. Am J Hosp Pharm. 1989;46(8):1576-1587.

86. Poisson J, Lemoinne S, Boulanger C, et al. Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J Hepatol. 2017;66(1):212-227.

87. Wisse E. An electron microscopic study of the fenestrated endothelial lining of rat liver sinusoids. J Ultrastruct Res. 1970;31(1):125-150.

88. Senior JH. Fate and behavior of liposomes in vivo: A review of controlling factors. Crit Rev

Ther Drug Carrier Syst. 1987;3(2):123-193.

89. Woodle MC, Lasic DD. Sterically stabilized liposomes. Biochim Biophys Acta. 1992;1113(2):171-199.

90. Wiklander OP, Nordin JZ, O’Loughlin A, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316.

(26)

3

SUPPLEMENTARY TABLES

Supplementary Table S1. Primers and probes used for real-time quantitative PCR analysis

(Taqman protocol).

Supplementary Table S2. Mouse primers used for real-time quantitative PCR analysis

(27)

Supplementary table S3. miRNA content of hASC and hASC-derived EVs

MiRNAs hASC (absolute

values) hASC (relative values) EVs (absolute values) EVs (relative values)

hsa-miR-21-5p 1957493 15.417559 1618929 20.70720178 hsa-miR-181a-5p 1132708 8.921407343 1086132 13.89236618 hsa-miR-142-5p 723206 5.696097599 835995 10.69294401 hsa-miR-22-3p 1257196 9.901896717 653737 8.361740366 hsa-miR-150-5p 364101 2.867723487 367610 4.701981647 hsa-miR-146b-5p 393219 3.097061974 313786 4.01353612 hsa-let-7f-5p 449131 3.537434715 285667 3.653875007 hsa-miR-26a-5p 378414 2.980455191 234806 3.003328263 hsa-miR-92a-3p 321236 2.530111211 179935 2.301490894 hsa-miR-16-5p 181177 1.426981904 175435 2.243932837 hsa-let-7g-5p 131485 1.035598976 133175 1.703398727 hsa-miR-21-3p 199794 1.573612669 132266 1.691771999 hsa-miR-30e-5p 124888 0.983639844 124599 1.59370586 hsa-let-7a-5p 382358 3.011518828 124648 1.594332603 hsa-miR-191-5p 162206 1.277562973 112377 1.437378177 hsa-miR-155-5p 143530 1.130467513 91447 1.169669257 hsa-miR-186-5p 90827 0.71536942 87732 1.122151883 hsa-miR-142-3p 55031 0.433433831 71319 0.912218463 hsa-let-7i-5p 188050 1.48111486 72879 0.932171923 hsa-miR-342-3p 54181 0.426739081 64070 0.819498828 hsa-miR-27a-3p 81227 0.639758132 63558 0.81295 hsa-miR-146a-5p 79218 0.623934895 57334 0.733340812 hsa-miR-28-3p 78058 0.614798531 52145 0.666969976 hsa-miR-29a-3p 60633 0.477556168 49823 0.637270019 hsa-miR-103a-3p 54663 0.430535398 46114 0.589829389 hsa-miR-26b-5p 39686 0.312573913 42524 0.54391085 hsa-miR-181b-5p 52766 0.415594293 40152 0.513571358 hsa-miR-10a-5p 453695 3.573381582 37633 0.481351637 hsa-miR-27b-3p 174210 1.372108587 36171 0.462651664 hsa-miR-25-3p 38499 0.303224892 35089 0.448812149 hsa-miR-148a-3p 64736 0.509872117 31416 0.401831983 hsa-miR-30c-5p 28691 0.22597536 26739 0.342009976 hsa-miR-221-3p 53521 0.421540805 26405 0.337737889 hsa-miR-107 28437 0.223974811 25828 0.330357667 hsa-miR-101-3p 28462 0.224171716 25533 0.326584417 hsa-miR-192-5p 21288 0.167668031 21655 0.276982162 hsa-miR-423-3p 46364 0.365171015 20595 0.263424042 hsa-miR-140-3p 20002 0.157539268 17595 0.225052004 hsa-miR-30d-5p 37024 0.291607533 16368 0.20935784 hsa-miR-146b-3p 17487 0.137730686 15578 0.199253203 hsa-miR-15a-5p 15285 0.120387347 14893 0.190491588 hsa-miR-98-5p 22695 0.17874981 14300 0.182906715 hsa-miR-222-3p 214607 1.690282461 12068 0.154357919 hsa-miR-423-5p 37389 0.294482337 10930 0.139802126 hsa-miR-92b-3p 79682 0.62758944 9389 0.120091689 hsa-miR-93-5p 10797 0.08503907 9423 0.120526572 hsa-miR-361-3p 9065 0.071397534 9398 0.120206805 hsa-miR-23a-3p 17727 0.139620968 8921 0.114105651 hsa-let-7d-5p 13519 0.106478021 8698 0.111253329 hsa-miR-34a-5p 6799 0.053550119 8312 0.106316127 hsa-miR-181a-3p 8398 0.066144124 7799 0.099754508 hsa-miR-29c-3p 6473 0.050982486 7426 0.094983585 hsa-miR-484 8933 0.070357878 7320 0.093627773 hsa-miR-1307-5p 6396 0.05037602 7259 0.092847542

(28)

3

hsa-miR-16-2-3p 8225 0.064781546 7419 0.09489405 hsa-miR-106b-3p 7959 0.062686483 7303 0.093410332 hsa-miR-941 7685 0.060528411 6591 0.084303368 hsa-miR-181c-5p 7283 0.057362188 6647 0.085019646 hsa-miR-378a-3p 7406 0.058330958 6393 0.081770813 hsa-miR-320a-3p 11803 0.092962503 6162 0.078816166 hsa-miR-28-5p 7914 0.062332055 5770 0.07380222 hsa-miR-128-3p 7567 0.059599022 5734 0.073341756 hsa-miR-425-5p 5486 0.0432087 5242 0.067048741 hsa-miR-19b-3p 5112 0.040263011 4853 0.062073167 hsa-miR-30e-3p 4835 0.03808131 4850 0.062034795 hsa-miR-148b-3p 9547 0.07519385 4504 0.05760922 hsa-miR-486-5p 10178 0.080163717 4247 0.054322015 hsa-miR-454-3p 3277 0.025810228 4019 0.05140574 hsa-miR-3615 4474 0.035238011 3903 0.049922022 hsa-miR-363-3p 3472 0.027346082 3929 0.050254579 hsa-miR-769-5p 6328 0.04984044 3647 0.046647608 hsa-miR-23b-3p 7679 0.060481154 3412 0.043641798 hsa-miR-182-5p 4311 0.033954194 3576 0.045739469 hsa-miR-20a-5p 3292 0.025928371 3067 0.039229014 hsa-miR-30b-5p 4076 0.032103293 2584 0.033051116 hsa-miR-15b-5p 3550 0.027960424 2746 0.035123206 hsa-miR-130b-3p 4436 0.034938716 2746 0.035123206 hsa-miR-1307-3p 3660 0.028826803 2760 0.035302275 hsa-miR-29b-3p 2995 0.023589147 2666 0.034099951 hsa-miR-223-3p 2206 0.017374844 2567 0.032833674 hsa-miR-421 3052 0.024038089 2493 0.031887164 hsa-miR-374a-3p 3112 0.024510659 2508 0.032079024 hsa-miR-339-3p 2689 0.021179037 2385 0.03050577 hsa-miR-24-3p 4973 0.039168222 2535 0.032424372 hsa-miR-532-5p 3201 0.025211639 2386 0.030518561 hsa-miR-424-3p 4159 0.032757015 2026 0.025913916 hsa-miR-301a-3p 2234 0.017595377 2014 0.025760428 hsa-miR-32-5p 1454 0.01145196 1814 0.023202292 hsa-miR-210-3p 2381 0.018753175 2116 0.027065078 hsa-miR-652-3p 1776 0.013988088 1857 0.023752292 hsa-miR-342-5p 2064 0.016256427 1918 0.024532523 hsa-miR-124-3p 1562 0.012302587 1840 0.02353485 hsa-miR-106b-5p 2210 0.017406349 1803 0.023061595 hsa-miR-17-5p 2238 0.017626881 1754 0.022434852 hsa-miR-19a-3p 2160 0.01701254 1798 0.022997642 hsa-miR-141-3p 1293 0.010183895 1539 0.019684856 hsa-miR-1285-3p 1632 0.012853919 1567 0.020042995 hsa-miR-197-3p 2380 0.018745298 1649 0.02109183 hsa-miR-378c 1217 0.009585306 1515 0.019377879 hsa-miR-99b-5p 40813 0.321450363 1488 0.019032531 hsa-miR-132-3p 5580 0.043949061 1397 0.017868579 hsa-miR-150-3p 1271 0.010010619 1414 0.018086021 hsa-miR-9985 1055 0.008309365 1194 0.015272071 hsa-miR-10399-3p 1157 0.009112735 1266 0.016193 hsa-miR-130a-3p 4989 0.039294241 1199 0.015336025 hsa-miR-501-3p 1397 0.011003018 1166 0.014913932 hsa-miR-125a-5p 40532 0.319237158 1089 0.01392905 hsa-miR-10399-5p 797 0.006277312 1097 0.014031375 hsa-miR-1246 1385 0.010908503 1087 0.013903468 hsa-miR-500a-3p 2764 0.02176975 1030 0.0131744 hsa-miR-589-5p 2432 0.01915486 965 0.012343006 hsa-miR-660-5p 1662 0.013090204 991 0.012675563

(29)

hsa-let-7b-5p 37884 0.298381044 894 0.011434867 hsa-miR-221-5p 7758 0.061103372 926 0.011844169 hsa-miR-17-3p 1632 0.012853919 823 0.010526729 hsa-miR-374a-5p 847 0.006671121 910 0.011639518 hsa-miR-671-3p 2630 0.020714342 895 0.011447658 hsa-miR-7706 1111 0.008750431 836 0.010693008 hsa-miR-3614-5p 1475 0.011617359 918 0.011741844 hsa-miR-151a-3p 24812 0.195423674 841 0.010756961 hsa-miR-330-5p 818 0.006442712 777 0.009938358 hsa-miR-361-5p 1430 0.011262931 865 0.011063938 hsa-miR-625-3p 1283 0.010105134 824 0.01053952 hsa-miR-223-5p 339 0.002670024 708 0.009055801 hsa-miR-331-3p 923 0.00726971 757 0.009682544 hsa-miR-194-5p 509 0.004008973 718 0.009183708 hsa-miR-26b-3p 1245 0.009805839 725 0.009273243 hsa-miR-148b-5p 1205 0.009490792 751 0.0096058 hsa-miR-27a-5p 3219 0.02535341 700 0.008953476 hsa-miR-766-3p 456 0.003591536 629 0.008045337 hsa-miR-151a-5p 18400 0.144921635 616 0.007879058 hsa-let-7d-3p 1973 0.015539695 607 0.007763942 hsa-let-7i-3p 643 0.005064381 649 0.008301151 hsa-miR-22-5p 753 0.00593076 619 0.007917431 hsa-miR-148a-5p 2222 0.017500863 558 0.007137199 hsa-miR-320b 1168 0.009199373 601 0.007687198 hsa-miR-140-5p 622 0.004898981 597 0.007636036 hsa-let-7c-5p 3830 0.030165753 503 0.006433712 hsa-miR-424-5p 1905 0.015004115 594 0.007597664 hsa-miR-143-3p 426115 3.356156653 573 0.007329059 hsa-miR-374b-5p 701 0.005521199 576 0.007367431 hsa-miR-328-3p 1164 0.009167869 536 0.006855804 hsa-miR-24-2-5p 1573 0.012389225 621 0.007943012 hsa-miR-1273h-3p 463 0.003646669 472 0.006037201 hsa-miR-12136 1182 0.00930964 486 0.00621627 hsa-miR-10b-5p 426981 3.362977421 437 0.005589527 hsa-miR-1843 639 0.005032876 498 0.006369758 hsa-miR-7-1-3p 452 0.003560031 471 0.00602441 hsa-miR-598-3p 442 0.00348127 498 0.006369758 hsa-miR-126-5p 640 0.005040753 499 0.006382549 hsa-miR-744-5p 3466 0.027298825 432 0.005525573 hsa-miR-450b-5p 1475 0.011617359 482 0.006165107 hsa-miR-345-5p 1349 0.010624961 415 0.005308132 hsa-miR-130b-5p 1473 0.011601607 442 0.00565348 hsa-miR-450a-5p 718 0.005655094 447 0.005717434 hsa-miR-576-5p 602 0.004741458 386 0.004937202 hsa-miR-378d 346 0.002725157 432 0.005525573 hsa-miR-330-3p 482 0.003796317 333 0.004259296 hsa-miR-4485-3p 413 0.003252861 402 0.005141853 hsa-miR-18a-5p 471 0.003709679 437 0.005589527 hsa-miR-5701 609 0.004796591 358 0.004579063 hsa-miR-128-1-5p 311 0.002449491 367 0.004694179 hsa-miR-454-5p 418 0.003292241 323 0.004131389 hsa-miR-548bc 281 0.002213205 309 0.00395232 hsa-miR-625-5p 601 0.004733582 408 0.005218597 hsa-miR-7705 288 0.002268339 301 0.003849994 hsa-miR-590-3p 424 0.003339499 310 0.003965111 hsa-miR-212-3p 661 0.005206152 295 0.00377325 hsa-miR-181c-3p 649 0.005111638 320 0.004093017 hsa-miR-3176 329 0.002591262 299 0.003824413

Referenties

GERELATEERDE DOCUMENTEN

Veel van deze soorten zijn niet goed aangepast aan het duinvalleimilieu, althans zullen de competitie met beter aangepaste soorten verliezen, zodat na 5-10 jaren een nieuwe

De overige kuilen leverden geen vondstmateriaal op, maar zijn vermoedelijk gezien de vele overeenkomsten in vorm, vulling en aflijning en hun ligging onder de Ap2 horizont overwegend

It is shown that the surface segregation behaviour in atomically clean Pt-Rh alloys can be understood quantitatively by taking into account the difference in

At sufficient rpm of the rotor the water stream closes the valve by the under pressure caused by the water acceleration through the slit between piston valve

Decoding therapeutic roles of adipose tissue-derived stromal cells and their extracellular vesicles in liver disease..

The overall aim of this thesis is to investigate the therapeutic potential of extracellular vesicles from human adipose-derived stromal cells (hASC-derived EVs) in the treatment

have been applied to analyze drug metabolism 37 and served as models of drug- induced cholestatic 38 , metabolic 39 , and fibrotic 35 liver disease, but this is the first-

Hoewel de aankoop frequentie onder deze light users uiterst marginaal is met gemiddeld 11 biologische aankopen per huishouden per jaar, vormt deze groep een belangrijke kans om de