Acta Biomaterialia - SOCRATES

13
Full length article Mesenchymal stem cell exosomes enhance periodontal ligament cell functions and promote periodontal regeneration Jacob Ren Jie Chew a , Shang Jiunn Chuah a , Kristeen Ye Wen Teo a , Shipin Zhang a , Ruenn Chai Lai b , Jia Hui Fu a , Lum Peng Lim a , Sai Kiang Lim b,c , Wei Seong Toh a,d,a Faculty of Dentistry, National University of Singapore, Singapore b Institute of Medical Biology, Agency for Science, Technology and Research, Singapore c Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore d Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore article info Article history: Received 5 December 2018 Received in revised form 8 March 2019 Accepted 11 March 2019 Available online 13 March 2019 Keywords: Mesenchymal stem cells Exosomes Bone Periodontal ligament Regeneration abstract Mesenchymal stem cells (MSCs) are potential therapeutics for the treatment of periodontal defects. It is increasingly accepted that MSCs mediate tissue repair through secretion of trophic factors, particularly exosomes. Here, we investigated the therapeutic effects of human MSC exosome-loaded collagen sponge for regeneration of surgically created periodontal intrabony defects in an immunocompetent rat model. We observed that relative to control rats, exosome-treated rats repaired the defects more efciently with regeneration of periodontal tissues including newly-formed bone and periodontal ligament (PDL). We also observed that concomitant with this, there was increased cellular inltration and proliferation. We therefore postulated that MSC exosomes enhanced regeneration through increased cellular mobilisa- tion and proliferation. Using PDL cell cultures, we demonstrated that MSC exosomes could increase PDL cell migration and proliferation through CD73-mediated adenosine receptor activation of pro-survival AKT and ERK signalling. Inhibition of AKT or ERK phosphorylation suppressed PDL cell migration and pro- liferation. Our ndings demonstrated for the rst time that MSC exosomes enhance periodontal regener- ation possibly by increasing PDL migration and proliferation. This study suggests that MSC exosome is a viable ready-to-use and cell-free MSC therapeutic for the treatment of periodontal defects. Statement of signicance Mesenchymal stem cell (MSC) therapies have demonstrated regenerative potential for the treatment of periodontal defects. However, translation of cellular therapies is hampered by challenges in maintaining optimal cell vitality and viability from manufacturing and storage to nal delivery to patients. Although the use of MSCs for tissue repair was rst predicated on their differentiation potential, the therapeutic efcacy of MSCs has increasingly been attributed to its paracrine secretion, particularly exosomes or small extracellular vesicles. In this study, MSC exosome-loaded collagen sponge enhanced periodontal regeneration in an immunocompetent rat periodontal defect model without any obvious adverse effects. These ndings provide the basis for future development of MSC exosomes as a cell-free strategy for peri- odontal regeneration. 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved. 1. Introduction Severe periodontitis is the sixth most prevalent disease [1]. It is the primary cause of irreversible destruction of the teeth- supporting periodontium, which is a complex structure composed of periodontal ligament (PDL), cementum and alveolar bone. If poorly managed or untreated, the disease often leads to tooth loss [2]. Current treatment modalities for periodontitis are effective in reducing/arresting the pathogenic condition but fail to predictably regenerate the lost periodontal structures in many clinical situa- tions [3]. Many of the common disease presentations, such as one-walled intrabony defects, class III and interproximal furcal defects are not amenable to current regenerative therapy, thus https://doi.org/10.1016/j.actbio.2019.03.021 1742-7061/2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved. Corresponding author at: Faculty of Dentistry, National University of Singapore, 9 Lower Kent Ridge Road, #10-01, National University Centre for Oral Health, 119085, Singapore. E-mail address: [email protected] (W.S. Toh). Acta Biomaterialia 89 (2019) 252–264 Contents lists available at ScienceDirect Acta Biomaterialia journal homepage: www.elsevier.com/locate/actabiomat

Transcript of Acta Biomaterialia - SOCRATES

Page 1: Acta Biomaterialia - SOCRATES

Full length article

Mesenchymal stem cell exosomes enhance periodontal ligament cellfunctions and promote periodontal regeneration

Jacob Ren Jie Chew a, Shang Jiunn Chuah a, Kristeen Ye Wen Teo a, Shipin Zhang a, Ruenn Chai Lai b,Jia Hui Fu a, Lum Peng Lim a, Sai Kiang Lim b,c, Wei Seong Toh a,d,⇑a Faculty of Dentistry, National University of Singapore, Singaporeb Institute of Medical Biology, Agency for Science, Technology and Research, SingaporecDepartment of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapored Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore

a r t i c l e i n f o

Article history:Received 5 December 2018Received in revised form 8 March 2019Accepted 11 March 2019Available online 13 March 2019

Keywords:Mesenchymal stem cellsExosomesBonePeriodontal ligamentRegeneration

a b s t r a c t

Mesenchymal stem cells (MSCs) are potential therapeutics for the treatment of periodontal defects. It isincreasingly accepted that MSCs mediate tissue repair through secretion of trophic factors, particularlyexosomes. Here, we investigated the therapeutic effects of human MSC exosome-loaded collagen spongefor regeneration of surgically created periodontal intrabony defects in an immunocompetent rat model.We observed that relative to control rats, exosome-treated rats repaired the defects more efficiently withregeneration of periodontal tissues including newly-formed bone and periodontal ligament (PDL). Wealso observed that concomitant with this, there was increased cellular infiltration and proliferation.We therefore postulated that MSC exosomes enhanced regeneration through increased cellular mobilisa-tion and proliferation. Using PDL cell cultures, we demonstrated that MSC exosomes could increase PDLcell migration and proliferation through CD73-mediated adenosine receptor activation of pro-survivalAKT and ERK signalling. Inhibition of AKT or ERK phosphorylation suppressed PDL cell migration and pro-liferation. Our findings demonstrated for the first time that MSC exosomes enhance periodontal regener-ation possibly by increasing PDL migration and proliferation. This study suggests that MSC exosome is aviable ready-to-use and cell-free MSC therapeutic for the treatment of periodontal defects.

Statement of significance

Mesenchymal stem cell (MSC) therapies have demonstrated regenerative potential for the treatment ofperiodontal defects. However, translation of cellular therapies is hampered by challenges in maintainingoptimal cell vitality and viability from manufacturing and storage to final delivery to patients. Althoughthe use of MSCs for tissue repair was first predicated on their differentiation potential, the therapeuticefficacy of MSCs has increasingly been attributed to its paracrine secretion, particularly exosomes orsmall extracellular vesicles. In this study, MSC exosome-loaded collagen sponge enhanced periodontalregeneration in an immunocompetent rat periodontal defect model without any obvious adverse effects.These findings provide the basis for future development of MSC exosomes as a cell-free strategy for peri-odontal regeneration.

� 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction

Severe periodontitis is the sixth most prevalent disease [1]. It isthe primary cause of irreversible destruction of the teeth-

supporting periodontium, which is a complex structure composedof periodontal ligament (PDL), cementum and alveolar bone. Ifpoorly managed or untreated, the disease often leads to tooth loss[2]. Current treatment modalities for periodontitis are effective inreducing/arresting the pathogenic condition but fail to predictablyregenerate the lost periodontal structures in many clinical situa-tions [3]. Many of the common disease presentations, such asone-walled intrabony defects, class III and interproximal furcaldefects are not amenable to current regenerative therapy, thus

https://doi.org/10.1016/j.actbio.2019.03.0211742-7061/� 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

⇑ Corresponding author at: Faculty of Dentistry, National University of Singapore,9 Lower Kent Ridge Road, #10-01, National University Centre for Oral Health,119085, Singapore.

E-mail address: [email protected] (W.S. Toh).

Acta Biomaterialia 89 (2019) 252–264

Contents lists available at ScienceDirect

Acta Biomaterialia

journal homepage: www.elsevier .com/locate /ac tabiomat

Page 2: Acta Biomaterialia - SOCRATES

posing an unmet need for new therapeutics to address theseconditions.

In recent years, mesenchymal stem cells (MSCs) have emergedas a promising therapy for periodontal regeneration in animalstudies [4,5] and in human studies [6]. However, this is a cell-based therapy that as with all cellular therapies, incurs significantoperational and logistical challenges in the manufacture, deliveryand storage of cells for transplantation. The use of MSCs for tissuerepair was originally rationalized on their differentiation potentialto generate various cell types that could replace the lost cells ininjured or dead tissues. However, it is increasingly evident thatMSCs secrete a wide spectrum of factors that have the capacitiesto alleviate tissue injury and promote repair and regeneration[7]. On this note, conditioned media of PDL stem cells and bonemarrow MSCs had been shown in animal studies to promoteperiodontal regeneration [8,9]. Among these secreted factors,exosomes which are nano-sized membrane vesicles of about50–200 nm have been shown to replicate the wide-ranging MSCtherapeutic efficacies in animal models for myocardial ischemiareperfusion injury [10], cutaneous wound [11], graft-versus-host-disease (GVHD) [12], drug-induced hepatic injury [13], and morerecently bone and cartilage injuries [14,15]. However, the implica-tion of these findings to periodontal regeneration remains to bedetermined.

Here, we investigate if MSC exosomes could facilitate periodon-tal regeneration. Specifically, we evaluated the effects of humanMSC exosomes in an immunocompetent rat model with surgicallycreated periodontal intrabony defects. We also evaluated theeffects of MSC exosomes on PDL cell migration and proliferation,and associated PDL expression of genes relevant to periodontalregeneration.

2. Materials and methods

2.1. Preparation of MSC exosomes

The exosome preparation followed a standard operating proce-dure that was based on the principles first described in 2010 [10].Briefly, MSC conditioned medium was size-fractionated, concen-trated 50� by tangential flow filtration (Sartorius, Gottingen,Germany, MWCO 100 kDa) and then characterized. Each batch ofexosome preparation was ascertained to have similarly sized par-ticles of 100–200 nm, a flotation density of 1.10–1.19 g/ml, andpresence of exosome-associated markers including CD81, ALIXand TSG101 [10,14]. The exosomes were then 0.22 mm filtered(Merck Millipore, Billerica, MA, USA) and stored at �20 �C untiluse. The exosomes were assayed for protein concentration usinga NanoOrange Protein Quantification Kit (Thermo Fisher Scientific),as per manufacturer’s instruction.

2.2. Isolation and culture of periodontal ligament (PDL) cells

Rat PDL cells were obtained from the extracted incisors of 8-week-old Sprague Dawley (SD) rats following an established proto-col with modification [16]. Briefly, the gingival soft tissues werefirst excised and the incisors were then extracted using forceps.The incisors were washed with phosphate-buffered saline (PBS)supplemented with 2% penicillin/streptomycin (PS; Thermo FisherScientific). Tissues from the coronal and apical thirds of the incisorswere removed. The tooth then underwent digestion in 2 mg/ml ofdispase II and collagenase I (Worthington, Lakewood, NJ, USA)overnight at 37 �C [17]. The cell suspension was passed througha 40-mm cell strainer (Corning�, Corning, NY, USA) to dispersethe cells into single cells before seeding at a density of 2 � 104 -cells/cm2. The culture was maintained in complete culture medium

composed of DMEM-F12 supplemented with 10% FBS, 25 mg/mlascorbic acid 2-phosphate (AA2P, Sigma, St. Louis, MO, USA) and1% PS under a humidified atmosphere of 5% CO2 at 37 �C. The cul-ture medium was changed every alternate day until confluencewas reached. Cells were then detached using TrypLe (ThermoFisher Scientific) and further sub-cultured to passages (P) 3 and 4for in vitro experiments.

2.3. Cellular uptake of exosomes

Exosomes were first labelled with Alexafluor 488 dye using aprotein labelling kit (Thermo Fisher Scientific) according to themanufacturer’s instruction. Briefly, 100 mg exosomes were incu-bated with 10 ml of Alexafluor 488 dye in PBS in a final volume of100 ml dye reaction mixture for 1 h at ambient temperature of20 �C before transferred into the Biospin P6 gel column and cen-trifuged at 1000 g for 4 min to remove the free dye. To determinethe kinetics of exosome uptake, PDL cells were incubated with 5mg/ml of Alexafluor 488-labelled exosomes and observed at 0.25,0.5, 1, 3 and 6 h by confocal microscopy (FV300, Olympus, Tokyo,Japan). In some experiments, PDL cells were labelled with Cell-MaskTM Deep Red plasma membrane stain (Thermo FisherScientific).

2.4. Cell migration

The migration of PDL cells in response to exosome treatmentwas assessed using a transwell system, as previously described[18]. Briefly, 5 � 104 cells in 300 ml of low serum culture medium(DMEM-F12 supplemented with 0.5% FBS and 1% PS) were placedin the upper chamber and 1, 5 or 10 mg/ml exosomes or vehiclecontrol (PBS) were added to the lower chambers. After 16 h, theupper surface of the transwell filters was swabbed free of cells.Cells on the underside of the filter were then fixed in 4% (v/v)paraformaldehyde and stained with haematoxylin and eosin(HE). The cells in five randomly-selected fields at 100� magnifica-tion were counted. The percentage of cell migration was calculatedby normalizing the number of cells on the underside of the tran-swell membrane to the initial number of cells seeded and com-pared against that of the PBS-treated vehicle control which wasset at 100%.

2.5. Cell viability and proliferation

Cells were seeded at 5000 cells/well in 96-well plates in com-plete culture medium for 16 h before the medium was changedto low serum culture medium for 24 h. The cells were then treatedwith 1, 5 and 10 mg/ml of exosomes or PBS as vehicle control. At 4,24, 48 and 72 h, cell viability and total DNA content were mea-sured. Cell viability was measured using the MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay kit (Promega, Madison, WI, USA) following themanufacturer’s specifications. 20 ml of MTS reagent was added toeach well and incubated at 37 �C for 2 h. Absorbance readings werethen taken at 490 nm and 650 nm (reference) using a microplatereader (Infinite� 200 PRO, TecanTM, Männedorf, Switzerland). Thevalue for cells treated with PBS at 4 h was set at 100% and all othervalues were normalised to this value. For measurement of DNAcontent, the cells in each well were lysed using 50 ml CelLytic M celllysis buffer (Sigma) and DNA concentration reflective of the cellnumber was measured using the Quant-iTTM Picogreen� dsDNAassay kit (Thermo Fisher Scientific) according to the manufac-turer’s instructions. The fluorescence readings were then taken atexcitation/emission wavelengths of 480/520 nm using a micro-plate reader.

J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264 253

Page 3: Acta Biomaterialia - SOCRATES

2.6. Exosome treatment and inhibitor studies

PDL cells were seeded at a density of 2 � 104 cells/cm2 andcultured for 16 h before the medium was changed to a low serumculture medium for 24 h, and then treated with 5 mg/ml ofexosomes or vehicle (PBS) as control for over 48 h. To investigatethe involvement of adenosine receptor and activation of AKT andERK pathways, PDL cells were pre-treated with 1 mM theophylline(a non-selective adenosine receptor antagonist; Sigma) [19], 1 mMwortmannin (inhibitor of PI3K/AKT pathway), 10 mM U0126 (inhi-bitor of MAPK/ERK pathway) (Cell Signaling Technology, Danvers,MA, USA), or equivalent volume of distilled water or DMSO(dimethyl sulfoxide) as vehicle controls for 1 h, before treatmentwith 5 mg/ml exosomes or PBS vehicle for over 48 h. Followingtreatment, PDL cells were rinsed with PBS and then harvested forfurther analysis. All in vitro experiments were performed intriplicates (n = 3) in at least two independent experiments.

2.7. Quantitative reverse transcription polymerase chain reaction(RT-PCR)

Total RNA was isolated from PDL cultures using PureLink� RNAMini kit (Thermo Fisher Scientific) according to manufacturer’sinstructions. The RNA was then reverse transcribed to cDNA usingiScriptTM reverse transcription Supermix (Bio-Rad Laboratories,Hercules, CA, USA). The cDNA samples were then amplified withiTaqTM Universal SYBR� Green Supermix (Bio-Rad) and primers(Table 1) using the CFX ConnectTM real-time PCR system (Bio-Rad). The PCR cycling condition comprised an initial denaturationat 95 �C for 30 s followed by 40 cycles of amplification consistingof a 15 s denaturation at 95 �C and a 30 s extension at 60 �C. Rela-tive mRNA expression levels were normalized against glyceralde-hyde 3-phosphate dehydrogenase (GAPDH) mRNA, and calculatedusing the comparative DCT method, and finally expressed as foldchanges [20].

2.8. Western blot hybridization

Western blot hybridization was performed using standard pro-tocols. Briefly, proteins were denatured, separated on 4–12% poly-acrylamide gels (Thermo Fisher Scientific), electro-blotted onto anitrocellulose membrane (GE Healthcare), probed with primaryantibody followed by incubation with horseradish peroxidase(HRP)-coupled secondary antibody against the primary antibody.

The primary antibodies used in this study were rabbit anti-AKT(pan) (1:1000), anti-phospho-AKT (Ser473) (1:1000), anti-ERK1/2(1:1000) and anti-phospho-ERK1/2 (Thr202/Tyr204) (1:1000) (CellSignaling Technology), and mouse anti-GAPDH (1:10000, Abcam,Cambridge, MA, USA). After incubating with the appropriate HRP-coupled secondary antibodies (GE Healthcare), the protein bandswere visualized by incubating with SuperSignal West Pico Chemi-luminescent Substrate (Thermo Fisher Scientific) and then docu-mented using the ChemiDocTM MP System (Bio-Rad). Finally, thebands were quantified with Image LabTM software (Bio-Rad).

2.9. Exosome incorporation into collagen sponges

Exosome-loaded collagen sponges were gifts from ParacrineTherapeutics, Biopolis, Singapore. Bovine collagen I/III sponges(HealiAid�, Maxigen Biotech Inc. Taoyuan City, Taiwan) were cutinto pieces of 2 mm width (W) � 2 mm length (L) � 1 mm depth(D) for exosome loading. The exosome-loaded collagen spongewhich is referred to as CS/Exosome hereafter was loaded with40 mg exosomes while the control collagen sponge referred to asCS/Control was similarly treated as per exosome loading but with-out exosomes. The sponges were lyophilized and kept at 4 �C untiluse.

2.10. Exosome loading efficiency and release

To determine the MSC exosome loading efficiency, theexosome-loaded sponges were each rehydrated with 200 ll PBSin a 96-well plate and incubated at ambient temperature of 20 �Cfor 10 min. The sponges were pressed repeatedly (�10 times) toextract the exosomes. To determine the release profile, theexosome-loaded sponges were each incubated in 200 ll of PBS atambient temperature of 20 �C and supernatant samples were har-vested at 10 min, 24 and 48 h. Supernatant samples were assayedfor the cholera toxin B chain (CTB)-CD81 assay as previouslydescribed [21]. CTB-CD81 assay had been reported to specificallydetect intact CTB-binding CD81 expressing MSC exosomes [21].

Briefly, CTB-CD81 assay was performed by incubating 20 llsupernatant sample with 0.25 lg biotinylated CTB in PBS in a finalvolume of 100 ll for 30 min, followed by immobilization using40 ll of pre-washed streptavidin-conjugated polystyrene beads(Spherotech, Inc. Lake Forest, IL, USA). The beads were then washedtwice with 200 ll PBS, incubated with 50 ll of anti-CD81 antibod-ies (1:500, Santa Cruz Biotech, Dallas, TX, USA), washed and incu-bated again with HRP-conjugated goat anti-mouse secondaryantibodies (1:5000, Santa Cruz Biotech). HRP activity was deter-mined by incubating with Amplex� Red Substrate (Thermo FisherScientific) as per manufacturer’s protocol. The loading efficiencyand release were determined by normalising the levels of CTB-CD81 to the initial level of CTB-CD81 present in 40 lg exosomesand expressed as a percentage.

2.11. Rat periodontal defect model

All procedures were performed according to the InstitutionalAnimal Care and Use Committee at National University of Singa-pore under protocol number: R17-0106. Eighteen 10-week oldmale SD rats weighting 289 to 413 g (mean: 306.2 g) were usedin this study. The animals were randomly assigned to three groups:defects left untreated (Untreated; n = 12), defects treated with col-lagen sponge containing 40 mg exosomes (CS/Exosome; n = 12) anddefects treated with control collagen sponge without exosome (CS/Control; n = 12). Under general anaesthesia, bilateral periodontaldefects of size 2 � 2 � 1.5 mm (W � L � D) were created on themesial surface of the first molar, as previously described [22,23](Fig. 1).

Table 1Primer sequence used for quantitative RT-PCR.

Gene Primer sequence Product size

FGF-2 F: CGGTACCTGGCTATGAAGGA 82 bpR: CCAGGCGTTCAAAGAAGAAA

PCNA F: CAACTTGGAATCCCAGAACAGGAG 82 bpR: TAAGGTCCCGGCATATACGTGC

COL1A1 F: TCCAGGGCTCCAACGAGA 58 bpR: CTGTAGGTGAATCGACTGTTGC

POSTN F: AGGGTCCTACACATACTTCG 149 bpR: GGTCCTTGGTTAGCATTCTC

TGF-ß1 F: AAGAAGTCACCCGCGTGCTA 70 bpR: TGTGTGATGTCTTTGGTTTTGTCA

IGF-1 F: ATAGACTCCGGCGCTACCTC 70 bpR: CCAGGGGATCTGGGTAGG

BAX F: TGGTTGCCCTCTTCTACTTTG 193 bpR: GTCACTGTCTGCCATGTGGG

Survivin F: AACTGGCCCTTCCTGGAG 89 bpR: TCAGGCTCATTCTCGGTAG

Bcl-2 F: GCCTTCTTTGAGTTCGGTG 199 bpR: GCCAGGAGAAATCAAACAGAG

GAPDH F: GGTCGGTGTGAACGGATTTGG 148 bpR: GCCGTGGGTAGAGTCATACTGGAAC

254 J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264

Page 4: Acta Biomaterialia - SOCRATES

At the beginning of surgery, an adrenaline containing dentallocal anaesthetic (Scandonest� 2% L, Mepivacaine HCl. 2% andLevonordefrin 1:20,000 Injection, USP) diluted to 0.5% was usedfor purpose of haemostasis. A full-thickness incision measuring4 mm was then made parallel to the alveolar ridge starting at themesio-palatal line angle of the first molar. The flap was elevatedto expose the alveolar bone. Under irrigation with sterile saline, adental hand-piece was used to create a defect without perforationinto the sinus. A 1.4-mm diameter dental round bur was used toestablish the depth, after which a 1.8-mm diameter dental roundbur was used to refine the defect. A periodontal probe was lastused to confirm the final size of the defect. The root surface wasgently debrided with a 0.7-mm diameter round bur to removeresidual PDL. The defect was then rinsed with saline and dried withsterile cotton pellets to ensure haemostasis was achieved before itwas left untreated or implanted with exosome-loaded collagensponge (CS/Exosome) or control collagen sponge (CS/Control).Finally, the flap was closed with single interrupted sutures (Vicryl6-0, Ethicon Products, Amersfoort, Netherlands) and primary clo-sure was achieved. Subcutaneous injections of antibiotics (Euflox-acin; 0.1 ml/100 g) and analgesics (Camprofen; 0.1 ml/100 g) weregiven once daily for 5 days post-operatively. The animals werehoused in pairs and allowed to move without restriction. Standardfood and water were provided ad libitum. At 2 and 4 weeks, ani-mals were euthanized by CO2 inhalation. The dentoalveolar com-plex of the maxillary molars were harvested for analysis.

2.12. Micro-computed tomography evaluation

The harvested tissues were fixed in 10% (v/v) neutral bufferedformalin (Sigma) for a week. Each maxilla was scanned usingmicro-computed tomography (micro-CT; Skyscan 1176, Kontich,Belgium) at 18-lm resolution. Using the multi-planar reconstruc-tion function, the cut was first oriented to the long axis of themesial root, and the sagittal view at the middle of the defect wasutilised for the 3-dimensional (3-D) analysis. This would minimiseinconsistencies in measurement arising from different angulations.A stack of 50 slides spanning the defect site were selected for mea-surements of the percentage of bone volume over total volume(BV/TV, %), trabecular separation (Tb.Sp, mm), trabecular thickness(Tb.Th, mm) and trabecular number (Tb.N, 1/mm) using CTAn ver-sion 1.13 (Burker micro-CT, Kontich, Belgium). For 3-D measure-ments, a region of interest (ROI) of approximately 0.125 mm3

was defined, coronally from the line angles of the mesial rootand amounting to a stack of 50 sections for analysis.

2.13. Histology, immunohistochemistry and histomorphometricanalysis

The harvested tissues were then decalcified in 30% (v/v) buf-fered formic acid for 1 week, trimmed into the individual rightand left dentoalveolar complexes, before another 2 weeks of decal-cification. Following decalcification, the samples were dehydrated

Fig. 1. Animal model and study design. (A) Baseline illustrating the condition prior to surgery. Following the incision and flap elevation, the naive condition of the alveolarbone is exposed. Defect creation on the mesial surface of the first molar in each rat was then performed. Each defect was 2 � 2 � 1.5 mm (W � L � D). (B) SD rats wererandomly allocated into three groups: defects untreated, defects treated with exosome-loaded collagen sponge (CS/Exosome; blue) and defects treated with control collagensponge (CS/Control; grey) (n = 12 per group). (C) The rats were euthanized at 2 or 4 weeks and samples were processed for micro-CT, histology, immunohistochemistry andhistomorphometric analyses. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264 255

Page 5: Acta Biomaterialia - SOCRATES

in increasing concentrations of ethanol and embedded in paraffin.Serial sections were cut at 5-mm thickness with a microtome (Leica,Hamburg, Germany) and were stained with haematoxylin andeosin (HE) to examine the general morphology and with Masson’strichrome (MT) to evaluate the fibre insertion and collagen matrixdeposition on the root surface. The stained sections were thenimaged using an inverted microscope (Olympus IX70). Histomor-phometric analysis was then carried out using ImageJ (NationalInstitutes of Health, Bethesda, MD, USA) as previously described[22]. As illustrated in Fig. 2C, four reference lines were drawn to

facilitate the measurements. Line 1 was drawn to estimate theposition of the occlusal plane. Line 2 was drawn to identify thelocation of the mesial cemento-enamel junction (CEJ) of the firstmolar, accounting for any possible damage during surgery. Line 2joined the distal CEJ of the first molar and the CEJ of the secondmolar. Line 3 was drawn to join the adjacent bone peaks to indicatethe alveolar bone levels [22]. Line 4 was drawn at the root tip par-allel to the occlusal plane. All measurements were parallel to thelong axis of the tooth. The tooth length (black line) extended fromLine 1 to 4, while the root length (green line) measured from Line 2

Fig. 2. Histological and histomorphometric evaluation of periodontal regeneration. Following 2 and 4 weeks of treatment, samples were harvested for histological andimmunohistochemical analysis. Histological analysis of periodontal regeneration by (A) haematoxylin and eosin (HE) and (B) Masson’s trichrome (MT) staining.Representative images (n = 6). The red box indicates the region of interest (ROI) for the magnified view of HE while the blue box indicates that for the MT. The blackarrowheads indicate the newly-formed bone, whereas the R indicates the root surface. (C) Schematic illustrating measurements of relative bone gap and PDL length. Fourreference lines were drawn to facilitate the measurements. Line 1 was drawn to demarcate the position of the occlusal plane. Line 2 was drawn to estimate the position of thecemento-enamel junction (CEJ) on the mesial surface of the first molar. Line 3 was drawn to join the adjacent bone peaks to indicate the alveolar bone levels. Line 4 wasdrawn at the root tip parallel to the occlusal plane. Measurements were parallel to the long axis of the tooth. The tooth length was represented by the black line that extendedfrom Line 1 to 4, while the root length was represented by the green line measuring from Line 2 to 4. The relative bone gap was assessed as the distance (blue line) from thealveolar bone level to the top of the new bone and expressed as a percentage of the tooth length. The relative PDL length was assessed as the length of the root surface (redline) with inserting functional PDL fibres (defined by an angle between long axis of the fibres to the root surface that was > 60�) and expressed as a percentage of the rootlength. The data are expressed as the mean ± SEM. *P < 0.05, **P < 0.01 compared to CS/Control or untreated group; #P < 0.05, ##P < 0.01 compared to CS/Exosome at 2 weeks(n = 6). CS/Exosome group showed significant improvements in the measurements of PDL length and bone gap indicative of new PDL and bone formation, compared to that ofCS/Control and untreated groups. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

256 J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264

Page 6: Acta Biomaterialia - SOCRATES

to 4. Inversely indicative of newly-formed bone, the relative bonegap was assessed as the distance (blue line) from the alveolar bonelevel to the top of the new bone and expressed as a percentage ofthe tooth length, parallel to the long axis of the mesial root as pre-viously described [22]. Using the MT-stained sections, the length ofthe root surface (red line) with inserting functional PDL fibres(defined by an angle between long axis of the fibres to the root sur-face that was >60�) was measured. This was then expressed as apercentage of the root length and reported as the relative PDLlength. Analysis was performed in a blinded fashion by two peri-odontal surgeons JRC and JHF, and the mean of two measurementswere tabulated.

Immunohistochemistry was performed using a biotin-streptavidin Lab Vision UltraVision detection system (ThermoFisher Scientific) to detect the proliferating cells expressing prolif-erative cell nuclear antigen (PCNA) as previously described [18].Briefly, the sections were hydrated before treated for antigenretrieval with sodium citric buffer (pH 6) at 60 �C for 1 h. Endoge-nous peroxidase was quenched using peroxidase block for 15 min,and non-specific protein binding was blocked by incubation withUltra V blocking solution for 5 min. The mouse anti-PCNA antibody(1:4000, Abcam) was then applied at ambient temperature for 1 hbefore rinsing with PBS and incubating with biotinylated sec-ondary antibody for 30 min. The DAB (3, 30-diaminobenzidine)chromogen was applied for 3 min to visualize the antibody-antigen reaction. Nuclear counterstaining was performed usingMayer’s haematoxylin (Sigma). The stained sections were dehy-drated and cover-slipped before imaged under an inverted micro-scope (Olympus IX70). For the quantification analysis of PCNA, aROI of approximately 0.155 mm2 was defined at the coronal mostaspect of the PDL. Positively-stained cells were counted at 200�magnification and expressed as a percentage of positive cells pre-sent in the ROI.

2.14. Statistical analysis

All quantitative data were reported as mean ± standard error ofthe mean (SEM). Shapiro-Wilk test was first used to determine thenormality of the data distribution. Student’s t-test or one-wayanalysis of variance (ANOVA) followed by Scheffe’s post-hoc testwas performed for normally distributed data. Mann-Whitney andKruskal-Wallis tests were performed for data that was not nor-mally distributed using a statistical package (SPSS version 25.0,Chicago, IL, USA). Statistical significance was set as P < 0.05.

3. Results

3.1. MSC exosomes promote regeneration of critical-size periodontaldefects

At 2 weeks, both CS/Exosome and CS/Control groups showedenhanced infiltration of fibroblast-like cells that probablyenhanced the formation of new soft tissues. Remnants of collagensponges were found. In contrast, the untreated group had lessercell infiltration and showed a collapse of gingival soft tissues. Allthe groups showed epithelial down-growth along the root surfaces,and the orientation of the collagen fibres was haphazard with nodefinitive direction.

The CS/Exosome group but not the CS/Control and untreatedgroups showed small amounts of newly-formed alveolar bone(indicated by black arrowheads) at the apical end of the defect(Fig. 2A). Consistent with these observations, our histomorphome-tric measurements showed reduced bone gap indicative ofincreased new bone formation in the CS/Exosome group(35.89 ± 0.38%) compared to those in CS/Control group

(41.72 ± 1.33%; P = 0.001) and untreated group (39.12 ± 0.61%;P = 0.058) (Fig. 2D).

At 4 weeks, the host cell infiltration of fibroblast-like cells hadsubsided in all the groups. The epithelial down-growth persistedin all the groups with the collagen fibres appearing parallel tothe root surfaces. CS/Exosome group displayed the most significantimprovements in bone and periodontal tissue regeneration com-pared to that in CS/Control and untreated groups. Five out of sixdefects (�83%) in the CS/Exosome group showed new bone forma-tion, as opposed to only two out of six defects (�33%) in the CS/Control group and three out of six defects (50%) in the untreatedgroup. Bone gap measurements showed significantly reduced bonegap of 28.02 ± 1.63% in the CS/Exosome group compared to37.17 ± 1.71% in CS/Control group (P = 0.003) and 38.01 ± 1.31% inuntreated group (P = 0.002). The bone gap in the exosome groupwas about 73.7% of that in the untreated group. Compared to2 weeks, only the exosome-treated defects significantly improvedtheir relative bone gap by 7.87% (P = 0.001), while the other twogroups, CS/Control and untreated groups showed no significantimprovements from week 2 to week 4. Ankylosis was also notobserved in all samples at both time points.

At 4 weeks, functionally oriented PDL fibres were seen to spanbetween the root surface and the newly-formed bone in four outof six defects in the CS/Exosome group. In contrast, no functionallyoriented PDL fibres were observed in CS/Control and untreatedgroups, instead disorganized collagen fibres parallel to thedenuded root surface were found. The mean PDL length was rela-tively longer in the CS/Exosome group (20.30 ± 0.96%) than in theCS/Control group (15.10 ± 1.46%; P = 0.046) and untreated group(14.44 ± 1.51%; P = 0.024) (Fig. 2B and E). Among the groups, theCS/Exosome group exhibited a significant increase in the PDLlength by 5.95% from week 2 to week 4 (P = 0.030). In contrast,CS/Control and untreated groups showed no significant improve-ments in the PDL length between the two time points.

Consistent with the bone gap measurements, the micro-CTassessment of bone structural parameters at 2 weeks revealed sig-nificantly higher percentage of bone volume over total volume (BV/TV) in the CS/Exosome group (35.69 ± 0.94%) compared to that inCS/Control group (29.13 ± 2.18%; P = 0.025) but not to theuntreated group (34.93 ± 1.07%; P = 0.938) (Fig. 3A and B). No sig-nificant differences in other parameters, Tb.Sp, Tb.Th and Tb.Nwere observed.

Differences in bone parameters were more evident at 4 weeks.At 4 weeks, significantly higher BV/TV of 45.77 ± 3.25% was foundin CS/Exosome group, compared to 31.37 ± 2.20% (P = 0.010) inCS/Control group and 30.69 ± 3.01% (P = 0.007) in untreated group(Fig. 3A and B). Trabecular separation (Tb.Sp) was also significantlylower in CS/Exosome group (0.099 ± 0.003 mm) compared to CS/Control (0.143 ± 0.013 mm) and untreated (0.141 ± 0.009 mm)(P < 0.05, Fig. 3C). However, no significant differences in Tb.Thand Tb.N were found among the three groups (Fig. 3D and E). Con-sistent with the improvements in bone gap from 2 to 4 weeks, sig-nificant improvements in BV/TV (35.69 ± 0.94% to 45.77 ± 3.25%;P = 0.025) and Tb.Sp (0.146 ± 0.007 to 0.099 ± 0.003 mm;P < 0.001) were observed with exosome treatment (Fig. 3A–C).

3.2. MSC exosomes enhance proliferation during periodontalregeneration

Cellular proliferation by staining for PCNA+ cells was examinedat the coronal most aspect of the PDL (Fig. 4A). The CS/Exosomegroup consistently displayed a significantly higher percentage ofproliferative PCNA+ cells at week 2 and week 4 (48.14 ± 3.20%and 35.39 ± 3.00%, respectively) compared to that of CS/Controlgroup (20.37 ± 3.01%; P = 0.024 and 17.31 ± 3.37%; P = 0.046,

J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264 257

Page 7: Acta Biomaterialia - SOCRATES

respectively) and untreated group (20.67 ± 6.71%; P = 0.012 and15.06 ± 6.62%; P = 0.024, respectively) (Fig. 4B & C).

3.3. Exosome loading and release from collagen scaffold

To determine the loading efficiency, the sponges were incu-bated in PBS for 10 min at ambient temperature and repeatedlypressed to release the exosomes. The CTB-CD81 level of releasedexosomes was measured and normalized against that in 40 lg exo-somes. By assaying for CTB-CD81, a surrogate marker for intactMSC exosomes, it was determined that the exosome-loaded colla-gen sponges contained the equivalent of 67.7 ± 1.8% of CTB-CD81present in 40 lg exosomes (Fig. 5). Hence, the loading efficiencywas 67.7 ± 1.8%.

To determine the release profile, the exosome-loaded spongeswere incubated in PBS at ambient temperature and monitoredfor CTB-CD81 which was normalized against that in 40 lg exo-somes. There was a rapid release of exosomes as indicated by theCTB-CD81 level following a 10-min of incubation (Fig. 5). By24 h, 69.1 ± 1.6% which approximated that of the loading efficiency(67.7%) was released, implying that most of the loaded exosomeswere released (Fig. 5). At 48 h, the CTB-CD81 level had rapidlydeclined to 30.1 ± 0.1% indicating a significant exosome loss ofabout 50% exosomes, possibly due to degradation (Fig. 5). Wehad previously reported that the integrity of MSC exosome is crit-ical for its bioactivity in tissue repair [24]. These findings suggestthat the bioavailability of exosomes was limited to about 48 hpost-implantation.

Fig. 3. Micro-CT assessment of periodontal regeneration. Following 2 and 4 weeks of treatment, samples were harvested for micro-CT analysis. (A) Sagittal view ofrepresentative micro-CT images for the different groups at the defect site (indicated by the red arrowheads). Quantitative micro-CT assessment of (B) BV/TV (%), (C) Tb.Sp(mm), (D) Tb.Th (mm) and (E) Tb.N (1/mm). Data are expressed as the mean ± SEM. *P < 0.05, **P < 0.01 compared to CS/Control or untreated group; #P < 0.05, ###P < 0.001compared to CS/Exosome at 2 weeks (n = 6). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

258 J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264

Page 8: Acta Biomaterialia - SOCRATES

3.4. Exosome uptake and effects on PDL cell migration andproliferation

As the exosome-mediated periodontal regeneration was charac-terized by enhanced cell infiltration and proliferation, we tested ifthese processes were directly modulated by MSC exosomes. Toassess if MSC exosomes interact directly with PDL cells, PDL cellswere incubated with labelled exosomes and monitored by confocalmicroscopy over 6 h. We observed that the cells rapidly take up theexosomes and became fluorescent within 30 min. They reached amaximum fluorescent intensity at 3 h before diminishing at 6 h(Fig. 6A). When viewed under a higher magnification at 1 h, thelabelled exosomes were found mainly in the cytoplasm (Fig. 6B).These findings implied that exosomes could be rapidly internalizedand interacted directly with the PDL cells. In our migration assay,MSC exosomes enhanced migration of PDL cells in a dose-dependent manner (Fig. 6C). At 10 mg/ml, exosomes induced a 2-fold higher migration rate than that of PBS control (P < 0.001,Fig. 6C). This finding suggests that MSC exosomes facilitate peri-

odontal defect repair, possibly by eliciting rapid migration of PDLcells into the defect. To further determine if MSC exosomes affectthe proliferation of PDL cells as observed in the animal model,PDL cells were treated with increasing concentrations of exosomesover 72 h. As shown in Fig. 6D, exosome treatment dose-dependently enhanced the number of viable PDL cells from 24 honwards, with significant enhancements at 5 mg/ml (P = 0.021)and 10 mg/ml (P < 0.001) compared to that of PBS treatment. Afterpeaking at 48 h, there was a general decrease in cell viability butthe exosome-treated PDL cells consistently showed higher viabilitywith 10 mg/ml exosomes showing at least 1.6-fold higher viabilitythan the PBS control (P < 0.001, Fig. 6D). As with cell viability,the DNA content reflective of the cell number increased withincreasing concentrations of exosomes (Fig. 6E). Consistent withthe changes in cell viability, 10 mg/ml of exosomes induced a signif-icant increase in cell number compared to the PBS control group(P = 0.014, Fig. 6E) as early as 24 h. By the end of 72 h, PDL cellsin the 10 mg/ml exosome treatment group proliferated 2.3-fold fas-ter than those in the PBS control group (P = 0.008, Fig. 6E).

Fig. 4. Evaluation of cellular proliferation during the course of exosome-induced periodontal regeneration. (A) Proliferative cell nuclear antigen (PCNA) staining and analysisof PCNA+ cells at the coronal most portion of the PDL. (B) Immunohistochemical staining of PCNA and (C) quantitative analysis of PCNA+ cells at 2 and 4 weeks. Representativeimages (n = 6). The PCNA+ cells were counted at 200 �magnification and expressed as the percentage of positively stained cells. Data are expressed as the mean ± SEM.*P < 0.05 compared to CS/Control or untreated group; #P < 0.05 compared to CS/Exosome at 2 weeks (n = 6).

J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264 259

Page 9: Acta Biomaterialia - SOCRATES

3.5. MSC exosomes enhance PDL cellular migration and proliferationthrough adenosine receptor-mediated activation of AKT and ERKsignalling pathways

MSC exosomes had been shown to activate pro-survival AKTand ERK signalling in vitro and in vivo [24,25] at least in partthrough exosomal CD73/ecto-5́-nucleotidase (NT5E) activity.CD73 is the only known extracellular NT5E that can hydrolyse ade-nosine monophosphate (AMP) to adenosines which in turn canactivate AKT and ERK phosphorylation via adenosine receptorinteraction [26]. We had also previously demonstrated that adeno-sine signalling promoted chondrocyte proliferation and migration[18]. We therefore postulated that exosomal CD73/NT5E-mediated AKT and ERK signalling pathways could similarly pro-mote PDL migration and proliferation to regenerate periodontaldefects in the animals. Consistent with our hypothesis, MSC exo-somes elicited rapid phosphorylation of AKT and ERK and thisphosphorylation was attenuated by theophylline, a non-selectiveadenosine receptor antagonist (Fig. 7A and B). Specifically at5 mg/ml exosome, phosphorylation of AKT and ERK peaked as earlyas 15 min. On contrary, AKT phosphorylation remained low whileERK phosphorylation was reduced with PBS treatment at 15 minbefore declining thereafter. Notably exosome-induced AKT andERK phosphorylation was consistently higher than that of PBS con-trol for up to 1 h (Fig. 7A). These results demonstrated that MSCexosomes can rapidly initiate pro-survival signalling in PDL cellsto enhance cell viability and induce gene expression changes

Next, we determine if AKT and ERK signalling mediateexosome-induced increases in PDL cellular migration and prolifer-ation. Phosphorylation of AKT and ERK induced by MSC exosomeswas abrogated by treatment with wortmannin and U0126, respec-tively (Fig. 7C). Functionally, MSC exosomes promoted migration ofPDL cells but this promotion was suppressed 3-fold and 2-fold by

AKT (P < 0.001) and ERK inhibition (P < 0.001, Fig. 7D), respectively.Similarly, exosome treatment enhanced PDL cell proliferation com-pared to PBS control (P < 0.01). However, this exosome-enhancedproliferation at 48 h was significantly reduced 1.7-fold and 1.4-fold with inhibition of AKT (P < 0.001) and ERK (P = 0.003,Fig. 7E), respectively. These observations corroborated our geneexpression analysis (Fig. 7F). Exosome-induced elevation of FGF-2and Bcl-2 at 24 h was partially suppressed by both AKT and ERKinhibition (P < 0.01). PCNA, Survivin, COL1A1 and TGF-b1 were sig-nificantly downregulated by AKT (but not ERK) inhibition(P < 0.05). On the other hand, exosome-induced suppression ofBAX was partially reversed with ERK inhibition (P = 0.03) that con-currently inhibited IGF-1 expression induced by MSC exosomes(P = 0.04). However, exosome-induced elevation of POSTN wasnot affected by AKT or ERK inhibition, suggesting that some ofthe exosome-induced gene expressions were not mediated byAKT or ERK signalling, and possibly not by CD73-mediated adeno-sine receptor signalling. Collectively, our findings suggest that MSCexosomes can mediate adenosine receptor activation of pro-survival kinases AKT and ERK, and related gene expression toenhance PDL cell migration and proliferation that are cellular pro-cesses relevant to periodontal regeneration.

4. Discussion

In this report, we demonstrated that human MSC exosome-loaded collagen sponge (CS/Exosome) enhanced periodontal regen-eration in an adult immunocompetent rat model. Consistent withour previous studies that used the same source of human MSC exo-somes, no adverse responses from the immunocompetent animalswere observed [10,13,14].

Previous studies have reported that transplantation of PDL stemcells and bone marrow MSCs, or their conditioned medium in col-lagen sponges were able to promote periodontal regeneration inanimal studies [9,27]. Since the therapeutic effect of the MSC con-ditioned medium had been increasingly attributed to MSC exo-somes, we investigated here the efficacy of exosome-loadedsponges in periodontal regeneration. In this study, we demon-strated that MSC exosomes replicated the therapeutic efficacy ofMSCs and conditioned medium in promoting periodontal regener-ation in a rat model. We assessed periodontal regeneration bystandard parameters, namely bone growth, functional PDL lengthand inhibition of epithelial down-growth. Notably, MSC exosomesenhanced bone growth and improved PDL length with significantimprovements over time. However, exosome treatment did notinhibit epithelial down-growth. This down-growth is typicallyobserved in small animal models where a barrier membrane toexclude epithelial cells was not applied because of the small defectsize of the animals [22].

Collagen sponges were used as the scaffold material to load exo-somes as they are widely used for space maintenance in conjunc-tion with other biologics for periodontal regeneration [28]. Beingradiolucent, resorbable and relatively inert compared to bone sub-stitutes (e.g. deproteinised bovine bone mineral) in bone regenera-tion, collagen sponge was ideal as a scaffold for testing MSCexosomes in regeneration of periodontal tissues. The inertness ofthe collagen sponge with limited capacity to promote periodontalregeneration was also confirmed in our study.

In this study, we observed that a single implantation ofexosome-loaded collagen sponge in a surgically created periodon-tal defect model promoted periodontal regeneration withenhanced bone growth and increased functional PDL length atweek 4. This observation suggests that despite the rapid releaseand decay of exosomes from the collagen sponges within the first48 h, the effect of exosome on periodontal regeneration appearedto persist and propagate for at least 4 weeks as evident by the

Fig. 5. Exosome loading efficiency and the release from exosome-loaded collagensponge. The loading efficiency and the release of intact exosomes from theexosome-loaded sponges were measured using the CTB-CD81 assay. Each spongewas loaded with 40 lg exosomes. To determine the loading efficiency, the spongeswere incubated in PBS for 10 min and repeatedly pressed to release the exosomes.To determine the release kinetics, the exosome-loaded sponges were incubated inPBS at ambient temperature for 10 min, 24 and 48 h. Supernatant samples werethen assayed for CTB-CD81. The levels of CTB-CD81 were normalized to the initiallevel of CTB-CD81 present in 40 lg exosomes and expressed as a percentage. Dataare expressed as the mean ± SEM. n = 2.

260 J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264

Page 10: Acta Biomaterialia - SOCRATES

significant improvements in bone regeneration and functionallyoriented PDL formation in the exosome-treated animals over a per-iod of 4 weeks. This observation that a transient exposure to MSCexosomes could trigger a sustained regenerative process is consis-tent with our long-standing hypothesis that MSC exosomes workrapidly to restore homeostasis and enable the initiation of endoge-nous tissue repair and regeneration processes [29,30].

In 2013, we demonstrated that MSC exosomes reduce myocar-dial ischemia reperfusion injury by rapidly increasing ATP produc-tion, activating survival kinase signalling and reducing apoptosis ina mouse model of reperfusion injury [24]. We subsequently attrib-uted this expeditious halt in a cascading injury process, restorationof homeostasis and the initiation of cellular recovery and repair tothe protein cargo of MSC exosomes [31–33]. We further postulatedthat as many of the key proteins to initiate cellular repair andrecovery were enzymes, they could potentially restore homeosta-sis in the injured tissue to facilitate endogenous tissue repair andregeneration [29].

Therefore, the relatively short 48-hour exposure to MSC exo-some released by the collagen sponge may be sufficient to promoteperiodontal regeneration. This is also supported by our observa-tions that MSC exosomes were rapidly taken up within minutesby PDL cells in vitro, induced signal transduction within 15 min,

and modulated gene expression and cellular activities such asmigration, viability and proliferation within 48 h. In particular,the exosome-mediated increase in expression of genes associatedwith cell migration (IGF-1, FGF-2), survival and anti-apoptosis(IGF-1, Survivin and Bcl-2), and proliferation (IGF-1, FGF-2 andPCNA) [34–37] has the potential to propagate the effects of exo-somes on these cellular activities beyond 48 h. We also observedexosome-mediated increase in PDL matrix-associated genes,namely the extracellular matrix proteins, COL1A1 and POSTN, andassociated growth factors for matrix synthesis, IGF-1 and TGF-b1[35,38]. Apart from matrix synthesis, IGF-1 also plays critical rolesin bone matrix mineralization [39] and had reportedly enhancedosteogenic differentiation of PDL stem cells [35]. Periostin (POSTN)is an extracellular matrix protein that supports adhesion andmigration of fibroblasts and osteoblasts, and is thought to con-tribute to periodontal regeneration by recruiting PDL fibroblastsand osteoblasts for new PDL and bone formation, respectively[40]. These could explain the significant bone formation in theexosome-treated animals. Together, these findings suggest thatMSC exosomes facilitated periodontal regeneration through a syn-ergistic combination of enhanced cell viability, migration, prolifer-ation, matrix synthesis and differentiation to form new bone andPDL attachment.

Fig. 6. Effects of MSC exosomes on PDL cell migration and proliferation. (A) Cellular uptake assay by confocal microscopy demonstrated rapid uptake of MSC exosomes(green). (B) Merged images of CellMaskTM Deep Red plasma membrane stain (red) and Alexafluor-488-labelled exosomes (green) at high magnification revealed cytoplasmiclocalization of exosomes. (C) Transwell migration assay showed dose-dependent effect of MSC exosomes on PDL cell migration. Data are expressed as the mean ± SEM.**P < 0.05, ***P < 0.001 compared to PBS control; ##P < 0.01 compared to 1 mg/ml exosome (n = 3). (D) Cell viability assay and (E) DNA assay showed potent dose and time-dependent effects of MSC exosomes on PDL cell viability and proliferation. Data are expressed as the mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 compared to PBS control;#P < 0.05 compared to 1 mg/ml exosome (n = 3). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264 261

Page 11: Acta Biomaterialia - SOCRATES

Fig. 7. MSC exosomes modulate PDL cellular activities in part through adenosine receptor activation of AKT and ERK signalling. (A) Western blot analysis revealed rapidactivation of AKT and ERK signalling pathways in PDL cells following exposure to 5 mg/ml exosomes. Representative results of 3 independent experiments. (B) Western blotanalysis of adenosine receptor inhibition by theophylline (a non-selective adenosine receptor antagonist; 1 mM). Adenosine receptor inhibition by theophylline (THEO)reduced exosome-mediated AKT and ERK phosphorylation. Representative results of 3 independent experiments. (C) Western blot analysis of AKT and ERK inhibition by PI3K/AKT inhibitor, wortmannin (WORT; 1 mM) and MAPK/ERK inhibitor (U0126; 10 mM). Wortmannin and U0126 suppressed exosome-mediated phosphorylation of AKT and ERK,respectively. Representative results of 3 independent experiments. (D) Transwell migration assay showed that exosome-mediated PDL cell migration was suppressed by AKTinhibition, and lesser extent of ERK. Data are expressed as the mean ± SEM. **P < 0.01, ***P < 0.001 compared to PBS control; ###P < 0.001 compared to cells with exosome andvehicle (DMSO) treatment (n = 3). (E) DNA assay showed that MSC exosomes enhanced PDL cell proliferation at 48 h, but this effect was significantly reduced by inhibition ofAKT and by a lesser extent with inhibition of ERK. Data are expressed as the mean ± SEM. **P < 0.01 compared to PBS control; ##P < 0.01, ###P < 0.001 compared to cellstreated with exosome and DMSO (n = 3). (F) Quantitative RT-PCR analysis at 24 h post-treatment showed involvement of AKT and ERK signalling pathways in exosome-mediated regulation of genes associated with the PDL cellular activities. Data are expressed as the mean ± SEM. *P < 0.05, **P < 0.01 compared to PBS control; #P < 0.05,##P < 0.01 compared to cells treated with exosome and DMSO (n = 5).

262 J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264

Page 12: Acta Biomaterialia - SOCRATES

To elucidate the molecular mechanism by which MSC exosomescould enhance this multitude of cellular activities, we investigatedif CD73-mediated adenosine receptor signalling was involved. MSCexosomes expresses CD73, the only known ecto-5́-nucleotidase(NT5E) that can hydrolyse extracellular AMP to adenosines to acti-vate the pro-survival AKT and ERK signalling through the adeno-sine receptors [25]. ATP is a danger signal released by cells instress or injury to remove or destroy ‘‘dangers” [41,42]. Excessiveextracellular ATP is deleterious, and could lead to excessive inflam-mation and cell death [42]. In periodontitis, extracellular ATP hasbeen implicated as a cause of alveolar bone loss through elevatedpro-inflammatory cytokine production and caspase activity [43],and as a therapeutic target to prevent further inflammation andperiodontal tissue destruction [43,44]. Extracellular ATP has a veryshort half-life measured in seconds [45] and is degraded by ecto-nucleotidases such as CD39 to ADP and AMP, and finally by CD73into adenosine, a potent activator of the survival kinases, AKTand ERK.

The role of CD73 in mediating the therapeutic activity of MSCexosome has been reported [18,24,25,46]. We had previouslyobserved that the MSC exosome-mediated cardioprotection wasconcomitant with AKT phosphorylation, a survival kinase sig-nalling in a mouse model of myocardial infarction reperfusioninjury [24], which is induced possibly by CD73-mediated adeno-sine signalling [24,25]. In addition, MSC exosomes also inducedAKT and ERK phosphorylation in chondrocytes that were stressedby reducing serum or adding inflammatory cytokine interleukin-1b in the culture medium [18,46]. This phosphorylation was abro-gated in the presence of theophylline, a non-selective antagonist ofadenosine receptors.

In this study, we also observed that adenosine signallingthrough phosphorylation of AKT and ERK played a significant rolein mediating exosome-induced PDL cellular migration and prolifer-ation. Attenuation of AKT or ERK phosphorylation by wortmanninor U0126 significantly inhibited these cellular activities. AKTappeared to be a more critical target of MSC exosomes than ERK,as attenuation of AKT phosphorylation resulted in a greater reduc-tion in PDL cell migration and proliferation. Together, these find-ings suggest that the enhanced cellular infiltration andproliferation observed during exosome-mediated periodontalregeneration could be attributed at least in part to exosomalCD73-mediated adenosine receptor activation of pro-survival AKTand ERK signalling. However, it is important to note that therecould be other exosome activators of AKT and/or ERK phosphoryla-tion since exosome-mediated AKT and ERK phosphorylation wasnot completely abolished by theophylline inhibition of the adeno-sine receptors. Indeed, MSC exosome cargo is highly diverse andcomplex [21,32,47,48] and contain growth factors such as TGF-b,IGF and FGF that could potentially activate AKT and/or ERK sig-nalling pathways in PDL cells [34–38].

This study provides a rationale for the development of an ‘off-the-shelf’ cell-free therapeutic alternative to cell-based MSC ther-apy for the treatment of periodontal defects. Comparing to cellulartherapies, exosome-based therapeutics offer unique advantages ofbeing cell-free, ready-to-use and more amenable to reformulationto support different routes of administration [49]. For instance,exosomemanufacture is more amenable to scalable manufacturingprocesses such as the use of immortalized clonal producer cellsources [50].

As in all studies, there are notable limitations to our study.Although there was regeneration of periodontal tissues, the overallextent of regeneration was not optimal. Future studies would beneeded to determine the dose of exosomes and to identify scaffoldmaterials for optimal tissue regeneration. The effects of exosomesshould also be monitored over a longer follow-up period. Further-more, the rat periodontal defect model that was surgically created

in our study might not be reflective of the chronic inflammatoryconditions of plaque-induced periodontitis. Nevertheless, it pro-vides a critical proof-of-concept to rationalize future investigationin larger, clinically relevant animal models.

5. Conclusion

Periodontal regeneration aims to reconstitute the lost periodon-tal tissues including PDL, cementum and alveolar bone. In thisstudy, we have shown that human MSC exosomes in combinationwith collagen sponge have the capacity to promote regeneration ofperiodontal tissues and newly-formed alveolar bone in a rat peri-odontal defect model without causing any adverse responses. Thisexosome-mediated periodontal regeneration could be attributed tothe effects of MSC exosomes in promoting PDL cellular activitiessuch as migration and proliferation in part through adenosinereceptor activation of AKT and ERK signalling pathways. Impor-tantly, our findings provide the basis for further investigation ofhuman MSC exosomes as a ready-to-use and cell-free therapeuticfor periodontal regeneration.

Author contributions

JRC performed the experiments, acquired, analysed and inter-preted the data, and drafted the manuscript. SJC, KYT, SZ and RCLperformed the experiments, acquired, analysed and interpretedthe data. JHF, LPL and SKL analysed and interpreted the data andedited the manuscript. WST designed the study, analysed andinterpreted the data, drafted and edited the manuscript. Allauthors approved the final version of the manuscript.

Disclosure

The authors report that they have no conflicts of interest in theauthorship and publication of this article. SKL holds founder sharesin Paracrine Therapeutics Pte Ltd.

Acknowledgements

Exosome-loaded collagen sponges were gifts from ParacrineTherapeutics, Biopolis, Singapore. This work was supported bygrants from the National University Healthcare System, NationalUniversity of Singapore (R221000114114 and R221000100101)and National Medical Research Council Singapore(R221000123213). RCL and SKL are supported by a core grant fromAgency for Science, Technology and Research (A*STAR) Singapore.

References

[1] N.J. Kassebaum, A.G.C. Smith, E. Bernabé, T.D. Fleming, A.E. Reynolds, T. Vos, C.J.L. Murray, W. Marcenes, Global, Regional, and national prevalence, incidence,and disability-adjusted life years for oral conditions for 195 countries, 1990–2015: a systematic analysis for the global burden of diseases, injuries, and riskfactors, J. Dent. Res. 96 (4) (2017) 380–387.

[2] N.J. Kassebaum, E. Bernabé, M. Dahiya, B. Bhandari, C.J.L. Murray, W. Marcenes,Global burden of severe periodontitis in 1990–2010: a systematic review andmeta-regression, J. Dent. Res. 93 (11) (2014) 1045–1053.

[3] L.J.A. Heitz-Mayfield, N.P. Lang, Surgical and nonsurgical periodontal therapy.Learned and unlearned concepts, Periodontology 62 (1 2013) (2000) 218–231.

[4] J. Du, Z. Shan, P. Ma, S. Wang, Z. Fan, Allogeneic bone marrow mesenchymalstem cell transplantation for periodontal regeneration, J. Dent. Res. 93 (2)(2014) 183–188.

[5] K. Hynes, D. Menicanin, J. Han, V. Marino, K. Mrozik, S. Gronthos, P.M. Bartold,Mesenchymal stem cells from iPS cells facilitate periodontal regeneration, J.Dent. Res. 92 (9) (2013) 833–839.

[6] F.-M. Chen, L.-N. Gao, B.-M. Tian, X.-Y. Zhang, Y.-J. Zhang, G.-Y. Dong, H. Lu, Q.Chu, J. Xu, Y. Yu, R.-X. Wu, Y. Yin, S. Shi, Y. Jin, Treatment of periodontalintrabony defects using autologous periodontal ligament stem cells: arandomized clinical trial, Stem Cell Res. Ther. 7 (1) (2016) 33.

J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264 263

Page 13: Acta Biomaterialia - SOCRATES

[7] L. da Silva Meirelles, A.M. Fontes, D.T. Covas, A.I. Caplan, Mechanisms involvedin the therapeutic properties of mesenchymal stem cells, Cytokine GrowthFactor Rev. 20 (5) (2009) 419–427.

[8] N. Mizuki, I. Kengo, A. Keiko, K. Motohiro, Y. Naoki, I. Yuichi, M. Ikuo,Conditioned medium from periodontal ligament stem cells enhancesperiodontal regeneration, Tissue Eng. Part A 23 (9–10) (2017) 367–377.

[9] T. Kawai, W. Katagiri, M. Osugi, Y. Sugimura, H. Hibi, M. Ueda, Secretomes frombone marrow–derived mesenchymal stromal cells enhance periodontal tissueregeneration, Cytotherapy 17 (4) (2015) 369–381.

[10] R.C. Lai, F. Arslan, M.M. Lee, N.S.K. Sze, A. Choo, T.S. Chen, M. Salto-Tellez, L.Timmers, C.N. Lee, R.M. El Oakley, G. Pasterkamp, D.P.V. de Kleijn, S.K. Lim,Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury,Stem Cell Res. 4 (3) (2010) 214–222.

[11] B. Zhang, M. Wang, A. Gong, X. Zhang, X. Wu, Y. Zhu, H. Shi, L. Wu, W. Zhu, H.Qian, W. Xu, HucMSC-exosome mediated-Wnt4 signaling is required forcutaneous wound healing, Stem Cells 33 (7) (2015) 2158–2168.

[12] Z. Bin, Y. Yijun, L.R. Chai, T.S. Sim, C.A.B. Hwa, L.S. Kiang, Mesenchymal stemcells secrete immunologically active exosomes, Stem Cells Dev. 23 (11) (2014)1233–1244.

[13] C.Y. Tan, R.C. Lai, W. Wong, Y.Y. Dan, S.-K. Lim, H.K. Ho, Mesenchymal stemcell-derived exosomes promote hepatic regeneration in drug-induced liverinjury models, Stem Cell Res. Ther. 5 (3) (2014) 76.

[14] S. Zhang, W.C. Chu, R.C. Lai, S.K. Lim, J.H.P. Hui, W.S. Toh, Exosomes derivedfrom human embryonic mesenchymal stem cells promote osteochondralregeneration, Osteoarthrit. Cartil. 24 (12) (2016) 2135–2140.

[15] J. Zhang, X. Liu, H. Li, C. Chen, B. Hu, X. Niu, Q. Li, B. Zhao, Z. Xie, Y. Wang,Exosomes/tricalcium phosphate combination scaffolds can enhance boneregeneration by activating the PI3K/Akt signaling pathway, Stem Cell Res.Ther. 7 (1) (2016) 136.

[16] C.C. Teixeira, E. Khoo, J. Tran, I. Chartres, Y. Liu, L.M. Thant, I. Khabensky, L.P.Gart, G. Cisneros, M. Alikhani, Cytokine expression and accelerated toothmovement, J. Dent. Res. 89 (10) (2010) 1135–1141.

[17] J. Han, D. Menicanin, V. Marino, S. Ge, K. Mrozik, S. Gronthos, P.M. Bartold,Assessment of the regenerative potential of allogeneic periodontal ligamentstem cells in a rodent periodontal defect model, J. Periodontal Res. 49 (3)(2014) 333–345.

[18] S. Zhang, S.J. Chuah, R.C. Lai, J.H.P. Hui, S.K. Lim, W.S. Toh, MSC exosomesmediate cartilage repair by enhancing proliferation, attenuating apoptosis andmodulating immune reactivity, Biomaterials 156 (2018) 16–27.

[19] K.A. Jacobson, Introduction to adenosine receptors as therapeutic targets, in: C.N. Wilson, S.J. Mustafa (Eds.), Adenosine Receptors in Health and Disease,Springer, Berlin Heidelberg, Berlin, Heidelberg, 2009, pp. 1–24.

[20] K.J. Livak, T.D. Schmittgen, Analysis of Relative gene expression data usingreal-time quantitative PCR and the 2�DDCT Method, Methods 25 (4) (2001)402–408.

[21] R.C. Lai, S.S. Tan, R.W.Y. Yeo, A.B.H. Choo, A.T. Reiner, Y. Su, Y. Shen, Z. Fu, L.Alexander, S.K. Sze, S.K. Lim, MSC secretes at least 3 EV types each with aunique permutation of membrane lipid, protein and RNA, J. Extracell. Vesicl. 5(1) (2016) 29828.

[22] N. Yu, D.A.W. Oortgiesen, A.L.J.J. Bronckers, F. Yang, X.F. Walboomers, J.A.Jansen, Enhanced periodontal tissue regeneration by periodontal cellimplantation, J. Clin. Periodontol. 40 (7) (2013) 698–706.

[23] Y. Shirakata, M. Eliezer, C.E. Nemcovsky, M. Weinreb, M. Dard, A. Sculean, D.D.Bosshardt, O. Moses, Periodontal healing after application of enamel matrixderivative in surgical supra/infrabony periodontal defects in rats withstreptozotocin-induced diabetes, J. Periodontal Res. 49 (1) (2014) 93–101.

[24] F. Arslan, R.C. Lai, M.B. Smeets, L. Akeroyd, A. Choo, E.N.E. Aguor, L. Timmers, H.V. van Rijen, P.A. Doevendans, G. Pasterkamp, S.K. Lim, D.P. de Kleijn,Mesenchymal stem cell-derived exosomes increase ATP levels, decreaseoxidative stress and activate PI3K/Akt pathway to enhance myocardialviability and prevent adverse remodeling after myocardial ischemia/reperfusion injury, Stem Cell Res. 10 (3) (2013) 301–312.

[25] R.C. Lai, R.W.Y. Yeo, S.S. Tan, B. Zhang, Y. Yin, N.S.K. Sze, A. Choo, S.K. Lim,Mesenchymal Stem Cell Exosomes: The Future MSC-Based Therapy?, in: L.G.Chase, M.C. Vemuri (Eds.), Mesenchymal Stem Cell Therapy, Humana Press,Totowa, NJ, 2013, pp. 39–61.

[26] S.P. Colgan, H.K. Eltzschig, T. Eckle, L.F. Thompson, Physiological roles for ecto-5’-nucleotidase (CD73), Purin. Signal. 2 (2) (2006) 351.

[27] K. Sakaguchi, W. Katagiri, M. Osugi, T. Kawai, Y. Sugimura-Wakayama, H. Hibi,Periodontal tissue regeneration using the cytokine cocktail mimickingsecretomes in the conditioned media from human mesenchymal stem cells,Biochem. Biophys. Res. Commun. 484 (1) (2017) 100–106.

[28] Y. Shirakata, R.J. Miron, T. Nakamura, K. Sena, Y. Shinohara, N. Horai, D.D.Bosshardt, K. Noguchi, A. Sculean, Effects of EMD liquid (Osteogain) on

periodontal healing in class III furcation defects in monkeys, J. Clin.Periodontol. 44 (3) (2017) 298–307.

[29] R.C. Lai, R.W.Y. Yeo, S.K. Lim, Mesenchymal stem cell exosomes, Semin. CellDev. Biol. 40 (2015) 82–88.

[30] W.S. Toh, R.C. Lai, J.H.P. Hui, S.K. Lim, MSC exosome as a cell-free MSC therapyfor cartilage regeneration: implications for osteoarthritis treatment, Semin.Cell Dev. Biol. 67 (2017) 56–64.

[31] R.C. Lai, R.W. Yeo, K.H. Tan, S.K. Lim, Mesenchymal stem cell exosomeameliorates reperfusion injury through proteomic complementation, Regen.Med. 8 (2) (2013) 197–209.

[32] W.S. Toh, R.C. Lai, B. Zhang, S.K. Lim, MSC exosome works through a protein-based mechanism of action, Biochem. Soc. Trans. 46 (4) (2018) 843–853.

[33] R.C. Lai, R.W.Yeo, S.S. Tan,B.Zhang,Y. Yin,N.S. Sze,A.Choo, S.K. Lim,MesenchymalStemCell Exosomes: The FutureMSC-based Therapy?, in: L.G. Chase,M.C. Vemuri(Eds.), Mesenchymal Stem Cell Therapy, , Humana Press, 2012.

[34] X. Han, S. Amar, IGF-1 Signaling Enhances Cell Survival in PeriodontalLigament Fibroblasts vs. Gingival Fibroblasts, J. Dent. Res. 82 (6) (2003) 454–459.

[35] Y. Yu, J. Mu, Z. Fan, G. Lei, M. Yan, S. Wang, C. Tang, Z. Wang, J. Yu, G. Zhang,Insulin-like growth factor 1 enhances the proliferation and osteogenicdifferentiation of human periodontal ligament stem cells via ERK and JNKMAPK pathways, Histochem. Cell Biol. 137 (4) (2012) 513–525.

[36] A.C.P. Sant’Ana, M.M. Marques, E.C. Barroso, E. Passanezi, M.L.R. de Rezende,Effects of TGF-b1, PDGF-BB, and IGF-1 on the Rate of Proliferation andAdhesion of a Periodontal Ligament Cell Lineage In Vitro, J. Periodontol. 78 (10)(2007) 2007–2017.

[37] Y. Shimabukuro, H. Terashima, M. Takedachi, K. Maeda, T. Nakamura, K.Sawada, M. Kobashi, T. Awata, H. Oohara, T. Kawahara, T. Iwayama, T.Hashikawa, M. Yanagita, S. Yamada, S. Murakami, Fibroblast growth factor-2stimulates directed migration of periodontal ligament cells via PI3K/AKTsignaling and CD44/hyaluronan interaction, J. Cell. Physiol. 226 (3) (2011)809–821.

[38] S. Fujii, H. Maeda, A. Tomokiyo, S. Monnouchi, K. Hori, N. Wada, A. Akamine,Effects of TGF-b1 on the proliferation and differentiation of human periodontalligament cells and a human periodontal ligament stem/progenitor cell line,Cell Tissue Res. 342 (2) (2010) 233–242.

[39] M. Zhang, S. Xuan, M.L. Bouxsein, D. von Stechow, N. Akeno, M.C. Faugere, H.Malluche, G. Zhao, C.J. Rosen, A. Efstratiadis, T.L. Clemens, Osteoblast-specificknockout of the insulin-like growth factor (IGF) receptor gene reveals anessential role of IGF signaling in bone matrix mineralization, J. Biol. Chem. 277(46) (2002) 44005–44012.

[40] J. Du, M. Li, Functions of Periostin in dental tissues and its role in periodontaltissues’ regeneration, Cell. Mol. Life Sci. 74 (23) (2017) 4279–4286.

[41] J. Lüthje, Origin, metabolism and function of extracellular adenine nucleotidesin the blood, Klinische Wochenschrift 67 (6) (1989) 317–327.

[42] A. Trautmann, Extracellular ATP in the immune system: more than just a‘‘Danger Signal”, Sci. Signaling 2(56) (2009) pe6-pe6.

[43] I. Binderman, H. Bahar, J. Jacob-Hirsch, S. Zeligson, N. Amariglio, G. Rechavi, S.Shoham, A. Yaffe, P2X4 is up-regulated in gingival fibroblasts after periodontalsurgery, J. Dent. Res. 86 (2) (2007) 181–185.

[44] I. Binderman, N. Gadban, A. Yaffe, Extracellular ATP is a key modulator ofalveolar bone loss in periodontitis, Arch. Oral Biol. 81 (2017) 131–135.

[45] J.G. Fitz, Regulation of cellular ATP release, Trans. Am. Clin. Climatol. Assoc.118 (2007) 199.

[46] S. Zhang, K.Y.W. Teo, S.J. Chuah, R.C. Lai, S.K. Lim, W.S. Toh, MSC exosomesalleviate temporomandibular joint osteoarthritis by attenuating inflammationand restoring matrix homeostasis, Biomaterials 200 (2019) 35–47.

[47] R.C. Lai, S.S. Tan, B.J. Teh, S.K. Sze, F. Arslan, D.P. de Kleijn, A. Choo, S.K. Lim,Proteolytic potential of the MSC exosome proteome: implications for anexosome-mediated delivery of therapeutic proteasome, Int. J. Proteom. 2012(2012) 14.

[48] T.S. Chen, R.C. Lai, M.M. Lee, A.B.H. Choo, C.N. Lee, S.K. Lim, Mesenchymal stemcell secretes microparticles enriched in pre-microRNAs, Nucleic Acids Res. 38(1) (2010) 215–224.

[49] A.T. Reiner, K.W. Witwer, B.W.M. van Balkom, J. de Beer, C. Brodie, R.L.Corteling, S. Gabrielsson, M. Gimona, A.G. Ibrahim, D. de Kleijn, C.P. Lai, J.Lötvall, H.A. del Portillo, I.G. Reischl, M. Riazifar, C. Salomon, H. Tahara, W.S.Toh, M.H.M. Wauben, V.K. Yang, Y. Yang, R.W.Y. Yeo, H. Yin, B. Giebel, E. Rohde,S.K. Lim, Concise review: developing best-practice models for the therapeuticuse of extracellular vesicles, Stem Cells Transl. Med. 6 (8) (2017) 1730–1739.

[50] T.S. Chen, F. Arslan, Y. Yin, S.S. Tan, R.C. Lai, A.B.H. Choo, J. Padmanabhan, C.N.Lee, D.P. de Kleijn, S.K. Lim, Enabling a robust scalable manufacturing processfor therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs, J. Transl. Med. 9 (1) (2011) 47.

264 J.R.J. Chew et al. / Acta Biomaterialia 89 (2019) 252–264