You are on page 1of 17

Article

www.acsnano.org

Sequential Release of Small Extracellular


Vesicles from Bilayered Thiolated Alginate/
Polyethylene Glycol Diacrylate Hydrogels for
Scarless Wound Healing
Yifan Shen,# Guanzhe Xu,# Huanxuan Huang,# Kaiyang Wang, Hui Wang, Meidong Lang,* Hong Gao,*
and Shichang Zhao*
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Cite This: ACS Nano 2021, 15, 6352−6368 Read Online


Downloaded via BUTLER UNIV on May 15, 2021 at 04:42:13 (UTC).

ACCESS Metrics & More Article Recommendations *


sı Supporting Information

ABSTRACT: Excessive scar formation has adverse physiological and


psychological effects on patients; therefore, a therapeutic strategy for rapid
wound healing and reduced scar formation is urgently needed. Herein,
bilayered thiolated alginate/PEG diacrylate (BSSPD) hydrogels were fabricated
for sequential release of small extracellular vesicles (sEVs), which acted in
different wound healing phases, to achieve rapid and scarless wound healing.
The sEVs secreted by bone marrow derived mesenchymal stem cells (B-sEVs)
were released from the lower layer of the hydrogels to promote angiogenesis
and collagen deposition by accelerating fibroblast and endothelial cell
proliferation and migration during the early inflammation and proliferation
phases, while sEVs secreted by miR-29b-3p-enriched bone marrow derived
mesenchymal stem cells were released from the upper layer of the hydrogels
and suppressed excessive capillary proliferation and collagen deposition during
the late proliferation and maturation phases. In a full-thickness skin defect
model of rats and rabbit ears, the wound repair rate, angiogenesis, and collagen deposition were evaluated at different time
points after treatment with BSSPD loaded with B-sEVs. Interestingly, during the end of the maturation phase in the in vivo
model, tissues in the groups treated with BSSPD loaded with sEVs for sequential release (SR-sEVs@BSSPD) exhibited a more
uniform vascular structure distribution, more regular collagen arrangement, and lower volume of hyperplastic scar tissue than
tissues in the other groups. Hence, SR-sEVs@BSSPD based on skin repair phases was successfully designed and has
considerable potential as a cell-free therapy for scarless wound healing.
KEYWORDS: thiolated alginate/polyethylene glycol diacrylate hydrogels, sequential release, small extracellular vesicles, scarless,
wound healing

scar (HS) formation,9 Complications of HS, including itching,

W ound healing is a complex and orderly physiological


process.1−3 When the integrity of the skin is
damaged, the body relies on an intricate interplay
among numerous biological pathways and various cell types to
compression, functional limitations, and anatomical deform-
ities, continue to irritate patients after the initial damage.10,11
Surgical resection is the most common therapeutic approach
restore tissue integrity and function.4,5 The process of skin for scarring; however, patients must go through the healing
wound healing is often accompanied by scar formation in process again, and the clinical therapeutic effect is often
adults.6 In the initial phase of healing, highly active fibroblasts unsatisfactory.10,12 Therefore, a therapeutic strategy allowing
promote wound closure and healing,7 but in the late phase of
wound healing, excessive deposition of the collagen and Received: September 13, 2020
extracellular matrix (ECM) caused by highly active fibroblasts Accepted: March 8, 2021
leads to inappropriate scar formation,8 which affects the Published: March 16, 2021
functional recovery of the original tissue. In particular,
excessive collagen and ECM deposition during wound healing
in severely burned patients results in keloid and hypertrophic

© 2021 American Chemical Society https://dx.doi.org/10.1021/acsnano.0c07714


6352 ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

rapid wound healing without scar formation is urgently and vascularization, which can instead result in HS
needed; such a strategy would benefit millions of patients formation.40 Therefore, sequential release of different bioactive
and has tremendous medical value. factors to fulfill the requirements of different wound healing
Recently, in tissue regeneration, considerable attention has phases may be a strategy to promote scarless wound healing.
been devoted to small extracellular vesicles (sEVs),13 which In the present study, we combined material technology and
have diameters between 30 and 200 nm, act as vehicles of biological methods to achieve optimal therapeutic outcomes.
bioactive proteins, DNAs, mRNAs, and noncoding RNAs, and Sodium alginate (SA), a natural marine polysaccharide, has
are increasingly considered important mediators of intercellular been widely applied in tissue engineering due to its excellent
communication,14 potential therapeutic bioactive factors, and biocompatibility.41−44 Our previous study indicated that
useful as continuation of stem cell therapy.15 Importantly, thiolated alginate in situ cross-linked (SA-SS-SA) hydrogels
scholars found that sEVs can be engineered or modified in a are potential hemostatic biomaterials.45 However, as a
modular fashion to improve therapeutic effects and overcome hemostatic agent, SA-SS-SA hydrogels degrade in liquid within
disadvantages.2,16−19 Liu et al. found that engineered human 48 h. By adjusting the grafting rate and optimizing the PEG
adipose stem cell sEVs delivered miR-21-5p to promote the ratio, hydrogels were fabricated to meet wound dressing
healing of diabetes-associated skin wounds.20 Ding et al. requirements. Different thiolated alginate/PEG diacrylate (SA-
suggested that sEVs derived from deferoxamine precondi- SH/PEG-DA) ratios endowed the lower and upper layers of
tioned stem cells could promote wound healing by increasing the hydrogel with different mechanical strengths and
proangiogenic ability.21 MicroRNAs (miRNAs) are a class of degradation rates, leading to spatial and temporal differences
small noncoding RNAs that play pivotal roles in multiple in bioactive factor release behavior. Therefore, bilayered SA-
biological events, including cell proliferation, differentiation, SH/PEG-DA hydrogels with different degradation rates for the
and apoptosis. MiR-29b regulates mitochondrial dysfunction sequential release of sEVs were fabricated as wound dressings.
and apoptosis in HEI-OC1 cells through the SIRT1/PGC-1α Based on the above rationale, an advanced strategy to
signaling pathway.22 MiR-29b-3p contained in sEVs released fabricate bilayered thiolated alginate/PEG diacrylate hydrogels
by human bone-marrow-derived mesenchymal stem cells loaded with sEVs for sequential release was developed in this
(hBMSCs) is a potential target for regulating aging-related study, aiming to overcome the problem of scar formation in
insulin resistance.23 In addition, previous studies demonstrated wound healing. Bilayered thiolated alginate/PEG diacrylate
that miR-29b suppressed collagen deposition and fibrotic gene (BSSPD) loaded with sEVs for sequential release (SRsEVs@
expression in scar tissues24,25 and might be a therapeutic target BSSPD) hydrogels were designed based on different phases of
for scarred wound healing.26−29 Therefore, miR-29b-3p- wound healing; these hydrogels not only promoted wound
enriched bone marrow derived mesenchymal stem cells (B- healing but also inhibited HS, as validated by in vitro and in
miR-29b-sEVs) might be a potential therapeutic intervention vivo studies.
to inhibit HSs.
Simple biological methods cannot easily simultaneously RESULTS AND DISCUSSION
promote wound healing and inhibit scar formation.30,31 The The skin is not only a vital organ for protecting internal
wound healing process typically comprises the coagulation, environmental stability, defending against invasion from
inflammation, proliferation, and maturation phases.32 The outside the body, evacuating moisture, and regulating body
wound healing response is dynamic, and the expression and temperature but also an immune system component.46,47
production of growth factors and cytokines are strictly When damage to the skin barrier occurs and invasion leads to a
spatiotemporally regulated to ensure that the correct wound wound, a complex healing cascade is activated, which can
healing events occur in the correct order.33,34 In the quickly restore the integrity and barrier function of skin
coagulation phase, the platelets release vasoactive substances tissue.47 As noted above, the dynamic wound healing process
such as 5-hydroxytryptamine (5-HT) and prostaglandins encompasses multiple overlapping events and involves a
(PGs), which further constrict the blood vessels and slow complex array of cellular and biochemical reactions,48
blood flow. Simultaneously, the release of phospholipids and including inflammatory cell recruitment, matrix deposition,
adenosine diphosphate induces additional platelet aggrega- epithelialization, and scar formation. Scars consist of fibrous
tion.35 Vasoconstriction leads to local tissue ischemia, resulting hyperplastic tissue with immature collagen. In this study, we
in the release of histamine and other vasoactive substances, combined material technology with sEVs therapy to rapidly
further leading to local vasodilation on the wound surface in promote wound healing and inhibit HS formation. sEVs
the inflammatory phase.36 During the early proliferation phase, therapy was used as the basis for modulating biological
vascular endothelial growth factor (VEGF) promotes tissue functions, while the BSSPD hydrogel was applied to allow
repair by expanding endothelial cells and increasing vascular sequential release of sEVs to meet the requirements during
permeability.37 Basic fibroblast growth factor (bFGF) and different repair phases.
platelet derived growth factor (PDGF) mainly regulate ECM Identification of sEVs. Recently, therapeutic sEVs have
component synthesis and deposition, but they can also been considered a continuation of stem cell therapy, which
enhance wound angiogenesis.38 PDGF stimulates macrophages might be a valuable direction for scarless wound healing.49 EVs
and the secretion of other growth factors important in the comprise four main types: exosomes, microvesicles, large
healing process as well as some matrix molecules such as oncosomes, and apoptotic bodies.50,51 Exosomes secreted by
fibronectin, collagen and proteoglycan.39 Dysregulation of the stem cells can promote angiogenesis and bone regenera-
wound repair process leads to excessive collagen and ECM tion.52,53 Compared with traditional stem cell therapy, sEVs
deposition; thus, normal tissue is replaced by permanent scar have the advantages of high stability, a lack of tumorigenicity,
tissue. During the late proliferation and maturation phases, easy storage, and quantitative use.14 A recent study confirmed
various cytokines, such as transforming growth factor β1 that MSC-derived exosomes can inhibit the acute inflamma-
(TGFβ1), VEGF, bFGF and PDGF, cause excessive fibrosis tory response in the wound microenvironment, accelerate
6353 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 1. (A) Representative TEM image of B-sEVs. (B) Presence of sEVs surface marker proteins (CD9, Alix, and Tsg101). (C) Size
distribution of sEVs. (D) Image of hBMSCs. (E) Uptake of sEVs by EA.hy926 cells and HFF-1 cells.

angiogenesis, promote epithelial cell proliferation and migra- narrowed our investigation and selected 52 highly possible
tion, and regulate collagen regeneration and deposition.54 targets (Figure 2A). Based on the intersection of 52 target
After isolation of sEVs from the cell culture supernatant, the genes and the antifibrogenic gene sets (Figure 2B), 6 putative
sEVs secreted by BMSCs (B-sEVs) were characterized in terms genes were selected from the various candidates. Among these
of morphology, size, and surface marker expression. Trans- 6 putative genes, Col1A1 was identified as a target gene of
mission electron microscopy (TEM) observations revealed miR-29b-3p based on bioinformatic algorithms. Bioinformatic
that the B-sEVs had a round spherical or cup-shaped algorithms showed that miR-29b-3p can bind to the 3′UTR of
morphology. Western blot analysis showed that B-sEVs were Col1A1 (Figure 2C). The luciferase reporter assay indicated
positive for expression of the exosomal marker proteins Alix, that miR-29b-3p mimics had an inhibitory effect on Col1A1-
TSG101, and CD9 (Figure 1B). Nanoparticle tracking analysis 3′UTR expression levels but not on Col1A1-3′UTR mutant
(NTA) showed that the sEVs were 30−100 nm in diameter (Mut) expression levels. These results confirmed that Col1A1
(Figure 1A, C). These results indicated that the collected B- is a direct target gene of miR-29b-3p and can be inhibited by
sEVs possessed typical morphological and molecular features. miR-29b-3p.
Successful delivery into key cells involved in wound healing, Effective miRNA delivery to target cells is a major problem
such as endothelial cells and fibroblasts,55 is a prerequisite to a that restricts miRNA-based clinical applications.63 As nano-
mechanistic understanding of sEV therapy.56 A membrane- carriers, sEVs protect key molecules, particularly RNA-like
labeling technique was used in which the PKH-26 red molecules, from being degraded by enzymes in plasma and
fluorescent exosome dye binds a green fluorescent dye triggering an immune response; they also help key molecules
(PKH-67) with a longer lipid tail on the lipid bilayer of the to penetrate various biofilms.64 Previous studies demonstrated
sEV membrane to produce a yellow-orange fluorescence. that the use of sEVs as therapeutic carriers had high application
Confocal micrographs showed obvious evidence of the cellular potential and great value for the treatment of various
attachment and internalization of B-sEVs by EA.hy926 cells diseases.65,66 Researchers have developed a variety of physical
and HFF-1 cells, and PKH-26-sEVs (red) were observed to and chemical methods to package exogenous interfering RNAs
surround the nucleus (blue). The cytoskeleton is stained green and chemical drugs into sEVs.67,68 In our study, miR-29b-3p
in Figure 1E. was overexpressed by lentivirus-mediated transfection to
Mechanism of miR-29b-3p and B-miR-29b-sEVs in generate engineered sEVs (Figure 2D). The miR-29b-3p
Scarless Wound Healing In Vitro. miRNAs play a critical lentiviral vector was obtained from Oligobio (Beijing, China).
role by binding to the 3′UTR of target genes or suppressing Lentiviral transfection was performed according to the
their transcription to reduce their expression level.57−59 In manufacturer’s protocol. In brief, hBMSCs were incubated in
previous studies, miR-29b was considered a key molecule in lentivirus-containing supernatant with 5 mg/mL of polybrene
inhibiting tumor angiogenesis.60 MiR-29b expression is for 24 h. After infection for 48 h, hBMSCs were selected with
significantly reduced in several tumors and is closely related puromycin dihydrochloride (Thermo Fisher). In general, when
to prognosis.61 Coincidentally, the level of miR-29b is also the miRNA target level in producer cells changed, the
closely related to the occurrence of scarring.62 MiR-29b-3p was corresponding miRNA level in sEVs showed the same change
predicted by three major miRNA target databases that were trend. MiRNA sorting to exosomes is modulated by cell
used to identify possible targets. Based on the intersection of activation-dependent changes in miRNA expression levels in
the miRNAs predicted as targets by the three databases, we the producer cells.69−71 Therefore, miRNA-29b-3p levels in
6354 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 2. (A) miR-29b-3p target prediction via the miRWalk, miRDB, and TargetScan databases. (B) Searching for miR-29b-3p target genes
via the intersection of possible miR-29b-3p targets and a collagen deposition gene set. (C) Binding sequences of Col1A1 and miR-29b-3p.
(D) Luciferase reporter activity of hsa-miR-29b-3p and Col1A1. (E) Lentiviral transduction efficiency. (F) Relative miRNA expression levels
compared with those in unmodified hBMSCs. (G, H) Akt, Smad3, Erk1/2, and TGF β1 phosphorylation levels in EA.hy926 and HFF-1 cells.

sEVs extracted from miR-29b-3p-enriched hBMSCs were also with B-miR-29b-sEVs downregulated the expression of those
high. Quantitative real-time polymerase chain reaction (qRT- proteins (Figure 2H). Regarding the mechanism of B-sEVs,
PCR) analysis confirmed significant upregulation of miR-29b- vascularization and fibrosis were promoted via activation of the
3p in miR-29b-3p-enriched hBMSCs and B-miR-29b-sEVs PI3K/Akt, Erk1/2, and Smad3/TGFβ1 signaling pathways. In
(Figure 2F). contrast, inhibition of these signaling pathways was observed in
One classical marker of HS is disproportionate fibrosis.55 the B-miR-29b-sEVs group. The PI3K/Akt, Erk1/2, and
Excessive ECM production, deposition and contraction Smad3/TGFβ1 signaling pathways are the most important
eventually lead to morphological changes and dysfunction at signaling pathways in the progression of vascularization and
the organ level.72,73 Excessive VEGF production and aberrant fibrosis. A recent study demonstrated the suppressive effect of
vascularization promote scar tissue formation through various miR-29b on the essential activity of Smad3 in TGF-β1-
mechanisms, such as compromised homeostasis and impaired mediated fibrosis.62 TGF-β1/Smad3 expression varies in
degradation and excessive accumulation of ECM.40 Scarless distinct stages of human growth; the TGF-β1/Smad3 gene is
healing and significantly reduced angiogenic responses are not expressed in the fetus, possibly explaining one mechanism
common features of oral and fetal wounds.74 The protein levels of scarless wound healing in fetal skin.75 Future treatments
of p-type AKT and p-type Erk1/2 in EA.hy926 cells were may be developed by investigating the molecular basis of
increased by treatment with B-sEVs. Similarly, the protein wound healing, such as blocking “pro-scarring” molecules such
levels of TGF-β1 and p-type smad3 in HFF-1 cells treated with as TGF-β1 and promoting the expression of TGF-β3, which
B-sEVs were increased (Figure 2G). By contrast, treatment aims to create an environment similar to that of the scarless
6355 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 3. Proliferation of EA.hy926 cells (A) and HFF-1 cells (B) incubated under different conditions. (C) Effects of the control, B-sEVs, B-
miR-29b-sEVs and B-NC-sEVs treatments on EA.hy926 and HFF-1 cell migration and EA.hy926 cell tube formation. Quantitative evaluation
of EA.hy926 (D) and HFF-1 (E) cell migration. (F) Quantitative evaluation of nodes. VEGF secretion from EA.hy926 cells (G) and Col I
secretion from HFF-1 cells (H). Immunofluorescence staining images of EA.hy926 (I) and HFF-1 cells (J).

healing process.75 These results imply that B-miR-29b-sEVs with B-miR-29b-sEVs inhibited VEGF and COL I expression.
might be a basis for the treatment of HS. Immunofluorescence (IF) assays confirmed the ELISA results
To further verify the hydrolytic action of B-sEVs and B-miR- (Figure 3I,J). The standard curve for ELISA is shown in Figure
29b-sEVs, additional in vitro evidence was provided. Pro- S1A,B. Studies have shown that miR-29b mimics can
liferation (Figure 3A,B), migration, tube formation, and significantly inhibit the excessive proliferation of blood vessels
VEGF/COL I expression and secretion were promoted in and collagen deposition in endothelial cells and fibroblasts.62
the B-sEVs and B-NC-sEVs groups of EA.hy926 and HFF-1 The inhibitory effect of B-miR-29b-sEVs on EA.hy926 and
cells. In contrast, the related functions were inhibited in the B- HFF-1 cells further demonstrated the feasibility of treatment
miR-29b-sEVs group of these two cell lines. On day 7, the with engineered sEVs.
number of proliferating cells in the B-miR-29b-sEVs group was Characterization of Hydrogels. Figure 4A shows a
significantly lower than that in the other groups. Similarly, both general view of the BSSPD hydrogels. Raman spectroscopy
the number of cells passing through the Transwell insert of SA-SH/PEG-DA (Figure 4B) showed no absorption peak at
membrane and the number of blood vessels on the Matrigel approximately 524 cm−1, indicating that no disulfide bonds
were lower in the B-miR-29b-sEVs group than in the other existed in the SA-SH/PEG-DA hydrogels. Figure 4C and
groups. (Figure 3C−F). The concentrations of VEGF and Col Figure S2A show the FTIR spectra of SA-SH/PEG-DA. Peaks
I in the cells were determined by enzyme-linked immuno- at wavenumbers of 1722 and 1105 cm−1 belonged to the
sorbent assay (ELISA) (Figure 3G,H). After culture for 3 days, characteristic peaks of carbonyls (PEG-DA) and ethers (PEG-
the supernatant of EA.hy926 cells in the B-sEVs and B-NC- DA), respectively. Combined, the Raman spectra and FTIR
sEVs groups had higher VEGF and COL I concentrations than spectra results proved that SA-SH and PEG-DA were cross-
that from the corresponding groups of HFF-1 cells. Treatment linked by thiol−ene click chemical bonds rather than disulfide
6356 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 4. (A) A photograph of SA-SH/PEG-DA hydrogel biolayers. (B) Raman spectra of SA-SH/PEG-DA hydrogels. (C) FTIR spectra of
SA-SH/PEG-DA hydrogels. (D) Swelling rate of SA-SH/PEG-DA hydrogels. (E) Scanning electron micrographs of lyophilized SA-SH/PEG-
DA hydrogels. (F) Fluorescent sEV tracer experiment: Blue indicates the nucleus, red indicates B-sEVs, and green indicates B-miR-29b-
sEVs. (G) Statistical analysis of the fluorescent sEV tracer experiment. (H) Curves showing the release of B-sEVs and B-miR-29b-sEVs in
PBS. (I) Statistical analysis of hydrogel degradation in PBS and cell culture medium. (J) Images of hydrogel degradation in PBS and cell
(EA.hy926) culture medium. (K) Reaction equation and diagram of the release assay.

bonds. The SA-SH-3/PEG-DA-700 hydrogels had the same networks. The swelling rate of the SA-SH-3/PEG-DA-700
cross-linking bonds and similar chemical structures as the SA- hydrogel with more hydrophobic network was lower than that
SH-3/PEG-DA-250 hydrogels, and the difference was the of SA-SH-3/PEG-DA-250 (Figure 4D and Figure S2B), and
molecular weights of the PEG-DAs. However, with the the difference in swelling rate between them was more than
increase of molecular weight of PEG-DA, the hydrophilicity 50%. Hydrogel group cross-linked by PEG-DA-700 had a
of its molecular chain gradually decreased which resulted in the flatter micromorphology than PEG-DA-250 group (Figure 4E
different hydrophilicity of the two SA-SH/PEG-DA hydrogel and Figure S2C). PKH-76 is a green fluorescent dye that can
6357 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 5. (A) Changes in the wound closure areas. (B) Simulation plots. (C) Quantitative evaluation of the wound closure rate. (D, E)
Images and quantitative analysis of H&E staining.

be used to label living cells. In this study, lipid molecules with hydrogel (Figure 4H). To detect BSSPD degradation, BSSPD
sEV membrane structures were labeled with green fluores- was placed in a Transwell chamber and transferred to a well
cence. The upper and lower layers of the hydrogel were loaded plate for detection in PBS and medium cell culture. Figure 4I,J
separately with 1 × 1010 B-sEVs and B-miR-29b-sEVs. Two shows a macroscopic illustration of the aqueous gel and the
different labeled sEVs were loaded into the hydrogels: B-miR- data analysis. The upper layer is dyed blue, and the lower layer
29b-sEVs were stained with PKH-76 (green), and B-sEVs were is dyed red. In PBS, the hydrogels were degraded slowly, and
stained with PKH-76 (red). The fluorescent images (Figure the hydrogels in the lower layer of culture medium were
4F) and statistical analysis (Figure 4G) showed that the degraded first and completely at day 7. The upper layer was
number of B-sEVs increased slowly with increasing immersion
degraded slowly at the beginning, accelerated after day 7, and
time. After 7 days, B-miR-29b-sEVs began to accumulate
degraded to 10% at day 14. In addition, ROS produced during
gradually. Both sEVs surrounded the nucleus (blue). The
cell culture76 could cause the disruption of glycosidic bonds on
amount of sEVs released was detected by an ELISA kit.
According to the curve in the figure, 80% of the sEVs of the the main chain of alginate,77,78 which accelerated the
SA-SH-3/PEG-DA-250 hydrogel were released at day 7, and degradation of SA-SH/PEG-DA hydrogels. In general, SA-
the release became slower after that. The SA-SH-3/PEG-DA- SH-3/PEG-DA-700 hydrogels had high storage modulus
700 hydrogel released 80% of sEVs at day 9, and the curve (Figure S2D), low swelling rate, and low sEVs release rate,
decreased after day 9. The sEVs release from the hydrogel into while SA-SH-3/PEG-DA-250 hydrogels had low storage
the PBS was a physical diffusion process, so the sEVs release modulus, high swelling rate and high sEVs release rate.
rate of the SA-SH-3/PEG-DA-700 hydrogel with a lower Therefore, hydrogels with different sEV release rates could play
swelling rate was lower than that of the SA-SH-3/PEG-DA-250 a graded release role in wound healing.
6358 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 6. (A) Reconstructed 3D images of new blood vessels and quantitative analysis of vessel number (B) and vessel area (C). (D) IHC
(CD31) and IF (CD31 and α-SMA) staining images. (E, F) Morphometric analysis of new and mature blood vessels.

In addition, the morphology of cells in these hydrogels was Blood vessels in each group were systematically evaluated by
not significantly altered (Figure S3A). The number of cells in microcomputed tomography (micro-CT), immunohistochem-
the SA-SH-3/PEG-DA-250 group and SA-SH-3/PEG-DA-700 istry, and immunofluorescence. The reconstructed three-
group on days 1, 3, and 7 was not significantly different from dimensional (3D) image (Figure 6A), immunohistochemical
that in the control group (Figure S3B). SA-SH/PEG-DA (IHC) staining, and dual immunofluorescence staining (Figure
hydrogels had excellent biocompatibility and were suitable for 6D) showed that both the density and number of blood vessels
cell growth. Most importantly, SA-SH/PEG-DA hydrogels in the SR-sEVs@BSSPD group and B-sEVs@BSSPD group
were compatible with skin healing and could deliver different were significantly higher than those in the control group,
sEVs to achieve sequential sEV release, indicating their BSSPD group, and BSSPD loaded with B-miR-29b-sEVs (B-
potential as dressings for scarless wound healing. miR-29b-sEVs@BSSPD) group. The same results were
Wound Healing In Vivo. Figure 5A shows images acquired obtained by quantitative analysis of the area of the region
with a digital camera, simulation diagrams (Figure 5B), and covered by new blood vessels and the number of new blood
vessels (Figure 6B,C) and the numbers of total blood vessels
quantitative data analysis results (Figure 5C) of the wounds in
and mature blood vessels (Figure 6E,F). The results of in vitro
each group at four time points. Results of the H&E staining are
experiments showed that B-miR-29b-sEVs had antivasculariza-
shown in Figure 5D that the B-sEVs@BSSPD and SR-sEVs@ tion and antifibrotic properties. However, analysis of the in vivo
BSSPD groups exhibited significantly faster wound healing experimental results indicated that B-miR-29b-sEVs released
than the others. Consistent with the quantitative analysis of the from the upper layer did not significantly affect wound healing
wound healing results on days 7 and 14, the B-sEVs@BSSPD and angiogenesis in the SR-sEVs@BSSPD group.
group and SR-sEVs@BSSPD group showed significantly more In the main skin structure, collagen fibers are divided into
favorable results than the other groups, although the difference type I and type III.79 Type I collagen is thick and is the main
between the B-sEVs@BSSPD group and SR-sEVs@BSSPD component of skin tissue, while type III collagen is thin and is
group was not significant (Figure 5E). Regarding wound the main component of reticular fibers. The proportions of
healing, no obvious impact of the antiangiogenic, antifibrotic collagen I and III in normal skin are affected by coordinated
effect of B-miR-29b-sEVs was seen in the SR-sEVs@BSSPD expression during development, and the normal proportions of
group. collagen I and III maintain the normal skin tissue structure.80
6359 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Therefore, the arrangement and distribution of collagens and effects during the different phases of wound healing. In the
the proportions of type I and type III collagen are important early wound healing phase, M2 macrophages inhibit the acute
criteria for assessing wound healing.81 The Masson staining inflammatory response in the wound microenvironment,
results (Figure 7A) indicated that collagen deposition was accelerate angiogenesis, promote epithelial cell proliferation
and migration, and regulate collagen regeneration and
deposition.82 Paradoxically, the M2 phenotype is a regulator
of both healing and scar hyperplasia. Moreover, they are the
main components in the HS microenvironment and play a key
role in both initial and supporting scar formation.83 In the late
wound healing phase, the M2 phenotype promotes excessive
fibroblast proliferation, resulting in dense fibrous tissue
overgrowth and excessive collagen deposition.83 Considering
the different roles of macrophage polarization at different
phases in the physiological wound healing process, we further
explored the mechanism of action of the two types of sEVs in
the relationship between sEVs and inflammation.
The in vitro flow cytometry results shown in Figure 8A,B
indicate that B-miR-29b-sEVs induced M1 macrophage
polarization and B-sEVs induced M2 macrophage polarization.
Moreover, B-miR-29b-sEVs reversed B-sEV-induced M2
macrophage polarization, as M2 macrophages were depleted
and M1 macrophages were enriched (Figure 8C−E). The
relative expression levels of genes, as determined by RT-PCR,
also proved the immunomodulatory function of B-miR-29b-
sEVs and B-sEVs. Anti-inflammatory genes (Arg1 and IL-4)
were upregulated by B-sEVs, and proinflammatory genes (IFN-
γ and IL-1β) were upregulated by B-miR-29b-sEVs (Figure
8F,G). Consistent with the flow cytometry results, proin-
flammatory genes were upregulated and anti-inflammatory
genes were downregulated in the B-sEVs/B-miR-29b-sEV
group. These results indicated different immunomodulatory
functions of B-miR-29b-sEVs and B-sEVs and suggested that
B-miR-29b-sEVs attenuated B-sEV-induced M2 polarization.
The immunomodulatory function was further characterized
in vivo by immunofluorescence staining (Figure 8H). At 7 days,
M2 polarization was significantly enhanced in the B-sEVs@
BSSPD and SR-sEVs@BSSPD groups compared to that in the
Figure 7. (A) Masson staining showing collagen deposition. (B) other groups, while the high CD206 expression was decreased
Representative slides stained with Sirius red. Sirius red staining in the B-miR-29b-sEVs@BSSPD group. M2 macrophage
can distinguish type I and type III collagen. Type I collagen is polarization was suppressed by B-miR-29b-sEVs, thus exacer-
stained yellow, and type III collagen is stained green. (C) Analysis bating the proinflammatory response in the early phase and
of the collagen I/III ratio in the five different treatment groups. inhibiting rapid epithelialization and wound healing. As the
time of wound healing is prolonged and the inflammatory
more extensive and the collagen fiber thickness was greater in response continues, the number of macrophages remains
the B-sEVs@BSSPD group than in the control group, while the high.84,85 Here, as the healing process was delayed and a large
collagen arrangement in the SR-sEVs@BSSPD group was more number of macrophages were recruited to the wound site,
regular and orderly. The arrangement of collagen fibers was CD206 expression in the B-miR-29b-sEVs@BSSPD group was
improved in the SR-sEVs@BSSPD group compared with the found to be high on day 14. In contrast, M2 polarization of
control group and was similar to that of normal skin, indicating macrophages was weakened in the B-sEVs@BSSPD and SR-
the positive effect of SR-sEVs@BSSPD treatment on ECM sEVs@BSSPD groups on day 14, as the wounds in these two
deposition and collagen arrangement. As shown in Figure groups were almost completely closed. Interestingly, M2
7B,C, the collagen I/III ratio in each group increased on day 7 polarization of macrophages was much weaker in the SR-
due to the increase in type I collagen. The B-sEVs@BSSPD sEVs@BSSPD group than in the B-sEVs@BSSPD group
group and SR-sEVs@BSSPD group had significantly higher (Figure 8I) due to the introduction of B-miR-29b-sEVs in
scores than the control group, BSSPD group, and B-miR-29b- the later phase. Based on these in vitro and in vivo results,
sEVs@BSSPD group. On day 14, the type I/III collagen ratios sequential release of B-sEVs and B-miR-29b-sEVs by SR-
in the three sEV treatment groups were lower than those in the sEVs@BSSPD led to M2 polarization in the early phase and
control group. Importantly, the type I/III collagen ratio in the attenuated M2 polarization in the later phase. Considering the
SR-sEVs@BSSPD group was more similar to that in normal biological phase of wound healing, rapid initiation of wound
skin than were the ratios in the other groups. healing and subsequent inhibition of excessive collagen
Macrophages are indispensable for tissue repair and production and fibrosis could be achieved by the immunor-
remodeling and play an essential role in cutaneous wound egulatory function of SR-sEVs@BSSPD. Therefore, sequential
healing. In particular, M2 polarization exerts distinct biological release of sEVs from BSSPD could account for both the speed
6360 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 8. (A) Histograms of CCR7 and CD206 expression after treatment with B-sEVs or B-miR-29b-sEVs for 48 h, as determined by flow
cytometry. (B) Quantitative analysis of the data in the histograms. (C) Histograms of CCR7 and CD206 expression after treatment with B-
sEVs or B-miR-29b-sEVs and sequential release of B-sEVs (48 h) and B-miR-29b-sEVs (48 h) for 96 h, as determined by flow cytometry. (D)
Quantitative analysis of the data in the histograms. (E) Analysis of the M2/M1 ratio. (F, G) Relative gene expression levels of Arg1, IL-4,
IFN-γ, and IL-1β. (H) Immunofluorescence staining of macrophages in vivo on days 7 and 14 (DAPI: blue, CD206: green). (I)
Immunofluorescence analysis of M2 polarization.

and the quality of wound healing to promote wound healing tion.88 Therefore, the classical rat model of full-thickness skin
and reduce HS. defects has natural limitations in the evaluation of hypertrophic
Rodent models of full-thickness skin wounds have scarring. A previous study showed that in rabbit ears,
fundamental biological and structural limitations in their epidermal hyperplasia occurs after wound healing, which is
ability to faithfully represent scarring in humans.86 For considered to be similar to hyperplastic scarring in humans.89
instance, rats have a panniculus carnosus, a muscle that Therefore, we used rabbit ear scar models to evaluate the effect
promotes wound contraction, in their dorsal skin.87 However, of different treatments on scarless wound healing. The wound
in human wounds, the healing process is driven mainly by healing rates in the B-sEVs@BSSPD and SR-sEVs@BSSPD
granulation and reepithelialization. Additionally, after wound groups were consistent with the results in Sprague−Dawley
healing, scars on the dorsal skin of rats rapidly shrink and are (SD) rats (Figure 9A,B). The collagen in the SR-sEVs@
not obvious, hindering long-term studies on scar reconstruc- BSSPD-treated rabbit ears was more orderly and more similar
6361 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Figure 9. (A) Changes in the wound closure areas in the rabbit ear were observed 0, 1, 2, and 4 weeks after surgery. (B) Traces of wound
closures. (C) Masson staining of wounds. (D) Ultrasonography of wounds: (a) Control, (b) BSSPD, (c) B-sEVs@BSSPD, (d) B-miR-29b-
sEVs@BSSPD, and (e) SR-sEVs@BSSPD. (E) Quantitative analysis of the SEI.

to that in normal skin sections than was the collagen in the TGF-β1 signaling pathways. In contrast, in the late
other treatment groups (Figure 9C). In addition, the skin in proliferation and maturation phases of wound healing, B-
the SR-sEVs@BSSPD group had a lower protrusion height miR-29b-sEVs released from the upper layer of the hydrogels
than that in the B-sEVs@BSSPD and B-miR-29b-sEVs@ inhibited excessive angiogenesis and collagen deposition, thus
BSSPD groups (Figure 9D). Doppler ultrasound was further reducing the formation of HSs. Collectively, these biomedical
utilized to measure scar thickness. Quantitative Doppler functions of SR-sEVs@BSSPD constitute an effective sympto-
ultrasound analysis showed that the scar elevation index matic treatment strategy that aims to improve skin
(SEI) in the SR-sEVs@BSSPD group was smaller than that in regeneration and inhibit HS.
the other four groups, indicating minimal scar formation in this
group (Figure 9E). In brief, SR-sEVs@BSSPD reduced HS METHODS
formation without affecting the wound closure speed.
This study was approved by the ethics committee of Shanghai Jiao
CONCLUSION Tong University Affiliated Sixth People’s Hospital, School of
Medicine, Shanghai Jiao Tong University. All animal experiments
Herein, we designed a strategy for the development of scarless were approved by the animal research ethics committee of Shanghai
wound healing dressings and the etiological treatment of scars Jiao Tong University Affiliated Sixth People’s Hospital and were
based on sequential sEVs releasing from SH/PEG-DA conducted in accordance with the National Institutes of Health Guide
hydrogels. Bilayered SA-SH/PEG-DA hydrogels were fabri- for the Care and Use of Experimental Animals (Animal Welfare Ethics
cated by thiol−ene click chemistry, exhibiting promising acceptance number (approval number) DWLL2020-0522).
wound protection ability and sequential sEVs release. In the Lentivirus Transfection. The miR-29b-3p lentiviral vector was
coagulation, inflammation, and early proliferation phases of obtained from Oligobio (Beijing, China). The transfection protocol
was performed according to the manufacturer’s instruction. In brief:
wound healing, B-sEVs released from the lower layer of the hBMSCs were incubated in retro-viral supernatant with 5 mg/mL of
hydrogels promoted angiogenesis and collagen deposition, polybrene for 24 h. After infection for 48 h, hBMSCs were treated
which accelerated wound healing. MiR-29b-3p was secreted by with puromycin dihydrochloride (Thermo Fisher).
B-miR-29b-sEVs to suppress endothelial cell and fibroblast Characterization of sEVs. Morphological characterization of
proliferation and migration and Col1A1 expression in vesicles was performed by TEM (JEM-1400, JEOL, Japan). Western
fibroblasts by inhibiting the PI3K/Akt, Erk1/2, and Smad3/ blot analysis was conducted to validate the sEV markers Alix (Protein

6362 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Tech, USA), TSG101 (Protein Tech, USA), and CD9 (Abcam, to dilute the lysates at a ratio of 1:5. Then, the lysates were heated to
USA). The size distribution of sEVs was determined by NTA. 95 °C for 5 min. Protein extracts were separated by 10% sodium
Internalization of sEVs. Purified sEVs were labeled with the dodecyl sulfate-polyacrylamide gel electrophoresis at 120 V for 1 h
lipophilic fluorescent PKH dye PKH-26 (Sigma-Aldrich, Germany). and transferred onto polyvinylidene difluoride membranes (Merck-
The PKH26-labeled sEVs were then resuspended in the culture Millipore) at 200 mA for 1.5 h. Membranes were blocked with 5%
medium of EA.hy926 and HFF-1 cells for 24 h. After fixation, the nonfat milk or BSA for 1 h and were then incubated with
sEVs were washed 3 times with PBS prior to immunostaining with combinations of primary antibodies overnight at 4 °C. Next,
QuickBlock blocking buffer (Beyotime, China). Then, the cytoske- membranes were incubated with combinations of horseradish
leton was stained with phalloidin (1:200, Yeasen, China) for 45 min at peroxidase-conjugated secondary antibodies for 1 h at room
room temperature prior to 3 washes with PBS for 5 min each. After temperature. The immunoreactive bands were visualized with an
the final wash, 4′,6-diamidino-2-phenylindole (DAPI; 1:2000, Solar- enhanced chemiluminescence reagent (Thermo Fisher Scientific).
bio, China) in PBS was added, and cells were stained for 10 min Characterization of Hydrogels. The surface morphology of the
before imaging via confocal microscopy (Leica, Solms, Germany). six freeze-dried SA-SH/PEG-DA hydrogels was observed by scanning
qRT-PCR. MiR-29-3p- and RNU6B (BioTNT, Shanghai, China) electron microscopy (S-3400N, Hitachi, Japan) at an acceleration
miRNA-specific qRT-PCR detection kits were used for cDNA voltage of 15 kV. The samples were sputtered with gold for 45 s
synthesis and qRT-PCR. MiR-29-3p expression levels were before visualization.
normalized to those of RNU6B. cDNA synthesis was performed Raman spectroscopy was carried out with an inVia Reflex Raman
using TransScript All-in-One First-Strand cDNA short SuperMix for spectrometer (Renishaw Company, UK). The wavelength was fixed at
qPCR (Transgen Biotech, Beijing, China). TransStart Top Green 514.5 nm, and the scanning range was set at 4000 cm−1 to 400 cm−1.
qPCR SuperMix (Transgen Biotech) was used for qRT-PCR, and the The sample was placed on the slide for direct laser irradiation.
results were processed by normalization. A Nicolet 5700 infrared spectrometer (Thermo Scientific, USA)
Cell Proliferation and Migration Assays. CCK-8 assays were was used for FTIR spectroscopy. The scanning wavenumber range
used to analyze the proliferation of EA.hy926 cells and HFF-1 cells. was set at 4000 cm−1 to 400 cm−1.
EA.hy926 cells were cultured at different densities and under different SA-SH/PEG-DA hydrogels with different ratios were placed in PBS
conditions. EA.hy926 cells were plated in flat-bottom 96-well plates buffer (pH = 7.4) at 37 °C for measurement of the swelling rate. After
(Corning, USA) at a density of 2 × 103 cells/well and were cultured swelling for 15 min, excessive liquid was absorbed, and the hydrogels
for 1, 3, and 7 days. HFF-1 cells (1.5 × 103) were cultured in a 96-well were reweighed. The swelling rate of the SA-SH/PEG-DA hydrogels
plate under similar conditions. Then, 100 μL of medium containing (Seq%) was calculated with the following equation:
10% CCK-8 reagent was added to each plate. The plates were then
incubated for 2 h, and the absorbance of the sample was immediately Weq − Wd
Seq % = × 100%
measured at 450 nm in a microtitration plate reader (Winooski Wd
BioTek, USA). The migration of EA.hy926 cells and HFF-1 cells was
evaluated by a Transwell assay (3422, Corning, USA). EA.hy926 cells where Weq is the mass of the swollen gel, and Wd is the mass of the
(2 × 104) were seeded in the upper chambers in 200 μL of serum-free dry gel.
medium. Five hundred μL of complete medium was added to the The rheological properties of the SA-SH/PEG-DA hydrogels were
lower chambers. Then, the cells were fixed, stained, imaged by evaluated with a MARS3 rotary rheometer (Thermo Scientific, USA).
microscopy (Olympus IX 70, Tokyo, Japan), and counted with Frequency conversion scanning was used to modulate the frequency
ImageJ software. The migration of HFF-1 cells was evaluated with the from 0.01 Hz to 10 Hz. The strain was set at 1%, and the temperature
same method. was set at 37 °C.
Tube Formation Assay. Two hundred μL of Matrigel (BD Fabrication of the SR-sEVs@BSSPD Hydrogel. The photo-
Bioscience) was added to each well of precooled 24-well plates, and initiator I2959 (1 mg) was dissolved in 400 μL of deionized water,
gel electrophoresis was conducted at 37 °C for 1 h. Cells were and 4 μL of PEG-DA (Mn = 250) was added to the solution. Then,
precultured for 48 h in complete growth medium under different 100 μL of an aqueous solution of B-sEVs (1 × 1011 sEV particles/mL)
conditions. A total of 1 × 105 pretreated EA.hy926 cells were then was added, and the solution was stirred thoroughly again. The above
seeded on Matrigel-coated plates and cultured. EA.hy926 cell tube solution was then added to a sample bottle containing 5 mg of SA-SH
formation was evaluated by microscopy (Olympus IX 70, Tokyo, powder (SA:SH 3:31.35%), and the sample was immediately
Japan) after 8 h of incubation. The number of tubes was analyzed with irradiated under a UV lamp at 365 nm after rapid stirring for 10
ImageJ software. min. After UV irradiation, the SA-SH/PEG-DA hydrogel was
VEGF/Col I Secretion Assay. Cells were maintained in medium removed and placed in a dialysis bag (MWCO = 14 kDa) for dialysis
at different concentrations for 3 days, and the cells were then fixed to remove excess I2959 and PEG-DA that were not involved in the
with paraformaldehyde (PFA), and washed with PBS, permeabilized reaction. The above method was used to add the same volume of an
with 0.25% Triton X-100 in PBS, and blocked with 3% bovine serum aqueous solution of B-miR-29b-sEVs to 11 μL of PEG-DA (Mn =
albumin (BSA). Anti-VEGF/Col I (1:200, ABclonal, China) anti- 700) to fabricate the corresponding SA-SH-3/PEG-DA-250 hydrogel.
bodies were added, and the cells were then incubated at 4 °C Fabrication of the B-sEVs@BSSPD Hydrogel. The photo-
overnight. The next day, the secondary antibodies were added, and initiator I2959 (1 mg) was dissolved in 400 μL of deionized water,
the cells were incubated in the dark for 1 h. Then, the cells were and 4 μL of PEG-DA (Mn = 250) was added to the solution. Then,
washed 3 times (5 min each time) with PBS. EA.hy926 cells and 100 μL of an aqueous solution of B-sEVs (1 × 1011 sEV particles/mL)
HFF-1 cells were then incubated with 5 g/mL of phalloidin (1:200, was added, and the solution was stirred thoroughly again. The above
Yeasen, China) for 45 min in the dark. After three PBS washes, the solution was then added to a sample bottle containing 5 mg of SA-SH
cells were stained with DAPI (1:200, Solarbio). A confocal powder (SA:SH 3:31.35%), and the sample was immediately
microscope (Leica, Solms, Germany) was used to visualize the cells. irradiated under a UV lamp at 365 nm after rapid stirring for 10
VEGF/Col I secretion from EA.hy926 cells/HFF-1 cells was assessed min. After UV irradiation, the SA-SH/PEG-DA hydrogel was
by ELISA (NeoBioscience, China). EA.hy926/HFF-1 cells (1 × 105) removed and placed in a dialysis bag (MWCO = 14 kDa) for dialysis
were seeded in medium at different concentrations in 6-well plates. to remove excess I2959 and PEG-DA that were not involved in the
The supernatant of the samples was collected after 3 days of reaction. The above method was used to add the same volume of an
incubation, and the VEGF/Col I concentrations were determined in aqueous solution of B-sEVs to 11 μL of PEG-DA (Mn = 700) to
accordance with the manufacturer’s instructions. fabricate the corresponding SA-SH-3/PEG-DA-250 hydrogel.
Western Blot Analysis of EA.hy926 Cells and HFF-1 Cells. Fabrication of the B-miR-29b-sEVs@BSSPD hydrogel. The
Cultured sEVs or cells were lysed in RIPA lysis buffer containing a photoinitiator I2959 (1 mg) was dissolved in 400 μL of deionized
supplement (Invitrogen, USA). Protein loading buffer (5×) was used water, and 4 μL of PEG-DA (Mn = 250) was added to the solution.

6363 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Then, 100 μL of an aqueous solution of B-miR-29b-sEVs (1 × 1011 the area occupied by new blood vessels were calculated, and the data
sEV particles/mL) was added, and the solution was stirred thoroughly for the five groups were analyzed with ImageJ software.
again. The above solution was then added to a sample bottle IHC and Immunofluorescence Analysis. Quantitative analysis
containing 5 mg of SA-SH powder (SA:SH 3:31.35%), and the sample of IHC staining was based on previous studies.90 To assess vascular
was immediately irradiated under a UV lamp at 365 nm after rapid system formation, CD31 (1:200, Abcam, Cambridge, UK) was
stirring for 10 min. After UV irradiation, the SA-SH/PEG-DA stained by IHC methods. CD31 (1:200, Abcam, Cambridge, UK) and
hydrogel was removed and placed in a dialysis bag (MWCO = 14 α-SAM (1:50, Abcam, Cambridge, UK) were stained with
kDa) for dialysis to remove excess I2959 and PEG-DA that were not immunofluorescence methods. Sections were observed via fluores-
involved in the reaction. The above method was used to add the same cence microscopy (Olympus IX 70, Tokyo, Japan) and confocal laser
volume of an aqueous solution of B-miR-29b-sEVs to 11 μL of PEG- scanning microscopy (Leica, Solms, Germany). ImageJ Plus software
DA (Mn = 700) to fabricate the corresponding SA-SH-3/PEG-DA- was used to calculate the number of blood vessels.
250 hydrogel. Histological Analysis. Samples were dehydrated in an ethanol
ELISA of SEVs. SR-sEVs@BSSPD hydrogels were immersed in a gradient, embedded in paraffin, and cut into 6 mm thick sections
six-well plate in PBS, and the plate was placed in a cell incubator for 3, perpendicular to the wound surface. The sections were stained with
6, and 12 h and 1, 2, 3, 4, 5, 6, 7, 8, and 9 days. The materials H&E for histological evaluation. The length of the new epithelium
immersed for 9 days were dissolved, the residual content of sEVs was was determined by the changes in the wound surface as observed by
measured after 9 days, and the total number of sEVs was calculated. H&E staining to assess the effect of each treatment on
The numbers of sEV particles were determined using a sEV ELISA kit epithelialization of the wound surface.91 Masson’s trichrome staining
for CD63 according to the instructions provided. In brief, standard was applied to detect collagen fibers in tissues. The collagen fibers
curves were generated by continuous dilution of the standard with appear blue, and the muscle fibers appear red. Sirius red staining was
sEVs combined with buffer solution. used mainly to analyze the degree and characteristics of fibrosis. The
Release of Fluorescent sEVs. B-miR-29b-sEVs were stained with slices were dyed in Biebrich scarlet dye for 8−10 min. Two or three
PKH-67 and B-sEVs with PKH-26, and the two types of sEVs that containers of anhydrous ethanol were used for dehydration. The
were successfully stained were placed on the upper and lower layers of samples were sliced, transferred to clean xylene for 5 min for clearing,
BSSPD in the original order. SR-sEVs@BSSPD hydrogels stained with and sealed using neutral gum. Sirius red staining can visualize specific
the sEV dye were placed in the upper compartment of the Transwell collagen tissue; under a polarized light source, type I collagen appears
chamber in a 6-well plate, and EA.hy926 cells (2 × 103) were added to yellow or red, type III collagen appears green, and type II collagen
the lower compartment in 500 μL of complete medium. The level of appears blue, green, or pale blue. ImageJ software was used to
the liquid was flush with the upper and lower boundaries of the calculate and analyze the collagen I/III ratio in tissues.
hydrogel. The 6-well plates were incubated in cell incubators for 1, 2,
Flow Cytometric, qRT-PCR, and Immunofluorescence
3, 4, 7, 9, 11, and 14 days. The cells were removed, and the plates Analysis of Macrophagocytes. Flow cytometric analysis: After
were fixed with PFA, washed three times with PBS, and treatment of RAW264.7 cells with B-sEVs, B-miR-29b-sEVs, B-sEVs,
immunostained with QuickBlock blocking buffer for 10 min. The or B-miR-29b-sEVs for 48 and 96 h, cells were detached with PBS
plates were then washed with PBS 3 times for 5 min each. Finally, solution (4 °C) and resuspended in cell staining buffer. After blocking
after washing, DAPI was added. After staining for 10 min, the cells with a rat antimouse CD16/32 antibody (Biolegend, USA),
were washed three times with PBS in the dark. Then, the cells were
RAW264.7 cells were stained with rat antimouse CD206 Alexa
visualized by confocal microscopy, and images were acquired. ImageJ
Fluor 647 antibody (BD Pharmingen, USA) and rat antimouse CCR7
was used for measurement and statistical analysis based on the
APC (Biolegend, USA) for 30 min at 4 °C in the dark. The samples
proportions of the three colors in the images.
were evaluated and analyzed with the CytoFLEX Platform (Beckman,
Degradation of BSSPD. The upper layer of BSSPD was stained
USA). For quantification of the relative gene expression levels of
blue with 1,9-dimethylmethylene blue, while the lower layer of
Arg1, IL-4, IFN-γ, and IL-1β in RAW264.7 cells cultured with B-sEVs,
BSSPD hydrogels was stained red with rhodamine B. The BSSPD
hydrogels were placed in the upper compartments of a 6-well B-miR-29b-sEVs, and B-sEVs/B-miR-29b-sEVs, qRT-PCR was
Transwell plate. EA.hy926 cells in PBS or culture medium were applied as described above. In immunofluorescence staining images
seeded in the lower compartments. The level of the liquid was just acquired 7 and 14 days after surgery, nuclei in the rat skin specimens
above the top of the lower compartment. The plates were then appear blue, and CD206 appears green. Statistical analysis was
incubated in a cell culture incubator. Images were acquired on days 0, performed with ImageJ software.
1, 2, 3, 5, 7, 9, 11, and 14. Statistical analysis was conducted based on Rabbit Model of HS and Surgical Procedure. Fifteen healthy
the hydrogel height. New Zealand white rabbits (2.5−3 kg) were obtained from HFK
SD Rats and Surgical Procedure. Thirty male SD rats (2 (HFK Bioscience Co., Ltd., Beijing). The operation was performed in
months old, 250 ± 15 g) were selected to establish a full-thickness a sterile environment after the animals were anesthetized with sodium
skin defect model in this study. BSSPD, B-sEVs@BSSPD, B-miR-29b- pentobarbital (Sigma, 30 mg/kg). The epidermis, dermis, and
sEVs, and SR-sEVs@BSSPD hydrogels were applied to the wound pericardium of each wound were thoroughly excised. On the ventral
surfaces on the SD rats. After the operation, the wounds were covered surface of each ear, four circular wounds 10 mm in diameter were
with sterile gauze. All wounds were observed daily to ensure that the established away from the middle auricular artery and the
materials and pressure dressing remained intact. periauricular vein. We randomly divided all treated rabbits into five
Microfil Perfusion and Micro-CT. SD rats were perfused with groups (n = 3 per group): the BSSPD, B-sEVs@BSSPD, B-miR-29b-
Microfil (MV-122; Flow Tech, USA), and neovascularization was sEVs, and SR-sEVs@BSSPD groups. The wounds in the control
evaluated 14 days after surgery. The experimental animals were group were untreated, and the wounds in the different treatment
euthanized and were then perfused intracardially with 100 mL of groups were covered with the corresponding hydrogel. The wound
heparinized saline, and 20 mL of Microfil was continuously injected at healing process was monitored and recorded every day until death 4
2 mL/min. The dye entered the neovascularized area of the wound weeks after the operation. The scar thickness was calculated using an
through the cardiac vessels. Finally, the experimental samples were ultrasound instrument (Mindray M9, Mindray, China). Scar tissue
incubated in a polymerizing contrast agent at 4 °C overnight. The was removed from the surrounding uninjured (2 mm) margin. The
next day, the wound samples were trimmed using micro-CT scar tissue samples from each group were divided into three parts. The
instrument (SkyScan). Red indicates the new blood vessels in the specimens were fixed with 4% PFA for 24 h, embedded in paraffin,
wound 14 days after surgery. SkyScan software (SkyScan Company) sectioned transversely, and subjected to histological analysis. The SEI
was used to reconstruct 3D images of wound neovascularization. The was determined by Masson’s trichrome staining. The dermis in
same section was selected, and neovascularization is marked in red in Masson’s trichrome-stained tissues was imaged in 5 randomly selected
the figure. Then, the number of new blood vessels per unit area and fields.

6364 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Statistical Analysis. All data are shown as the mean ± standard Notes
deviation (SD) values. Independent samples t tests were used to The authors declare no competing financial interest.
compare means between two groups. One-way analysis of variance
(ANOVA) was used to determine the level of significance with
GraphPad Prism software, and P < 0.05 was considered to indicate ACKNOWLEDGMENTS
statistical significance. *P < 0.05, **P < 0.01, ***P < 0.001 compared The authors acknowledge the support of the National Natural
to the control group; #P < 0.05 compared to the B-sEVs@BSSPD
group; &P < 0.05 compared to the B-miR-29b-sEVs@BSSPD group.
Science Foundation of China (nos. 51972212, 51802326, and
81572178) and Shanghai Municipal Commission of Health
ASSOCIATED CONTENT and Family Planning (no. 20124356).
*
sı Supporting Information
The Supporting Information is available free of charge at REFERENCES
https://pubs.acs.org/doi/10.1021/acsnano.0c07714. (1) Willyard, C. Unlocking the Secrets of Scar-Free Skin Healing.
Nature 2018, 563, S86−S88.
Additional experimental methods and supplementary
(2) Kim, H.; Wang, S. Y.; Kwak, G.; Yang, Y.; Kwon, I. C.; Kim, S.
figures. Figure S1: Elisa standard curve. Figure S2: H. Exosome-Guided Phenotypic Switch of M1 to M2Macrophages for
Further information and data plots about SA-SH/PEG- Cutaneous Wound Healing. Adv. Sci. (Weinh). 2019, 6, 1900513.
DA hydrogels. Figure S3: Biocompatibility of SA-SH/ (3) Lim, L.; Bui, H.; Farrelly, O.; Yang, J.; Li, L.; Enis, D.; Ma, W.;
PEG-DA hydrogels (PDF) Chen, M.; Oliver, G.; Welsh, J. D.; Kahn, M. L. Hemostasis Stimulates
Lymphangiogenesis through Release and Activation of VEGFC. Blood
AUTHOR INFORMATION 2019, 134, 1764−1775.
(4) Hajialyani, M.; Tewari, D.; Sobarzo-Sanchez, E.; Nabavi, S. M.;
Corresponding Authors Farzaei, M. H.; Abdollahi, M. Natural Product-Based Nanomedicines
Hong Gao − Department of Orthopaedic Surgery, Shanghai for Wound Healing Purposes: Therapeutic Targets and Drug Delivery
Jiao Tong University Affiliated Sixth People’s Hospital, Systems. Int. J. Nanomed. 2018, 13, 5023−5043.
Shanghai 200233, China; orcid.org/0000-0001-9347- (5) Evans, J.; Infusini, G.; McGovern, J.; Cuttle, L.; Webb, A.; Nebl,
3293; Email: honggao630@163.com T.; Milla, L.; Kimble, R.; Kempf, M.; Andrews, C. J.; Leavesley, D.;
Shichang Zhao − Department of Orthopaedic Surgery, Salamonsen, L. A. Menstrual Fluid Factors Facilitate Tissue Repair:
Shanghai Jiao Tong University Affiliated Sixth People’s Identification and Functional Action in Endometrial and Skin Repair.
FASEB J. 2019, 33, 584−605.
Hospital, Shanghai 200233, China;
(6) Monavarian, M.; Kader, S.; Moeinzadeh, S.; Jabbari, E.
Email: zhaoshichang0404@163.com Regenerative Scar-Free Skin Wound Healing. Tissue Eng., Part B
Meidong Lang − Shanghai Key Laboratory of Advanced 2019, 25, 294−311.
Polymeric Materials, Key Laboratory for Ultrafine Materials (7) Long, Y.; Wei, H.; Li, J.; Yao, G.; Yu, B.; Ni, D.; Gibson, A. L.;
of Ministry of Education, School of Materials Science and Lan, X.; Jiang, Y.; Cai, W.; Wang, X. Effective Wound Healing
Engineering, East China University of Science and Enabled by Discrete Alternative Electric Fields from Wearable
Technology, Shanghai 200237, China; orcid.org/0000- Nanogenerators. ACS Nano 2018, 12, 12533−12540.
0003-2053-9284; Email: mdlang@ecust.edu.cn (8) Duffield, J. S.; Lupher, M.; Thannickal, V. J.; Wynn, T. A. Host
Responses in Tissue Repair and Fibrosis. Annu. Rev. Pathol.: Mech.
Authors Dis. 2013, 8, 241−76.
Yifan Shen − Department of Orthopaedic Surgery, Shanghai (9) Pal, P.; Dadhich, P.; Srivas, P. K.; Das, B.; Maulik, D.; Dhara, S.
Jiao Tong University Affiliated Sixth People’s Hospital, Bilayered Nanofibrous 3D Hierarchy as Skin Rudiment by Emulsion
Shanghai 200233, China Electrospinning for Burn Wound Management. Biomater. Sci. 2017, 5,
Guanzhe Xu − Shanghai Key Laboratory of Advanced 1786−1799.
(10) Amini-Nik, S.; Yousuf, Y.; Jeschke, M. G. Scar Management in
Polymeric Materials, Key Laboratory for Ultrafine Materials Burn Injuries Using Drug Delivery and Molecular Signaling: Current
of Ministry of Education, School of Materials Science and Treatments and Future Directions. Adv. Drug Delivery Rev. 2018, 123,
Engineering, East China University of Science and 135−154.
Technology, Shanghai 200237, China; Internet of Things (11) Coentro, J. Q.; Pugliese, E.; Hanley, G.; Raghunath, M.;
Research Center, Advanced Institute of Information Zeugolis, D. I. Current and Upcoming Therapies to Modulate Skin
Technology, Peking University, Hangzhou 311200, China Scarring and Fibrosis. Adv. Drug Delivery Rev. 2019, 146, 37−59.
Huanxuan Huang − Shanghai Key Laboratory of Advanced (12) Chawla, S.; Ghosh, S. Regulation of Fibrotic Changes by the
Polymeric Materials, Key Laboratory for Ultrafine Materials Synergistic Effects of Cytokines, Dimensionality and Matrix: Towards
of Ministry of Education, School of Materials Science and the Development of an in Vitro Human Dermal Hypertrophic Scar
Engineering, East China University of Science and Model. Acta Biomater. 2018, 69, 131−145.
(13) Mead, B.; Tomarev, S. Extracellular Vesicle Therapy for Retinal
Technology, Shanghai 200237, China Diseases. Prog. Retinal Eye Res. 2020, 79, 100849.
Kaiyang Wang − Department of Orthopaedic Surgery, (14) Pegtel, D. M.; Gould, S. J. Exosomes. Annu. Rev. Biochem. 2019,
Shanghai Jiao Tong University Affiliated Sixth People’s 88, 487−514.
Hospital, Shanghai 200233, China (15) Fuhrmann, G.; Chandrawati, R.; Parmar, P. A.; Keane, T. J.;
Hui Wang − Green Chemical Engineering Technology Maynard, S. A.; Bertazzo, S.; Stevens, M. M. Engineering Extracellular
Research Center, Shanghai Advanced Research Institute, Vesicles with the Tools of Enzyme Prodrug Therapy. Adv. Mater.
Chinese Academy of Sciences, Shanghai 201210, China 2018, 30, No. 1706616.
(16) Cabral, J.; Ryan, A. E.; Griffin, M. D.; Ritter, T. Extracellular
Complete contact information is available at: Vesicles as Modulators of Wound Healing. Adv. Drug Delivery Rev.
https://pubs.acs.org/10.1021/acsnano.0c07714 2018, 129, 394−406.
(17) Nanbo, A.; Katano, H.; Kataoka, M.; Hoshina, S.; Sekizuka, T.;
Author Contributions Kuroda, M.; Ohba, Y. Infection of Epstein(−)Barr Virus in Type III
#
These authors contributed equally to the work. Latency Modulates Biogenesis of Exosomes and the Expression

6365 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

Profile of Exosomal MiRNAs in the Burkitt Lymphoma Mutu Cell (34) Seeger, M. A.; Paller, A. S. The Roles of Growth Factors in
Lines. Cancers 2018, 10, 237. Keratinocyte Migration. Adv. Wound Care (New Rochelle) 2015, 4,
(18) Wang, C.; Wang, M.; Xu, T.; Zhang, X.; Lin, C.; Gao, W.; Xu, 213−224.
H.; Lei, B.; Mao, C. Engineering Bioactive Self-Healing Antibacterial (35) Rajkumar, R.; Konishi, K.; Richards, T. J.; Ishizawar, D. C.;
Exosomes Hydrogel for Promoting Chronic Diabetic Wound Healing Wiechert, A. C.; Kaminski, N.; Ahmad, F. Genomewide Rna
and Complete Skin Regeneration. Theranostics 2019, 9, 65−76. Expression Profiling in Lung Identifies Distinct Signatures in
(19) Song, Y.; Li, Z.; He, T.; Qu, M.; Jiang, L.; Li, W.; Shi, X.; Pan, Idiopathic Pulmonary Arterial Hypertension and Secondary Pulmo-
J.; Zhang, L.; Wang, Y.; Zhang, Z.; Tang, Y.; Yang, G. Y. M2Microglia- nary Hypertension. Am. J. Physiol. Heart Circ. Physiol. 2010, 298,
Derived Exosomes Protect the Mouse Brain from Ischemia- H1235−48.
Reperfusion Injury via Exosomal MiR-124. Theranostics 2019, 9, (36) Patel, A. N.; Selzman, C. H.; Kumpati, G. S.; McKellar, S. H.;
2910−2923. Bull, D. A. Evaluation of Autologous Platelet Rich Plasma for Cardiac
(20) Lv, Q.; Deng, J.; Chen, Y.; Wang, Y.; Liu, B.; Liu, J. Engineered Surgery: Outcome Analysis of 2000 Patients. J. Cardiothorac. Surg.
Human Adipose Stem-Cell-Derived Exosomes Loaded with MiR-21- 2016, 11, 62.
5p to Promote Diabetic Cutaneous Wound Healing. Mol. (37) Zeng, Z.; Huang, W. D.; Gao, Q.; Su, M. L.; Yang, Y. F.; Liu, Z.
Pharmaceutics 2020, 17, 1723−1733. C.; Zhu, B. H. Arnebin-1 Promotes Angiogenesis by Inducing eNOS,
(21) Ding, J.; Wang, X.; Chen, B.; Zhang, J.; Xu, J. Exosomes VEGF and HIF-1alpha Expression through the PI3K-Dependent
Derived from Human Bone Marrow Mesenchymal Stem Cells Pathway. Int. J. Mol. Med. 2015, 36, 685−97.
Stimulated by Deferoxamine Accelerate Cutaneous Wound Healing (38) Yum, M. J.; Koppula, S.; Kim, J. S.; Shin, G. M.; Chae, Y. J.;
by Promoting Angiogenesis. Biomed. Res. Int. 2019, 2019, 9742765. Yoon, T.; Chun, C. S.; Lee, J. D.; Song, M. Protective Effects of
(22) Hao, S.; Wang, L.; Zhao, K.; Zhu, X.; Ye, F. Rs1894720 Ampelopsis Brevipedunculata against in Vitro Hepatic Stellate Cells
Polymorphism in MIAT Increased Susceptibility to Age-Related System and Thioacetamide-Induced Liver Fibrosis Rat Model. Pharm.
Hearing Loss by Modulating the Activation of MiR-29b-Sirt1/PGC- Biol. 2017, 55, 1577−1585.
1alpha Signaling. J. Cell. Biochem. 2019, 120, 4975−4986. (39) Muthumalage, T.; Lamb, T.; Friedman, M. R.; Rahman, I. E-
(23) Su, T.; Xiao, Y.; Xiao, Y.; Guo, Q.; Li, C.; Huang, Y.; Deng, Q.; Cigarette Flavored Pods Induce Inflammation, Epithelial Barrier
Wen, J.; Zhou, F.; Luo, X. H. Bone Marrow Mesenchymal Stem Cells- Dysfunction, and DNA Damage in Lung Epithelial Cells and
Derived Exosomal MiR-29b-3p Regulates Aging-Associated Insulin Monocytes. Sci. Rep. 2019, 9, 19035.
Resistance. ACS Nano 2019, 13, 2450−2462. (40) Korntner, S.; Lehner, C.; Gehwolf, R.; Wagner, A.; Grutz, M.;
(24) Xu, B. H.; Sheng, J.; You, Y. K.; Huang, X. R.; Ma, R. C. W.; Kunkel, N.; Tempfer, H.; Traweger, A. Limiting Angiogenesis to
Wang, Q.; Lan, H. Y. Deletion of SMAD3 Prevents Renal Fibrosis and Modulate Scar Formation. Adv. Drug Delivery Rev. 2019, 146, 170−
Inflammation in Type 2 Diabetic Nephropathy. Metab., Clin. Exp. 189.
2020, 103, 154013. (41) Ruvinov, E.; Cohen, S. Alginate Biomaterial for the Treatment
(25) Jafarinejad-Farsangi, S.; Gharibdoost, F.; Farazmand, A.; of Myocardial Infarction: Progress, Translational Strategies, and
Kavosi, H.; Jamshidi, A.; Karimizadeh, E.; Noorbakhsh, F.; Clinical Outlook: From Ocean Algae to Patient Bedside. Adv. Drug
Mahmoudi, M. MicroRNA-21 and MicroRNA-29a Modulate the Delivery Rev. 2016, 96, 54−76.
Expression of Collagen in Dermal Fibroblasts of Patients with (42) Matricardi, P.; Di Meo, C.; Coviello, T.; Hennink, W. E.;
Systemic Sclerosis. Autoimmunity 2019, 52, 108−116. Alhaique, F. Interpenetrating Polymer Networks Polysaccharide
(26) Guo, J.; Lin, Q.; Shao, Y.; Rong, L.; Zhang, D. MiR-29b Hydrogels for Drug Delivery and Tissue Engineering. Adv. Drug
Promotes Skin Wound Healing and Reduces Excessive Scar Delivery Rev. 2013, 65, 1172−87.
Formation by Inhibition of the TGF-Beta1/SMAD/CTGF Signaling (43) Branco da Cunha, C.; Klumpers, D. D.; Li, W. A.; Koshy, S. T.;
Pathway. Can. J. Physiol. Pharmacol. 2017, 95, 437−442. Weaver, J. C.; Chaudhuri, O.; Granja, P. L.; Mooney, D. J. Influence
(27) Zhu, Y.; Li, Z.; Wang, Y.; Li, L.; Wang, D.; Zhang, W.; Liu, L.; of the Stiffness of Three-Dimensional Alginate/Collagen-I Inter-
Jiang, H.; Yang, J.; Cheng, J. Overexpression of MiR-29b Reduces penetrating Networks on Fibroblast Biology. Biomaterials 2014, 35,
Collagen Biosynthesis by Inhibiting Heat Shock Protein 47 during 8927−36.
Skin Wound Healing. Transl. Res. 2016, 178, 38−53. (44) Zeng, Q.; Chen, W. The Functional Behavior of a Macrophage/
(28) Yu, J.; Luo, H.; Li, N.; Duan, X. Suppression of Type I Collagen Fibroblast Co-Culture Model Derived from Normal and Diabetic
Expression by MiR-29b via PI3K, AKT, and SP1 Pathway, Part II: An Mice with a Marine Gelatin-Oxidized Alginate Hydrogel. Biomaterials
in Vivo Investigation. Invest. Ophthalmol. Visual Sci. 2015, 56, 6019− 2010, 31, 5772−81.
28. (45) Xu, G.; Cheng, L.; Zhang, Q.; Sun, Y.; Chen, C.; Xu, H.; Chai,
(29) Cheng, J.; Yu, H.; Deng, S.; Shen, G. MicroRNA Profiling in Y.; Lang, M. In Situ Thiolated Alginate Hydrogel: Instant Formation
Mid- and Late-Gestational Fetal Skin: Implication for Scarless Wound and Its Application in Hemostasis. J. Biomater. Appl. 2016, 31, 721−
Healing. Tohoku J. Exp. Med. 2010, 221, 203−9. 729.
(30) Nosenko, M. A.; Moysenovich, A. M.; Zvartsev, R. V.; (46) Brazil, J. C.; Quiros, M.; Nusrat, A.; Parkos, C. A. Innate
Arkhipova, A. Y.; Zhdanova, A. S.; Agapov, I. I.; Vasilieva, T. V.; Immune Cell-Epithelial Crosstalk during Wound Repair. J. Clin. Invest.
Bogush, V. G.; Debabov, V. G.; Nedospasov, S. A.; Moisenovich, M. 2019, 129, 2983−2993.
M.; Drutskaya, M. S. Novel Biodegradable Polymeric Microparticles (47) Deniz, A. A. H.; Abdik, E. A.; Abdik, H.; Aydin, S.; Sahin, F.;
Facilitate Scarless Wound Healing by Promoting Re-Epithelialization Tasli, P. N. Zooming in across the Skin: A Macro-to-Molecular
and Inhibiting Fibrosis. Front. Immunol. 2018, 9, 2851. Panorama. Adv. Exp. Med. Biol. 2019, 1247, 157−200.
(31) Wang, L.; Yang, J.; Ran, B.; Yang, X.; Zheng, W.; Long, Y.; (48) Nikoloudaki, G.; Brooks, S.; Peidl, A. P.; Tinney, D.; Hamilton,
Jiang, X. Small Molecular TGF-Beta1-Inhibitor-Loaded Electrospun D. W. JNK Signaling as a Key Modulator of Soft Connective Tissue
Fibrous Scaffolds for Preventing Hypertrophic Scars. ACS Appl. Physiology, Pathology, and Healing. Int. J. Mol. Sci. 2020, 21, 1015.
Mater. Interfaces 2017, 9, 32545−32553. (49) Chen, B.; Li, Q.; Zhao, B.; Wang, Y. Stem Cell-Derived
(32) Chin, J. S.; Madden, L.; Chew, S. Y.; Becker, D. L. Drug Extracellular Vesicles as a Novel Potential Therapeutic Tool for
Therapies and Delivery Mechanisms to Treat Perturbed Skin Wound Tissue Repair. Stem Cells Transl. Med. 2017, 6, 1753−1758.
Healing. Adv. Drug Delivery Rev. 2019, 149−150, 2−18. (50) Chen, M.; Xu, R.; Ji, H.; Greening, D. W.; Rai, A.; Izumikawa,
(33) Noh, S. M.; Abdul Kadir, S. H.; Crowston, J. G.; Subrayan, V.; K.; Ishikawa, H.; Takahashi, N.; Simpson, R. J. Transcriptome and
Vasudevan, S. Effects of Ranibizumab on TGF-Beta1 and TGF-Beta2 Long Noncoding Rna Sequencing of Three Extracellular Vesicle
Production by Human Tenon’s Fibroblasts: An in Vitro Study. Mol. Subtypes Released from the Human Colon Cancer Lim1863 Cell
Vis. 2015, 21, 1191−1200. Line. Sci. Rep. 2016, 6, 38397.

6366 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

(51) O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L. C.; (67) Sawada, S. I.; Sato, Y. T.; Kawasaki, R.; Yasuoka, J. I.; Mizuta,
Breakefield, X. O. RNA Delivery by Extracellular Vesicles in R.; Sasaki, Y.; Akiyoshi, K. Nanogel Hybrid Assembly for Exosome
Mammalian Cells and Its Applications. Nat. Rev. Mol. Cell Biol. Intracellular Delivery: Effects on Endocytosis and Fusion by Exosome
2020, 21, 585−606. Surface Polymer Engineering. Biomater. Sci. 2020, 8, 619−630.
(52) Zhang, Y.; Hao, Z.; Wang, P.; Xia, Y.; Wu, J.; Xia, D.; Fang, S.; (68) Tao, S. C.; Yuan, T.; Zhang, Y. L.; Yin, W. J.; Guo, S. C.;
Xu, S. Exosomes from Human Umbilical Cord Mesenchymal Stem Zhang, C. Q. Exosomes Derived from MiR-140−5p-Overexpressing
Cells Enhance Fracture Healing through HIF-1alpha-Mediated Human Synovial Mesenchymal Stem Cells Enhance Cartilage Tissue
Promotion of Angiogenesis in a Rat Model of Stabilized Fracture. Regeneration and Prevent Osteoarthritis of the Knee in a Rat Model.
Cell Proliferation 2019, 52, No. e12570. Theranostics 2017, 7, 180−195.
(53) Zhu, Y.; Jia, Y.; Wang, Y.; Xu, J.; Chai, Y. Impaired Bone (69) Squadrito, M. L.; Baer, C.; Burdet, F.; Maderna, C.; Gilfillan, G.
Regenerative Effect of Exosomes Derived from Bone Marrow D.; Lyle, R.; Ibberson, M.; De Palma, M. Endogenous RNAs
Mesenchymal Stem Cells in Type 1 Diabetes. Stem Cells Transl. Modulate MicroRNA Sorting to Exosomes and Transfer to Acceptor
Med. 2019, 8, 593−605. Cells. Cell Rep. 2014, 8, 1432−46.
(54) Casado-Diaz, A.; Quesada-Gomez, J. M.; Dorado, G. (70) Fulzele, S.; Mendhe, B.; Khayrullin, A.; Johnson, M.; Kaiser, H.;
Extracellular Vesicles Derived from Mesenchymal Stem Cells Liu, Y.; Isales, C. M.; Hamrick, M. W. Muscle-Derived MiR-34a
(MSC) in Regenerative Medicine: Applications in Skin Wound Increases with Age in Circulating Extracellular Vesicles and Induces
Healing. Front. Bioeng. Biotechnol. 2020, 8, 146. Senescence of Bone Marrow Stem Cells. Aging 2019, 11, 1791−1803.
(55) Wang, S.; Li, C.; Yu, Y.; Qiao, J. Decreased Expression of Micro (71) Wang, Z.; Yan, K.; Ge, G.; Zhang, D.; Bai, J.; Guo, X.; Zhou, J.;
RNA-145 Promotes the Biological Functions of Fibroblasts in Xu, T.; Xu, M.; Long, X.; Hao, Y.; Geng, D. Exosomes Derived from
Hypertrophic Scar Tissues by Upregulating the Expression of MiR-155−5p-Overexpressing Synovial Mesenchymal Stem Cells
Transcription Factor SOX-9. Exp. Ther. Med. 2019, 18, 3450−3460. Prevent Osteoarthritis via Enhancing Proliferation and Migration,
(56) Piffoux, M.; Nicolas-Boluda, A.; Mulens-Arias, V.; Richard, S.; Attenuating Apoptosis, and Modulating Extracellular Matrix Secretion
Rahmi, G.; Gazeau, F.; Wilhelm, C.; Silva, A. K. A. Extracellular in Chondrocytes. Cell Biol. Toxicol. 2021, 37, 85−96.
Vesicles for Personalized Medicine: The Input of Physically Triggered (72) Chen, Q.; Zhao, T.; Xie, X.; Yu, D.; Wu, L.; Yu, W.; Sun, W.
Production, Loading and Theranostic Properties. Adv. Drug Delivery MicroRNA-663 Regulates the Proliferation of Fibroblasts in Hyper-
Rev. 2019, 138, 247−258. trophic Scars via Transforming Growth Factor-Beta1. Exp. Ther. Med.
(57) Song, X.; You, W.; Zhu, J.; Cui, X.; Hu, J.; Chen, Y.; Liu, W.; 2018, 16, 1311−1317.
Wang, L.; Li, S.; Wei, Y.; Yang, L.; Li, F. A Genetic Variant in (73) Yu, H. N.; Li, X. M.; Kong, L. L.; Ren, J.; Wu, H.; Bu, L. G.;
MiRNA-219−1 Is Associated with Risk of Esophageal Squamous Cell Ding, N. Z.; Ni, H. Connective Tissue Growth Factor Gene
Carcinoma in Chinese Kazakhs. Dis. Markers 2015, 2015, 541531. Expression in Goat Endometrium during Estrous Cycle and Early
(58) Liu, T.; Wu, Y.; Huang, T.; Zhang, X.; Cai, Y. MiR-590 Pregnancy. Theriogenology 2020, 153, 85−90.
(74) Zhang, J.; Nie, D.; Williamson, K.; Rocha, J. L.; Hogan, M. V.;
Promotes the Proliferation of Humscs and Induces ECM Synthesis by
Wang, J. H. Selectively Activated PRP Exerts Differential Effects on
Targeting SMAD7. Oncol. Lett. 2017, 14, 3941−3946.
Tendon Stem/Progenitor Cells and Tendon Healing. J. Tissue Eng.
(59) Nie, Z. C.; Weng, W. H.; Shang, Y. S.; Long, Y.; Li, J.; Xu, Y. T.;
2019, 10, 2041731418820034.
Li, Z. MicroRNA-126 Is Down-Regulated in Human Esophageal
(75) van der Veer, W. M.; Bloemen, M. C.; Ulrich, M. M.; Molema,
Squamous Cell Carcinoma and Inhibits the Proliferation and
G.; van Zuijlen, P. P.; Middelkoop, E.; Niessen, F. B. Potential
Migration in EC109 Cell via PI3K/AKT Signaling Pathway. Int. J.
Cellular and Molecular Causes of Hypertrophic Scar Formation.
Clin. Exp. Pathol. 2015, 8, 4745−4754.
Burns 2009, 35, 15−29.
(60) Liu, H. J.; Lam, H. C.; Baglini, C. V.; Nijmeh, J.; Cottrill, A. A.;
(76) Bielli, A.; Scioli, M. G.; Mazzaglia, D.; Doldo, E.; Orlandi, A.
Chan, S. Y.; Henske, E. P. Rapamycin-Upregulated MiR-29b Antioxidants and Vascular Health. Life Sci. 2015, 143, 209−16.
Promotes Mtorc1-Hyperactive Cell Growth in TSC2-Deficient Cells (77) Hao, T.; Li, J.; Yao, F.; Dong, D.; Wang, Y.; Yang, B.; Wang, C.
by Downregulating Tumor Suppressor Retinoic Acid Receptor Beta Injectable Fullerenol/Alginate Hydrogel for Suppression of Oxidative
(Rarbeta). Oncogene 2019, 38, 7367−7383. Stress Damage in Brown Adipose-Derived Stem Cells and Cardiac
(61) Sur, S.; Steele, R.; Shi, X.; Ray, R. B. MiRNA-29b Inhibits Repair. ACS Nano 2017, 11, 5474−5488.
Prostate Tumor Growth and Induces Apoptosis by Increasing Bim (78) Kelishomi, Z. H.; Goliaei, B.; Mahdavi, H.; Nikoofar, A.;
Expression. Cells 2019, 8, 1455. Rahimi, M.; Moosavi-Movahedi, A. A.; Mamashli, F.; Bigdeli, B.
(62) Gallant-Behm, C. L.; Piper, J.; Lynch, J. M.; Seto, A. G.; Hong, Antioxidant Activity of Low Molecular Weight Alginate Produced by
S. J.; Mustoe, T. A.; Maari, C.; Pestano, L. A.; Dalby, C. M.; Jackson, Thermal Treatment. Food Chem. 2016, 196, 897−902.
A. L.; Rubin, P.; Marshall, W. S. A MicroRNA-29 Mimic (Remlarsen) (79) Davison-Kotler, E.; Marshall, W. S.; Garcia-Gareta, E. Sources
Represses Extracellular Matrix Expression and Fibroplasia in the Skin. of Collagen for Biomaterials in Skin Wound Healing. Bioengineering
J. Invest. Dermatol. 2019, 139, 1073−1081. 2019, 6, 56.
(63) Peng, Y.; Wang, X.; Guo, Y.; Peng, F.; Zheng, N.; He, B.; Ge, (80) Karayi, A. K.; Basavaraj, V.; Narahari, S. R.; Aggithaya, M. G.;
H.; Tao, L.; Wang, Q. Pattern of Cell-to-Cell Transfer of MicroRNA Ryan, T. J.; Pilankatta, R. Human Skin Fibrosis: Up-Regulation of
by Gap Junction and Its Effect on the Proliferation of Glioma Cells. Collagen Type III Gene Transcription in the Fibrotic Skin Nodules of
Cancer Sci. 2019, 110, 1947−1958. Lower Limb Lymphoedema. Trop. Med. Int. Health 2020, 25, 319−
(64) Syn, N. L.; Wang, L.; Chow, E. K.; Lim, C. T.; Goh, B. C. 327.
Exosomes in Cancer Nanomedicine and Immunotherapy: Prospects (81) Cakin, M. C.; Ozdemir, B.; Kaya-Dagistanli, F.; Arkan, H.;
and Challenges. Trends Biotechnol. 2017, 35, 665−676. Bahtiyar, N.; Anapali, M.; Akbas, F.; Onaran, I. Evaluation of the in
(65) Won Lee, G.; Thangavelu, M.; Joung Choi, M.; Yeong Shin, E.; Vivo Wound Healing Potential of the Lipid Fraction from Activated
Sol Kim, H.; Seon Baek, J.; Woon Jeong, Y.; Eun Song, J.; Platelet-Rich Plasma. Platelets 2020, 31, 513−520.
Carlomagno, C.; Miguel Oliveira, J.; Luis Reis, R.; Khang, G. (82) Liu, W.; Wang, M.; Cheng, W.; Niu, W.; Chen, M.; Luo, M.;
Exosome Mediated Transfer of MiRNA-140 Promotes Enhanced Xie, C.; Leng, T.; Zhang, L.; Lei, B. Bioactive Antiinflammatory
Chondrogenic Differentiation of Bone Marrow Stem Cells for Antibacterial Hemostatic Citrate-Based Dressing with Macrophage
Enhanced Cartilage Repair and Regeneration. J. Cell. Biochem. 2020, Polarization Regulation for Accelerating Wound Healing and Hair
121, 3642−3652. Follicle Neogenesis. Bioact. Mater. 2021, 6, 721−728.
(66) Kluszczynska, K.; Czernek, L.; Cypryk, W.; Peczek, L.; Duchler, (83) Chen, J.; Zhou, R.; Liang, Y.; Fu, X.; Wang, D.; Wang, C.
M. Methods for the Determination of the Purity of Exosomes. Curr. Blockade of LncRNA-ASLNC5088-Enriched Exosome Generation in
Pharm. Des. 2020, 25, 4464−4485. M2Macrophages by GW4869 Dampens the Effect of M2Macrophages

6367 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368
ACS Nano www.acsnano.org Article

on Orchestrating Fibroblast Activation. FASEB J. 2019, 33, 12200−


12212.
(84) Smith, A.; Watkins, T.; Theocharidis, G.; Lang, I.; Leschinsky,
M.; Maione, A.; Kashpur, O.; Raimondo, T.; Rahmani, S.; Baskin, J.;
Mooney, D.; Veves, A.; Garlick, J. A Novel Three-Dimensional Skin
Disease Model to Assess Macrophage Function in Diabetes. Tissue
Eng., Part C 2021, 27, 49.
(85) MacLeod, A. S.; Mansbridge, J. N. The Innate Immune System
in Acute and Chronic Wounds. Adv. Wound Care (New Rochelle)
2016, 5, 65−78.
(86) Golberg, A.; Villiger, M.; Khan, S.; Quinn, K. P.; Lo, W. C. Y.;
Bouma, B. E.; Mihm, M. C., Jr.; Austen, W. G., Jr.; Yarmush, M. L.
Preventing Scars after Injury with Partial Irreversible Electroporation.
J. Invest. Dermatol. 2016, 136 (11), 2297−2304.
(87) Ramos, M. L.; Gragnani, A.; Ferreira, L. M. Is There an Ideal
Animal Model to Study Hypertrophic Scarring? J. Burn Care Res.
2008, 29 (2), 363−8.
(88) Zhou, S.; Wang, W.; Zhou, S.; Zhang, G.; He, J.; Li, Q. A Novel
Model for Cutaneous Wound Healing and Scarring in the Rat. Plast.
Reconstr. Surg. 2019, 143 (2), 468−477.
(89) Liu, S.; Jiang, L.; Li, H.; Shi, H.; Luo, H.; Zhang, Y.; Yu, C.; Jin,
Y. Mesenchymal Stem Cells Prevent Hypertrophic Scar Formation via
Inflammatory Regulation When Undergoing Apoptosis. J. Invest.
Dermatol. 2014, 134 (10), 2648−2657.
(90) Shen, Y. F.; Huang, J. H.; Wang, K. Y.; Zheng, J.; Cai, L.; Gao,
H.; Li, X. L.; Li, J. F. Pth Derivative Promotes Wound Healing via
Synergistic Multicellular Stimulating and Exosomal Activities. Cell
Commun. Signaling 2020, 18 (1), 40.
(91) Tao, S. C.; Guo, S. C.; Li, M.; Ke, Q. F.; Guo, Y. P.; Zhang, C.
Q. Chitosan Wound Dressings Incorporating Exosomes Derived from
MicroRNA-126-Overexpressing Synovium Mesenchymal Stem Cells
Provide Sustained Release of Exosomes and Heal Full-Thickness Skin
Defects in a Diabetic Rat Model. Stem Cells Transl. Med. 2017, 6 (3),
736−747.

6368 https://dx.doi.org/10.1021/acsnano.0c07714
ACS Nano 2021, 15, 6352−6368

You might also like