ArticlePDF AvailableLiterature Review

Engineering exosomes for bone defect repair

Frontiers
Frontiers in Bioengineering and Biotechnology
Authors:

Abstract and Figures

Currently, bone defect repair is still an intractable clinical problem. Numerous treatments have been performed, but their clinical results are unsatisfactory. As a key element of cell-free therapy, exosome is becoming a promising tool of bone regeneration in recent decades, because of its promoting osteogenesis and osteogenic differentiation function in vivo and in vitro. However, low yield, weak activity, inefficient targeting ability, and unpredictable side effects of natural exosomes have limited the clinical application. To overcome the weakness, various approaches have been applied to produce engineering exosomes by regulating their production and function at present. In this review, we will focus on the engineering exosomes for bone defect repair. By summarizing the exosomal cargos affecting osteogenesis, the strategies of engineering exosomes and properties of exosome-integrated biomaterials, this work will provide novel insights into exploring advanced engineering exosome-based cell-free therapy for bone defect repair.
Content may be subject to copyright.
Engineering exosomes for bone
defect repair
Shaoyang Ma, Yuchen Zhang, Sijia Li, Ang Li, Ye Li* and
Dandan Pei*
Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of
Stomatology, Xian Jiaotong University, Xian, Shaanxi, China
Currently, bone defect repair is still an intractable clinical problem. Numerous
treatments have been performed, but their clinical results are unsatisfactory. As
a key element of cell-free therapy, exosome is becoming a promising tool of
bone regeneration in recent decades, because of its promoting osteogenesis
and osteogenic differentiation function in vivo and in vitro. However, low yield,
weak activity, inefcient targeting ability, and unpredictable side effects of
natural exosomes have limited the clinical application. To overcome the
weakness, various approaches have been applied to produce engineering
exosomes by regulating their production and function at present. In this
review, we will focus on the engineering exosomes for bone defect repair.
By summarizing the exosomal cargos affecting osteogenesis, the strategies of
engineering exosomes and properties of exosome-integrated biomaterials, this
work will provide novel insights into exploring advanced engineering exosome-
based cell-free therapy for bone defect repair.
KEYWORDS
bone regeneration, engineering exosomes, exosomal cargos, exosome-integrated
biomaterials, osteogenesis
1 Introduction
Bone is the central element in skeletal tissues of the human body, and provides a
framework for attachment of muscles and other tissues, enables body movements, provides
protection of internal organs from injury, promotes blood cells production, and balances
calcium and acid/base homeostasis (Elefteriou, 2018). However, the regeneration of critical-
size bone defects is still a major clinical challenge and globally costs up to $45 billion per year
(Mauffrey et al., 2015;Bharadwaz and Jayasuriya, 2020). Recently, stem cell therapy is
considered as a potential strategy for bone defect regeneration (TanSHS.etal.,2020), and
several clinical studies have demonstrated mesenchymal stromal/stem cells (MSCs) to be safe
and efcacious for the treatment of bone defects and diseases (Liebergall et al., 2013;Chen
et al., 2016;Castillo-Cardiel et al., 2017;Hernigou et al., 2018a;Hernigou et al., 2018b).
Nevertheless, cellular therapies incur signicant costs and challenges as they require
stringently monitored manufacturing, handling, and storage to ensure optimal viability
and potency of cells needed for transplantation (Tan et al., 2021). More importantly,
accumulating evidence indicates that the positive effect of MSCs on tissue repair is to
stimulate the activity of tissue-resident recipient cells through paracrine, such as exosomes,
OPEN ACCESS
EDITED BY
Jianxun Ding,
Changchun Institute of Applied
Chemistry (CAS), China
REVIEWED BY
Shiyu Liu,
Fourth Military Medical University, China
Qianli Ma,
University of Oslo, Norway
Mingming Yang,
Northwest A&F University, China
*CORRESPONDENCE
Ye Li,
lisa.l0309@163.com
Dandan Pei,
peidandan@xjtu.edu.cn
SPECIALTY SECTION
This article was submitted
to Biomaterials,
a section of the journal
Frontiers in Bioengineering and
Biotechnology
RECEIVED 07 November 2022
ACCEPTED 28 November 2022
PUBLISHED 07 December 2022
CITATION
Ma S, Zhang Y, Li S, Li A, Li Y and Pei D
(2022), Engineering exosomes for bone
defect repair.
Front. Bioeng. Biotechnol. 10:1091360.
doi: 10.3389/fbioe.2022.1091360
COPYRIGHT
© 2022 Ma, Zhang, Li, Li, Li and Pei. This
is an open-access article distributed
under the terms of the Creative
Commons Attribution License (CC BY).
The use, distribution or reproduction in
other forums is permitted, provided the
original author(s) and the copyright
owner(s) are credited and that the
original publication in this journal is
cited, in accordance with accepted
academic practice. No use, distribution
or reproduction is permitted which does
not comply with these terms.
Frontiers in Bioengineering and Biotechnology frontiersin.org01
TYPE Review
PUBLISHED 07 December 2022
DOI 10.3389/fbioe.2022.1091360
rather than directly differentiate into parenchymal cells to repair or
replace damaged tissue (Liang et al., 2014;Zhang et al., 2016). Such
concerns have driven the search for alternate therapeutic strategies
and cell-free therapies based on exosomes have become strongly
established in the landscape of regenerative medicine.
Exosome is a subclass of membrane-coated extracellular
vesicles with sizes of 30150 nm (Tkach and Thery, 2016). As
one of the most revolutionary contributions to cell biology in the
past 30 years (Wang Y. et al., 2019), exosomes can exert multiple
biological functions by targeting recipient cells and inducing
signaling via receptor-ligand interactions, endocytosis and/or
phagocytosis (Bobrie et al., 2012;Colombo et al., 2014;
Hoshino et al., 2015;Shang et al., 2021). Exosomes have been
experimented with many animal models for the regeneration of
bone, osteochondral, and cartilage injury/diseases such as
osteoarthritis (OA), osteoporosis, osteonecrosis, and
inammatory bone loss in periodontitis with enhanced tissue
formation and integration (Kuang et al., 2019;Kim et al., 2020;
Lei et al., 2022). Furthermore, several exosome-based clinical
experiments of orthopedic diseases have been performed based
on US-NIH clinical trial database (https://clinicaltrials.gov/).
However, there still are several constraints to exosome clinical
applications for bone defect repair: 1) unclear mechanism of
promoting bone tissue regeneration; 2) poor retention and
targeting ability of exosome at the bone defect site; 3) low
extraction rate and complex separation process.
In view of the shortcomings of natural exosomes, a growing
number of studies are aiming to develop engineering exosomes
based on modifying exosomal cargos or/and incorporating
biomaterials (Bei et al., 2021;Lathwal et al., 2021;Liang et al.,
2021). Here we will review the recent research of engineering
exosome used in bone defect repair, and highlight the bioactive
cargos and construction strategies. Additionally, we will also
summarize the application of biomaterials to impregnate
exosome and focus on how the properties of biomaterials
assist exosome to promote bone regeneration. By reviewing
currently available knowledge, this present review will
contribute to the clinical knowledge and may have
implications for the engineering design of exosomes used in
bone defect repair.
2 Osteogenic cargos in exosomes
In the past decade, numerous exosomal bioactive cargos have
been revealed (Kalluri and LeBleu, 2020). Exosomal cargos are
dependent on the parent cell type and vary between different
physiological or pathological conditions (Meng et al., 2019). The
vesicular structure of exosome provides an enclosed space to
protect exosomal cargos against degradation. In return, exosomal
cargos are the foundation to endow exosomes with various
biological functions. In this section we will review recent
research about exosomes in bone regeneration and focus on
the functions of exosomal cargos and their molecular
mechanisms (Figure 1).
2.1 Non-Coding RNA
Non-coding RNAs (ncRNAs) refer to the RNAs that lack
protein-coding regions, and have the potential to regulate gene
expression at transcriptional, post-transcriptional, and
translational levels, thereby modulating associated signaling
networks (Bhat et al., 2020). NcRNAs have become a hot
topic of increasing concern after the completion of the
Human Genome Project (Lander et al., 2001), which showed
only 1.2% of genes in the genome could encode proteins, whereas
the rest were considered as non-coding. Accumulating
evidence demonstrates that a variety of ncRNAs can be
encapsulated and transported by exosomes, among which
exosomal microRNAs (miRNAs), long non-coding RNAs
(lncRNAs), and circular RNAs (circRNAs) are the most
attractive subclasses in the eld of bone regeneration.
2.1.1 miRNAs
MiRNAs are small, highly conserved ncRNAs with ~22 nt
length (Prattichizzo et al., 2021). The biogenesis of miRNAs
involves the processing of larger primary miRNAs (pri-miRNAs)
into shorter pre-miRNAs, and the maturation of pre-miRNA to
produce active miRNAs (Ha and Kim, 2014). MiRNAs mediate
post-transcriptional gene silencing by binding to the target
mRNAs 3-untranslated region (UTR) or open reading frames
(ORFs) to regulate the translational process in a wide range of
physiological processes (Yang et al., 2017).
Since the rst observation of exosomal miRNAs in 2007
(Valadi et al., 2007), miRNAs have become the most studied
cargos in exosome. Recently, a massive number of studies have
demonstrated that miRNAs in natural exosomes derived from
multiple cell types can promote osteogenesis (Table 1). These
studies conrmed miRNAs from exosomes of different cellular
origin can enter recipient cells with the help of exosome
internalization, and then regulate the expressions of genes
associated with osteogenic at the translational level to regulate
bone regeneration.
2.1.2 lncRNAs
As a heterogeneous group of non-protein-coding
transcripts with length of greater than 200 nucleotides,
lncRNAs are emerging regulators involved in diverse
physiological and pathological processes (Kopp and
Mendell, 2018;Nair et al., 2020). Notably, lncRNAs can be
selectively packaged into exosomes (Valadi et al., 2007), which
enable them as biomarkers of certain disease. For instance, the
expressions of lncRNAs in serum exosomes from persons with
or without osteoporosis showed signicant differences (Teng
et al., 2020).
Frontiers in Bioengineering and Biotechnology frontiersin.org02
Ma et al. 10.3389/fbioe.2022.1091360
Beyond as molecular markers, lncRNAs can sponge miRNAs
and regulate the expression of downstream genes, called
competing endogenous RNA (ceRNA) mechanism (Salmena
et al., 2011). Accumulating evidence showed that lncRNAs
from multiple cells-derived exosomes can enter the receptor
cells and have the potential to regulate bone regeneration
(Table 1).
2.1.3 circRNAs
CircRNA, a special subclass of lncRNAs with a circular
structure, has recently gained interest because of their
extraordinary stability, much longer half-life and diverse
biological functions (Jeck and Sharpless, 2014;Liu and Chen,
2022). CircRNAs can be selectively packaged into exosomes
similar to lncRNAs (Ma et al., 2021). Additionally, exosomal
circRNA also has the potential to regulate gene expression by
ceRNA mechanism (Zhi et al., 2021;Du et al., 2022). Number of
studies have revealed the regulatory function of exosomal
circRNA in bone regeneration (Table 1). Interestingly, the
effect of exosomal circRNAs in regulating bone regeneration,
it seems, is a double-edged sword. For example, Zhi et al. (2021)
reported that serum exosomal hsa_circ_0006859 was
upregulated in patients with osteoporosis, and suppressed
osteogenesis and promoted adipogenesis. Therefore, the
regulatory functions of exosomal circRNAs are still unclear,
which needs further studies.
2.1.4 tsRNAs
Transfer RNA (tRNA)-derived small RNA (tsRNA) is a class
of small ncRNAs generated from precursor or mature tRNAs,
which has recently received considerable attention (Zhu et al.,
2018;Zhu et al., 2019). With the deepening of research, tsRNAs
have been reported to regulate stem cell maintenance (Blanco
et al., 2016), cancer (Balatti et al., 2017), viral infection (Nunes
et al., 2020), neurological diseases (Zhang et al., 2020), epigenetic
inheritance (Zhang Y. et al., 2018), and symbiosis (Ren et al.,
2019). The mechanisms of action of tsRNAs include playing as
mimicry/replacement of tRNAs with sequence/structure effects,
associating with ribonucleoproteins and binding to the target
genes like miRNAs (Chen et al., 2021). Although the function of
exosomal tsRNAs is an emerging eld with a paucity of research,
Fang et al. (2020) explored the osteogenic effect of exosomal
tsRNA (Table 1). They found tsRNA-10277 in the exosome
derived from BMSCs could enhance osteogenic differentiation
ability of dexamethasone-induced BMSCs.
2.2 mRNAs
As the Central Dogma of molecular biology presented
mRNA as the fundamental ingredient in genetic translational
machinery (Crick, 1970), it seemed that transferring mRNA via
exosomes to affect the biological processes of recipient cells
would be a more simple and efcient method compared with
transferring ncRNAs. However, there has been remarkably little
work about exosomal mRNA. This is probably because miRNAs
and lncRNAs are the vast majority of exosomal RNAs
(Hergenreider et al., 2012;Zhang et al., 2015;Zhang et al.,
2017), and exosomal mRNAs were classically thought to be in
the form of fragments, but not their intact forms (Valadi et al.,
2007;Wei et al., 2017). With further research, it was estimated
that on average, one intact mRNA can be found within every
1,000 exosomes produced endogenously without external
stimulation (Yang Z. et al., 2020). Therefore, it is essential to
conrm the integrity, high expression and regulatory function of
mRNAs in the research based on exosomal mRNAs. In recent
FIGURE 1
Schematic of exosomal cargos (miRNA, lncRNA, circRNA, tsRNA, mRNA and protein) with the function of promoting bone regeneration.
Frontiers in Bioengineering and Biotechnology frontiersin.org03
Ma et al. 10.3389/fbioe.2022.1091360
TABLE 1 The exosomal cargos involved in bone regeneration.
Cargos Sources Target cell Function References
ncRNA
miRNA
miR-23a-3p UCMSCs Chondrocytes BMSCs Promoting the migration, proliferation and differentiation of
chondrocytes and BMSCs
Hu et al. (2020)
miR-21 UCMSCs EPCs Enhancing angiogenesis Zhang et al.
(2021c)
miR-378a M2 polarized
macrophages
MSCs Inducing osteogenic differentiation Kang et al. (2020)
miR-100-5p IPFP-MSCs Chondrocytes Enhancing the autophagy level of chondrocytes Wu et al. (2019)
miR-335 Mature DCs BMSCs Promoting the proliferation and osteogenic differentiation of BMSCs Cao et al. (2021)
miR-126 EPCs Endothelial cells Enhancing the proliferation, migration, and angiogenic capacity of
endothelial cells
Jia et al. (2019)
miR-451a ADSCs Macrophages Inhibiting inammation and promoting the polarization of
M1 macrophages to M2 macrophages
Li et al. (2022)
miR-1265p SCAP HUVECs Promoting angiogenesis Jing et al. (2022)
miR-1505p MC3T3-E1 Promoting osteogenesis
miR-29a BMSCs HUVECs Promoting angiogenesis Lu et al. (2020)
lncRNA
NEAT1 Prostate cancer cells BMSCs Inducing osteogenic differentiation Mo et al. (2021)
MALAT1 EPCs Bone marrow-derived
macrophages
Enhancing recruitment and differentiation of osteoclast precursors Cui et al. (2019)
MALAT1 BMSCs hFOB1.19 Enhancing osteoblast activity Yang et al. (2019)
MEG-3 BMSCs Chondrocytes Reducing senescence and apoptosis Jin et al. (2021)
LYRM4-AS1 BMSCs Chondrocytes Regulating the growth of chondrocytes Wang et al.
(2021c)
H19 BMSCs CD31
+
ECs and BMSCs Promoting endothelial angiogenesis and BMSCs osteogenesis Behera et al.
(2021)
BMSCs Affecting osteogenic differentiation Wang et al.
(2021d)
circRNA
circLPAR1 Osteogenic-induced
DPSCs
DPSCs Promoting osteogenic differentiation of the recipient DPSCs Xie et al. (2020)
circRNA_0001236 BMSCs BMSCs Promoting chondrogenic differentiation Mao et al. (2021)
circ_003564 BMSCs Primary neurons and PC-12
cells
Attenuating inammasome-related pyroptosis Zhao et al. (2022)
circ-Rtn4 BMSCs MC3T3-E1 cells Attenuating TNF-α-induced cytotoxicity and apoptosis Cao et al. (2020)
circ_0008542 MC3T3-E1 cells Osteoclast Promoting osteoclast differentiation and bone resorption Wang et al.
(2021b)
circRNA3503 SMSCs Chondrocytes Promoting chondrocyte renewal to alleviate the progressive loss of
chondrocytes
Tao et al. (2021)
cirHmbox1 Osteoclasts Osteoclasts and osteoblasts Regulating osteoclasts differentiation and osteoblasts differentiation Liu et al. (2020)
tsRNA
tsRNA-10277 BMSCs Dexamethasone-induced
BMSCs
Enhancing osteogenic differentiation ability Fang et al. (2020)
mRNA
TFAM SHED DPSCs Promoting osteogenic differentiation Guo et al. (2022)
IL-10 M2 polarized
macrophages
BMSCs Regulating cell differentiation and bone metabolism Chen et al. (2022)
Protein
CD73 MSCs Chondrocytes Suppressing inammation and restoring matrix homeostasis Zhang et al.
(2019)
Wnt-3a ADSCs Primary osteoblastic cells Promoting the proliferation and osteogenic differentiation Lu et al. (2017)
(Continued on following page)
Frontiers in Bioengineering and Biotechnology frontiersin.org04
Ma et al. 10.3389/fbioe.2022.1091360
research, the regulatory function of exosomal mRNA in bone
regeneration have been revealed (Table 1). These studies showed
exosomal mRNAs also could be a useful tool to aid the healing of
bone defects, as long as improving the loading efciency of
intrinsically encapsulate transcribed mRNA into secreted
exosomes.
2.3 Protein
A variety of proteins have been observed in exosomes,
including cytoskeletal proteins, tetraspanins (CD9, CD63,
CD81, and CD82), ESCRT-associated components (Alix and
TSG101), heat shock proteins (HSP60, HSP70, and HSP90),
antigen presentation proteins (MHC I and MHC II), and
integrins (Kalluri and LeBleu, 2020;Zhu et al., 2020). As the
main executor of life activities, proteins are not only the markers
of exosomes but also endow exosomes with many biofunctions
including regulating bone regeneration (Table 1).
Despite above research drawn inspiring conclusions, the
controversy about the function of exosomal protein in bone
regeneration persists. Take BMP2, an important regulator of
osteogenesis, as an example. Han L. et al. (2022) reported that
BMP2 in BMSC-derived exosomes could promote tendon bone
healing in rotator cuff tear by activating Smad/RUNX2 signaling
pathway. Conversely, in another study, exosomes derived from
MSCs overexpressing BMP2 did not contain BMP2 protein, and
the function of promoting bone regeneration was possibly due to
the changes of exosomal miRNA composition (Huang et al.,
2020). Additionally, Furuta et al. (2016) found MSC exosomes
could promote mice fracture healing, but the levels of SDF-1,
MCP-1, and MCP-3, essential factors in the initial phase of
fracture healing (Kitaori et al., 2009;Toupadakis et al., 2012;
Ando et al., 2014;Ishikawa et al., 2014), in MSC exosomes were
signicantly lower, suggesting that bone regeneration may be
mediated by other exosome components (such as miRNAs) but
not exosomal proteins. The controversy above suggests that the
mechanisms by which exosomal proteins work may be complex
and remain to be determined.
To sum up, the function of various exosomal cargos makes
exosomes have the ability to regulate bone regeneration in
different ways. Predictably, more and more exosomal cargos
would be revealed to function in bone regeneration by
conventional or novel mechanism in the near future.
Meanwhile, the explorations of mechanism inspired
investigators to design engineering exosomes for bone
regeneration by modifying the exosomal cargos, which will be
discussed in the next section.
3 Strategies of engineering exosomes
for bone defect repair
Although numerous exosomal cargos have been revealed to
function in promoting osteogenic differentiation in the past
decade, the clinical application of exosome in bone
regeneration is still facing major challenges. The reason may
be the low exosome yield, low content of functional exosomal
cargos and low targeting efciency of native exosomes (Song
et al., 2022).
To improve the yield of exosomes, it is necessary to simplify the
exosome extraction procedure. Until now, six classes of exosome
separation strategies have been reported, including ultra-speed
centrifugation, ultraltration, immunoafnity capture, charge
neutralization-based polymer precipitation, size-exclusion
chromatograph, and microuidic techniques, with unique sets of
advantages and disadvantages for each technique (Yang D. et al.,
2020). These rapid development in separation technology has in a
large extent solved the problem of exosome isolation.
In order to enrich the exosomal cargo and increase exosome
targeting efciency, engineering exosome is rapidly expanding in the
past decade. Engineering exosomes are the exosomes created
through changing parent cells or directly on exosomes by
biochemical or physical treatment (Kojima et al., 2018;Yerneni
et al., 2019). In this section, we summarized the three strategies of
engineering exosomes for bone regeneration (Figure 2): 1) direct
modication of exosomes, 2) chemical or physical treatment of
parent cells, and 3) genetic modication of parent cells.
3.1 Direct modication of exosomes
The direct modication of exosomes means decorating the
surface proteins to improve the targeting ability of exosomes; or
TABLE 1 (Continued) The exosomal cargos involved in bone regeneration.
Cargos Sources Target cell Function References
Mutant HIF-1αBMSCs BMSCs Promoting osteogenic differentiation capacity and angiogenesis Li et al. (2017)
HUVECs
BMP2 BMSCs BMSCs Promoting tendon bone healing in rotator cuff tear Han et al. (2022a)
UCMSCs, umbilical cord-derived mesenchymal stem cells; IPFP-MSCs, infrapatellar fat pad mesenchymal stem cells; DCs, dendritic cells; EPCs, endothelial progenitor cells; ADSCs,
adipose-derived stem cells; SCAP, stem cells from apical papilla; BMSCs, bone marrow mesenchymal stem cells; ECs, endothelial cells; DPSCs, dental pulp stem cells; SMSCs, synovium
mesenchymal stem cells; SHED, stem cells from human exfoliated deciduous teeth; DPSCs, dental pulp stem cells; MSCs, mesenchymal stem cells; HUVECs, human umbilical vein
endothelial cells.
Frontiers in Bioengineering and Biotechnology frontiersin.org05
Ma et al. 10.3389/fbioe.2022.1091360
embellishing exosomal cargos or exogenous bioactive molecules
to enhance the regulatory function through chemical methods
(conjugation of peptides to exosomal surface (Gao X. et al.,
2018)) or physical methods (electroporation (Tian et al., 2014)
or sonication (Wang P. et al., 2019)) directly. This strategy has
been extensively used to enhance the targeting ability and/or
deliver specic cargo to the lesion region in numerous diseases,
such as cancers (Gilligan and Dwyer, 2017;Zhang and Yu, 2019;
Zhou et al., 2021), acute lung injury/acute respiratory distress
syndrome (Zoulikha et al., 2022), inammatory bowel disease
(Ocansey et al., 2020) and Alzheimers disease (Alvarez-Erviti
et al., 2011).
In bone regeneration, several studies have revealed the enhanced
function of exosomes modied by electroporation and sonication.
For example, Wang et al. (2021e) used electroporation to introduce
activating transcription factor 4 (ATF4)mRNAintomiceserum
exosomes, and found the ATF4-overloading exosomes could
promote chondrocyte autophagy and inhibit chondrocyte
apoptosis, which in turn protected cartilage and alleviated
osteoarthritic progression. Zha et al. (2021) encapsulated plasmid
carrying the vascular endothelial growth factor (VEGF)into
exosomes via electroporation, and the gene-activated engineering
exosomes could effectively induce the bulk of vascularized bone
regeneration. Choi et al. (2019) inactivated pre-osteoblast exosomal
let-7, a critical miRNA regulating osteogenesis regulation, by
transfecting let-7 inhibitor into exosomes via electroporation, and
found these exosomes lost the ability to recover osteogenic
differentiation, which conrmed the availability of direct
modication of exosomes strategy from the opposite.
Additionally, although data are scarce, sonication is another
method to load hydrophilic molecules into exosomes, which is
considered much more efcient than electroporation (Kim et al.,
2016). In several studies, the mixture of BMP2 protein and exosomes
was sonicated on ice to construct BMP2-loaded exosomes (Haney
et al., 2015;Yerneni et al., 2021;Yerneni et al., 2022), and these
engineering exosomes could enhance the osteogenic potential of
MC3T3-E1 cells (Yerneni et al., 2022).
Direct modication of exosomes seems a simple and useful
approach to obtain engineering exosomes, but the application of
this strategy is still facing several challenges. The loading
efciency of electroporation is largely suppressed when
transferring oligonucleotides with more than 750 bp length
into exosomes (Lamichhane et al., 2015). Another important
point to consider is that sonication is reported to be the most
damaging technique for exosomal membrane integrity (Donoso-
Quezada et al., 2020). Besides, the size and zeta potential were
reported to affect the efciency of exosome internalization
(Caponnetto et al., 2017;Patel et al., 2019), which should be
taken into consideration in the further research. Therefore, when
using this strategy to product engineering exosomes, it must be
carefully designed to increase loading and internalization
efciency and avoid exosome rupture.
3.2 Chemical or physical treatment of
parent cells
Directly treating parent cells with chemical or physical factors is
an available strategy for engineering exosomes. As originated from
parent cells, the characteristics of exosomes will be reected by the
FIGURE 2
Three strategies of engineering exosomes for bone regeneration. ODM: osteogenic differentiation medium.
Frontiers in Bioengineering and Biotechnology frontiersin.org06
Ma et al. 10.3389/fbioe.2022.1091360
physiological and biochemical alterations of parent cells. Numerous
studies have conrmed that the preconditioning of stem cells via
hypoxia, pharmacological agents, chemical agents, trophic factors,
cytokines, and physical factors could improve stem cellsfunction
in vitro and in vivo (Liu et al., 2011;Ferrer et al., 2013;Yang et al.,
2016;Kheirandish et al., 2017;Yin et al., 2017;Hu and Li, 2018).
Chemical agents and metal ions are the two main treatment
modalities of producing engineering exosomes by chemical
treatment. Culturing parent cells in the osteogenic differentiation
medium (ODM) is the most common method. Liu A. et al. (2021)
isolated exosomes from BMSCs after osteoinductive culturing and
found these engineering exosomes enhanced the bone forming
capacity and induced rapid initiation of bone regeneration. In
other research, umbilical cord mesenchymal stem cells (Ge and
Wang, 2021) and dental pulp stem cells (Xie et al., 2020)were
cultured in the ODM to produce engineering exosomes, which could
enhance osteogenesis. Besides the ODM, many other chemical
agents, TNF-α(Lu et al., 2017), short peptide (Zhao W. et al.,
2021), parathyroid hormone (Shao et al., 2022),
dimethyloxalylglycine (Liang et al., 2019)andBMP2(Wei et al.,
2019), were also used to produce engineering exosomes for bone
defect repair. The metal ions treatment of parent cells can also
endow exosomes with the ability to enhance bone regeneration. The
exosomes derived from BMSCs stimulated by strontium-substituted
calcium silicate ceramics could regulate osteogenesis and
angiogenesis of human umbilical vein endothelial cells (Liu et al.,
2021c). Similarly, macrophage-derived exosomes upon stimulation
with titania nanotubes simultaneously enhanced osteogenesis and
angiogenesis (Wang et al., 2022d).
Moreover, various physical modications of parent cells also
could yield engineering exosomes. Wu et al. (2021a) collected
exosomes from BMSCs stimulated by magnetic nanoparticles and
a static magnetic eld and found these exosomes could improve
osteogenesis and angiogenesis. As oxygen concentration plays a
crucial role in proliferation (Silván et al., 2009), Shen et al. (2022)
found exosomes derived from hypoxia preconditioned MSCs
promote cartilage regeneration via the miR-2055p/PTEN/AKT
pathway. The mechanical force is an essential factor to regulate
the differentiation of stem cells (Halder et al., 2012). Lv et al. (2020)
found exosomes derived from osteocyte induced by mechanical
strain could promote the proliferation and osteogenic differentiation
of human periodontal ligament stem cell. Low yield is one of the
main challenges for the application of engineering exosomes. To
overcome this, Fan et al. (2020) employed an extrusion approach to
amass exosome mimetics (EMs) from human MSCs, and the EMs
demonstrated robust bone regeneration. In other studies, low-
intensity pulsed ultrasound not only promoted BMSC-exosome
release, but enhances the effects of BMSC-exosomes on cartilage
regeneration in osteoarthritis (Liao et al., 2021;Xia et al., 2022).
According to above research, chemical or physical treatment of
parent cells indeed is an effective strategy to produce engineering
exosomes for bone regeneration. It is worthwhile to mention that the
function of the engineering exosomes produced by this strategy still
relies on the exosomal cargos in substantially all these studies.
Therefore, modifying the nucleic acids of parent cells to produce
engineering exosomes with bioactive cargos seems another ideal
strategy, which will be elaborated on below.
3.3 Genetic modication of parent cells
With advances in molecular biological techniques, gene-
editing has become one of the most commonly used
methodologies in molecular research. Consequently,
FIGURE 3
The properties of biomaterial (hydrogel and metal scaffold) help exosomes to promote bone regeneration.
Frontiers in Bioengineering and Biotechnology frontiersin.org07
Ma et al. 10.3389/fbioe.2022.1091360
engineering exosomes with more or totally new bioactive
molecules can be performed by editing certain genes in parent
cells. As described previously, cargos are the basis of exosomes
function, and numerous exosomal cargos (mRNAs, miRNAs,
lncRNAs, circRNAs and proteins) have been conrmed to
promote bone regeneration, which enlightened researchers to
produce engineering exosomes by genetic modication of the
parent cells.
As the most extensively studied exosomal cargos, miRNAs with
the function of promoting bone regeneration received tremendous
attention, and a vast variety of studies have attempted to enhance the
biofunction of exosomes by gene-editing of parent cellsmiRNAs.
Wang N. et al. (2022) transfected BMSCs with lentivirus to obtain
exosomes overexpressing miR-1403p, and found these exosomes
promoted bone defect remodeling. A lentiviral infection system was
also used to overexpress miR-940 in MDA-MB-231 cells to attain
engineering exosomes, which could promote the osteogenic
differentiation of human MSCs (Hashimoto et al., 2018).
Transferring parent cells with miRNAs by Lipofectamine reagent
is another genetic modication method. By this way, exosomes
overexpressing miR-378 (Nan et al., 2021), miR-181b (Liu W. et al.,
2021)andmiR-1225p (Liao et al., 2019) have been demonstrated to
promote osteogenic differentiation.
The mRNA is also an important target for this strategy. Li
et al. (2017) transfected adenovirus carrying mutant HIF-1αinto
BMSCs, and found the mutant protein was highly expressed in
BMSCs exosomes, which markedly accelerated the bone
regeneration and angiogenesis. Interestingly, in several other
TABLE 2 The properties of exosome-integrated biomaterials for bone defect repair.
Properties Biomaterials References
Maintaining exosomes stability Nanocomposite hydrogels Li et al. (2021b)
Enhancing local concentrations of exosomes Acellular extracellular matrix hydrogel Xing et al. (2021)
Injectable hyaluronic acid hydrogel Zhang et al. (2021c)
Gelatin methacrylate/nanoclay hydrogel Hu et al. (2020)
3D matrix hydrogels Holkar et al. (2021)
Injectable thermosensitive hydrogel Ma et al. (2022)
Enhancing exosomes activity 3D matrix hydrogels Yu et al. (2022b)
Alginate hydrogel Holkar et al. (2021)
Optimizing the 3D distribution ABM/P-15 CMC-hydrogel Matos et al. (2012)
3D-printed porous bone scaffolds Zha et al. (2021)
Antibacterial property Food-grade probiotic-modied implant Tan et al. (2020a)
Multifunctional HA hydrogel Liu et al. (2022)
Yu et al. (2022a)
Natural polymer HA hydrogel Mi et al. (2022)
Chitosan hydrogel Shen et al. (2020)
Adapting to irregular bone defects Injectable thermosensitive hydrogel Xing et al. (2021)
PDLLA-PEG-PDLLA triblock copolymer gels Tao et al. (2021)
Chitosan hydrogel Fan et al. (2020)
Wu et al. (2021b)
Nanocomposite hydrogel based on gelatin and Laponite Liu et al. (2021b)
PG/TCP Zhang et al. (2021a)
HA hydrogel Yu et al. (2022a)
Alginate Huang et al. (2020)
Holkar et al. (2021)
Gel-ADH Lin et al. (2022)
Silk broin Shen et al. (2022)
Hyaluronic acid Sang et al. (2022)
Yang et al. (2020b)
SIS-CA hydrogel Ma et al. (2022)
Favorable adhesion Crosslinked network of alginate-dopamine, chondroitin sulfate, and regenerated silk broin Zhang et al. (2021b)
HA hydrogel modied with the PPFLMLLKGSTR peptide Li et al. (2020)
Thermo-sensitivity SIS-CA hydrogel Ma et al. (2022)
ABM/P-15, CMC-hydrogel: bovine-derived mineral bound to a P-15 carboxymethyl cellulose-hydrogel; HA, hyaluronic acid; PDLLA-PEG-PDLLA, poly (D,L-lactide)-b-poly (ethylene
glycol)-b-poly (D, L-lactide); PG/TCP, poly ethylene glycol maleate citrate with β-TCP; Gel-ADH, hydrazide grafted gelatin; SIS-CA, small intestinal submucosa with propionic acid.
Frontiers in Bioengineering and Biotechnology frontiersin.org08
Ma et al. 10.3389/fbioe.2022.1091360
studies, exosomes derived from parent cells with gene-editing of
BMP2 (Huang et al., 2020), Scx (Feng et al., 2021) and P2X7R (Xu
et al., 2020) performed the enhanced osteogenic ability. However,
this modulating function was due to the changes of exosomal
miRNA rather than the transfection of these genes. This might be
due to two reasons: rstly, the cellular components are selectively
packaged into exosomes to be exosomal cargos (Ma et al., 2021),
and gene-editing of certain genes may not inevitably result in
their expression change in exosome; secondly, miRNAs are the
most abundant exosomal cargos, which may be more sensitive to
the gene-editing modication.
Several studies revealed the effect of genetic modication on
other exosomal bioactive molecules (lncRNAs and circRNAs). Cui
et al. (2019) inhibited lncRNA-MALAT1 expression in endothelial
progenitor cells-derived exosomes by transfecting lncRNA-
MALAT1-targeting siRNA, which disrupted bone regeneration.
Cao et al. (2020) subcloned the full sequence of circ-Rtn4 into
the pcDNA-3.1 vector and transfected the vector into BMSCs using
Lipofectamine 2000 to overexpress exosomal circ-Rtn4.
Nevertheless, to date, the research in this eld has remained
limited for the technical reason. Take upregulating circRNAs as
an example, it is difcult to deplete or generate the circular form
without affecting the linear counterpart of circRNA (Nielsen et al.,
2022). In addition, low cyclization efcacy and accuracy also limited
the modication of circRNAs by gene-editing. Therefore,
investigation into novel and high-efciency genetic modication
technologies is required to combat these problems.
4 Properties of exosome-integrated
biomaterials essential for bone defect
repair
Although the strategies of engineering exosomes could
enhance exosome yield and biofunction, exosomes used for
clinical bone defect treatment are still limited (van der Meel
et al., 2014;Lener et al., 2015). Currently, the major modes of
exosome application are direct injection or carrier loading, which
is mainly aimed at systemic diseases, such as osteoporosis (Song
et al., 2019), hematological malignancies (De Luca et al., 2017),
and myocardial ischemia-reperfusion injury (Zhao et al., 2019).
Nevertheless, it has been reported that no signicant effect was
observed with free exosomes treatment by direct injection,
because of its rapid excretion from the site of application
(Zhang Z. et al., 2018;Wang C. et al., 2019;Xing et al., 2022),
suggeting the medand for exosome-integrated biomaterials.
Currently, more and more biomaterials have been designed
and applied in bone regeneration (Cui et al., 2020;Zhao D.
et al., 2021;Zhu et al., 2022a;Zhu et al., 2022b;Wang et al., 2022c;
Zhang et al., 2022). Therefore, the selection of available
biomaterials with appropriate stability and integrity to load
and release exosomes at the bone defect site to increase their
retention and stability may be necessary for bone regeneration.
Several excellent and informative reviews have addressed the
types, synthetic procedure and/or encapsulation approaches of
biomaterials used to carry exosomes (Riau et al., 2019;Pishavar
et al., 2021;Wang D. et al., 2022;Sun et al., 2022). Instead, we
propose to streamline the properties of biomaterial to dissect how
and by what mechanisms the biomaterials help exosomes to
promote bone regeneration (Figure 3;Table 2). By summarizing
the previous studies, we expected to represent a promising
strategy for the use of engineering exosomes in combination
with biomaterials for clinical bone regeneration.
4.1 Maintaining the exosome stability
The rst consideration is how to maintain the stability of
exosomes. Despite the bilayer membrane structures making
exosomes resist degradation to some extent, exosomes are
unstable and maintain for less 48 h at room temperature
(Chew et al., 2019). The time will be even shorter at 37°C, at
which exposed functional substances (proteins and RNA) will be
rapidly degraded and metabolized. In fact, stability is an
important but often overlooked point in the research of
biomaterials loading exosomes, which should be given
sufcient attention in the further. Hydrogel encapsulated
exosomes was reported to protect them without degradation
and supply therapeutic effects with persistent exosomes delivery
(Riau et al., 2019). Li et al. (2021b) used the gelatin and laponite
to prepare nanocomposite hydrogels as a carrier for exosomes to
extend the time of BMSC-exosomes in the periodontal pocket
and enhance their osteoinductive function.
4.2 Enhancing local concentrations of
exosomes
The therapeutic effect of exosomes depends strongly on the
local concentrations. However, it is demanding to produce
exosomes in large quantities with high quality and purity,
making clinical applications of exosomes more expensive.
Additionally, free exosomes diffused out from the defect
rapidly, resulting in no exertion of exosomal cargo activity
(Riau et al., 2019). According to the research of Lai et al.
(2012), biodistribution proceeds in the stages of liver and
lungs for 30 min after direct injection of exosomes, and
exosomes are removed within 1 h6 h after administration via
liver and kidney treatment. Thus, much research was performed
with the aim of enhancing the retention and sustained-releasing
of exosomes at the defect site. Xing et al. (2021) synthesized an
acellular extracellular matrix hydrogel coupled with adipose-
derived mesenchymal stem cell exosomes to regulate the
intervertebral disc microenvironment for the treatment of
intervertebral disc degeneration. The decomposition of the
hydrogel was slowed down, allowing exosomes to remain in
Frontiers in Bioengineering and Biotechnology frontiersin.org09
Ma et al. 10.3389/fbioe.2022.1091360
the disc for up to 28 days Zhang Y. et al. (2021) fabricated an
injectable hyaluronic acid hydrogel encapsulated with umbilical
MSC-derived exosomes through three-dimensional (3D)
printing technology, and the hydrogel showed good sustained-
releasing features in the rat critical-size cranial defect model. Hu
et al. (2020) fabricated Gelatin methacrylate/nanoclay hydrogel
for sustained release of exosomes. The hydrogels with a 3D
matrix prevent the dispersion of exosomes and maintained
their local concentration, which enable the controlled release
of exosomes at bone defect sites (Holkar et al., 2021). In another
study, exosomes were incorporated into an injectable
thermosensitive hydrogel by constructing fusion peptides,
which also enhanced the retention of exosomes and improved
the biological activity of exosomes (Ma et al., 2022). Generally,
the consensus has been achieved that it is essential to enhance
local exosome concentrations at the bone defect site, and the
biomaterials loading exosomes should possess the property of
enhancing the retention and sustained-releasing of exosomes.
4.3 Enhancing exosomes activity
The hydrogel with 3D microenvironment can enhance exosome
activity and affect the interaction of integrin membrane protein
between cells and the cell matrix, which promotes cell proliferation
and differentiation in a bone regeneration environment. Yu W. et al.
(2022) encapsulated exosomes derived from periodontal ligament
stem cells into a hydrogel with 3D microenvironment, which
enhanced osteoinductive ability and signicantly promoted bone
defect repair in rats. Another study demonstrated alginate hydrogels
combined with exosomes promoted osteogenesis by increasing cell-
exosomes interactions, cell aggregation, and long-term viability
(Holkar et al., 2021).
4.4 Optimizing the 3D distribution of
exosomes
Biomaterials with a highly porous and 3D structure mimic
the porosity, pore size, and interconnectedness of native bone
ideally. In bone defects repair, bioactive materials with good
mechanical properties not only provide temporary mechanical
support for the bone at the implant site, but also modulate
extracellular matrix formation, facilitate better cell-cell and
cell-matrix interactions, retain the cell morphology, provide
mechanical stimulations, and support cell growth and
exosomes secretion, the features which are akin to in vivo
systems (Tibbitt and Anseth, 2009). Matos et al. (2012)
showed that the lyophilized biomaterials created a more
homogenous interparticle spacing, allowed a more suitable
particle distribution and stabilization, then promoting a faster
bone regeneration with relevant clinical benets. Similarly,
biocompatible 3D porous biomaterials ensured a uniform
spacing and stable distribution of MSC-exosomes compared
with compacted materials (Zha et al., 2021).
4.5 Antibacterial property
During bone defect healing, the bacterial infection is one of the
risk factors (Blair et al., 2015). Therefore, antibacterial property of
biomaterials should also be taken into consideration. Tan L. et al.
(2020) developed a food-grade probiotic-modied implant to
prevent methicillin-resistant Staphylococcus aureus infection and
accelerated bone integration. Liu et al. (2022) designed a
multifunctional hyaluronic acid (HA) hydrogel with antibacterial
property. Then, this group loaded plasma exosomes to this hydrogel
for promoting infected fracture healing (Yu C. et al., 2022). Mi et al.
(2022) combined engineered exosomes and a natural polymer HA
hydrogel, which performed an anti-inammatory and antibacterial
function on fracture repair acceleration. As a cationic natural
polymer biomaterial, chitosan has anti-microbial property (Dai
et al., 2011), and many reports have shown cationic loaded
engineering exosomes could promote bone regeneration (Fan
et al., 2020;Shen et al., 2020;Wang et al., 2020;Wu et al.,
2021b;Bahar et al., 2022;Nikhil and Kumar, 2022), although
some of them did not look at the antibacterial property. The
biomaterials with antibacterial property have been designed in
some studies, but these materials are still rarely used for bone
defect repair, which merits further investigation.
4.6 Adapting to irregular bone defects
Clinical bone defects caused by trauma, neoplasia,
infection or corrective osteotomies are always irregular.
Hence, biomaterials should have the injectable property to
ll irregular defects and promote in situ bone tissue
regeneration. Xing et al. (2021) constructed an injectable
thermosensitive hydrogel system via a coordinative crossing
of ADSC-derived exosomes and acellular extracellular matrix
hydrogels to effectively protect nucleus pulposus cells from
pyroptosis after intervertebral disc degeneration. By taking
advantage of injectable, reversible, and thermosensitive
abilities, Tao et al. (2021) used PDLLA-PEG-PDLLA
triblock copolymer gels as a carrier of synovium
mesenchymal stem cells-derived exosomes for intra-
articular injection to prevent osteoarthritis progression.
Additionally, multiple biomaterials, including chitosan
hydrogel (Fan et al., 2020;Wu et al., 2021b),
nanocomposite hydrogel (based on gelatin and Laponite)
(Liu et al., 2021b), PG/TCP (PEGMC with β-TCP) (Zhang
B. et al., 2021), HA hydrogel (Yu C. et al., 2022), alginate
(Huang et al., 2020;Holkar et al., 2021), Gel-ADH (hydrazide
grafted gelatin) (Lin et al., 2022), silk broin (Shen et al.,
2022), hyaluronic acid (Yang S. et al., 2020;Sang et al., 2022)
Frontiers in Bioengineering and Biotechnology frontiersin.org10
Ma et al. 10.3389/fbioe.2022.1091360
and SIS-CA (small intestinal submucosa (SIS) with propionic
acid (CA)) hydrogel (Ma et al., 2022)werereportedtoadapt
irregular bone defects and were used to carry exosomes for
skeletal regeneration. Generally, although local injection
therapy is not suitable for certain types of bone
regeneration, such as spinal cord repair (Han M. et al.,
2022), injectable biomaterials loading engineering exosomes
have been used extensively to repair irregular bone defects.
4.7 Other properties
In a moist environment, the favorable adhesion of
biomaterials is essential for in situ bone regeneration (Hasani-
Sadrabadi et al., 2020;Li et al., 2020;Zhang FX. et al., 2021).
Inspired by mussel materials, which exhibit underwater robust
adhesion (Gao Z. et al., 2018), Zhang and the colleague (2021b)
prepared a hydrogel with high bonding strength to the wet
surface using a crosslinked network of alginate-dopamine,
chondroitin sulfate, and regenerated silk broin, which
promote cartilage defect regeneration by combining with
BMSCs-exosomes. Peptide-modication is another strategy for
enhancing biomaterial adhesion. Li et al. (2020) prepared a
biomaterial with high adhesion by modifying HA hydrogel
with the PPFLMLLKGSTR peptide, which could locally deliver
human placenta amniotic membrane mesenchymal stem cell-
derived exosomes in spinal cord tissue.
Biomaterials with thermo-sensitivity are also of particular
concern for their property, changing between a liquid state and a
solid-state based on the ambient temperature (López-Noriega et al.,
2014;Ni et al., 2014). Several thermo-sensitive biomaterials have
been used in bone defect repair (Fu et al., 2012;Kim et al., 2018;Yu
et al., 2020;Wang QS. et al., 2021). Further, Ma et al. (2022) designed
a novel thermo-sensitive biomaterial by loading BMSCs-exosomes
with SIS-CA hydrogel to regulate bone regeneration.
Collectively, the bio-functional materials not only provide a
scaffold or carrier for engineering exosomes, but also play an
essential role by their own variety properties. Along with major
advancements in chemical engineering techniques, more and
more novel biomaterials with various properties have been
synthesized for bone regeneration. In the future, selection of
appropriate biomaterials to integrate engineering exosomes
should be one of the leading focuses of bone defect repair.
5 Conclusion and perspective
Coexistence of challenges and opportunities have greatly
stimulated the study of engineering exosomes for bone
regeneration in the past 10 years. In the present review, we
mainly addressed the molecular basis of exosomal cargos, the
strategies of engineering exosomesandthepropertiesofexosome-
integrated biomaterials required for bone regeneration. The research
about engineering exosomes for bone defect repair is undeniably in
its infancy. The rapid development of engineering exosomes is
impeded by several key challenges, especially the consistency of
exosomes production. As a result, these difculties inspired the
development of new and cutting-edge approaches, often distinct
from those in the conventional study of cells, to address both
exosome production and function. Rening the isolation,
purication and storage techniques of exosomes may be an
effective means of improving the consistency of exosomes
production (Colao et al., 2018;Zeng et al., 2022). Additionally,
excellent biomaterials emerged continuously, which greatly
promoted research self-renewal. The effective combination of
engineering exosomes and biomaterials will be greater than the
sum of their parts and exhibit synergy effects in bone
regeneration. We optimistically foresee that novel biomaterials will
be constructed and more sophisticated engineering exosomes will be
implemented for bone tissue regeneration. This huge progress is sure
to benet both biomedical research and therapeutic modalities in the
eld of bone regeneration.
Author contributions
Conceptualization, SM, YL, and DP; writingoriginal draft
preparation, SM, YZ, and SL; writingreview and editing, SM,
AL, and DP; visualization, SM and YZ; funding acquisition, DP,
YL, and SM. All authors have read and agreed to the published
version of the manuscript.
Funding
This work was supported by National Natural Science
Foundation of China (No. 81870798 and 82170927) and the
Fundamental Research Funds for the Central Universities, Xian
Jiaotong University (xzy012021066).
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could
be construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the
authors and do not necessarily represent those of their afliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or
claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Frontiers in Bioengineering and Biotechnology frontiersin.org11
Ma et al. 10.3389/fbioe.2022.1091360
References
Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., and Wood, M. J. (2011).
Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes.
Nat. Biotechnol. 29, 341345. [PubMed: 341]. doi:10.1038/nbt.1807
Ando, Y., Matsubara, K., Ishikawa, J., Fujio, M., Shohara, R., Hibi, H., et al. (2014).
Stem cell-conditioned medium accelerates distraction osteogenesis through
multiple regenerative mechanisms. Bone 61, 8290. [PubMed: 82]. doi:10.1016/j.
bone.2013.12.029
Bahar, D., Gonen, Z. B., Gumusderelioglu, M., Onger, M. E., Tokak, E. K., Ozturk-
Kup, F., et al. (2022). Repair of rat calvarial bone defect by using exosomes of
umbilical cord-derived mesenchymal stromal cells embedded in chitosan/
hydroxyapatite scaffolds. Int. J. Oral Maxillofac. Implants 37, 943950.
[PubMed: 943]. doi:10.11607/jomi.9515
Balatti, V., Nigita, G., Veneziano, D., Drusco, A., Stein, G. S., Messier, T. L., et al.
(2017). tsRNA signatures in cancer. Proc. Natl. Acad. Sci. U. S. A. 114, 80718076.
[PubMed: 28696308]. doi:10.1073/pnas.1706908114
Behera, J., Kumar, A., Voor, M. J., and Tyagi, N. (2021). Exosomal lncRNA-H19
promotes osteogenesis and angiogenesis through mediating Angpt1/Tie2-NO
signaling in CBS-heterozygous mice. Theranostics 11, 77157734. [PubMed:
34335960]. doi:10.7150/thno.58410
Bei, H. P., Hung, P. M., Yeung, H. L., Wang, S., and Zhao, X. (2021). Bone-a-
Petite: Engineering exosomes towards bone, osteochondral, and cartilage repair.
Small 17, e2101741. [PubMed: 34288410]. doi:10.1002/smll.202101741
Bharadwaz, A., and Jayasuriya, A. C. (2020). Recent trends in the application of
widely used natural and synthetic polymer nanocomposites in bone tissue
regeneration. Mater. Sci. Eng. C 110, 110698. [PubMed: 32204012]. doi:10.1016/
j.msec.2020.110698
Bhat, A. A., Younes, S. N., Raza, S. S., Zarif, L., Nisar, S., Ahmed, I., et al. (2020).
Role of non-coding RNA networks in leukemia progression, metastasis and drug
resistance. Mol. Cancer 19, 57. [PubMed: 32164715]. doi:10.1186/s12943-020-
01175-9
Blair, J. M., Webber, M. A., Baylay, A. J., Ogbolu, D. O., and Piddock, L. J. (2015).,
13. PubMed, 42424251. doi:10.1038/nrmicro3380Molecular mechanisms of
antibiotic resistanceNat. Rev. Microbiol.
Blanco, S., Bandiera, R., Popis, M., Hussain, S., Lombard, P., Aleksic, J., et al.
(2016). Stem cell function and stress response are controlled by protein synthesis.
Nature 534, 335340. [PubMed: 27306184]. doi:10.1038/nature18282
Bobrie, A., Krumeich, S., Reyal, F., Recchi, C., Moita, L. F., Seabra, M. C., et al.
(2012). Rab27a supports exosome-dependent and -independent mechanisms that
modify the tumor microenvironment and can promote tumor progression. Cancer
Res. 72, 49204930. [PubMed: 4920]. doi:10.1158/0008-5472.Can-12-0925
Cao, G., Meng, X., Han, X., and Li, J. (2020). Exosomes derived from circRNA
Rtn4-modied BMSCs attenuate TNF-α-induced cytotoxicity and apoptosis in
murine MC3T3-E1 cells by sponging miR-146a. Biosci. Rep. 40, BSR20193436.
[PubMed: 32400849]. doi:10.1042/bsr20193436
Cao, Z., Wu, Y., Yu, L., Zou, L., Yang, L., Lin, S., et al. (2021). Exosomal miR-335
derived from mature dendritic cells enhanced mesenchymal stem cell-mediated
bone regeneration of bone defects in athymic rats. Mol. Med. 27, 20. [PubMed:
33637046]. doi:10.1186/s10020-021-00268-5
Caponnetto, F., Manini, I., Skrap, M., Palmai-Pallag, T., Di Loreto, C., Beltrami,
A. P., et al. (2017). Size-dependent cellular uptake of exosomes. Nanomedicine
Nanotechnol. Biol. Med. 13 (3), 10111020. [PMID: 27993726]. doi:10.1016/j.nano.
2016.12.009
Castillo-Cardiel, G., López-Echaury, A. C., Saucedo-Ortiz, J. A., Fuentes-Orozco,
C., Michel-Espinoza, L. R., Irusteta-Jiménez, L., et al. (2017). Bone regeneration in
mandibular fractures after the application of autologous mesenchymal stem cells, a
randomized clinical trial. Dent. Traumatol. 33, 3844. [PubMed: 27513920]. doi:10.
1111/edt.12303
Chen, C., Qu, Z., Yin, X., Shang, C., Ao, Q., Gu, Y., et al. (2016). Efcacy of
umbilical cord-derived mesenchymal stem cell-based therapy for osteonecrosis of
the femoral head: A three-year follow-up study. Mol. Med. Rep. 14, 42094215.
[PubMed: 27634376]. doi:10.3892/mmr.2016.5745
Chen, Q., Zhang, X., Shi, J., Yan, M., and Zhou, T. (2021). Origins and evolving
functionalities of tRNA-derived small RNAs. Trends Biochem. Sci. 46, 790804.
[PubMed: 790]. doi:10.1016/j.tibs.2021.05.001
Chen, X., Wan, Z., Yang, L., Song, S., Fu, Z., Tang, K., et al. (2022). Exosomes
derived from reparative M2-like macrophages prevent bone loss in murine
periodontitis models via IL-10 mRNA. J. Nanobiotechnol. 20, 110. [PubMed:
35248085]. doi:10.1186/s12951-022-01314-y
Chew, J. R. J., Chuah, S. J., Teo, K. Y. W., Zhang, S., Lai, R. C., Fu, J. H., et al.
(2019). Mesenchymal stem cell exosomes enhance periodontal ligament cell
functions and promote periodontal regeneration. Acta Biomater. 89, 252264.
[PubMed: 252]. doi:10.1016/j.actbio.2019.03.021
Choi, S. Y., Han, E. C., Hong, S. H., Kwon, T. G., Lee, Y., and Lee, H. J. (2019).
Regulating osteogenic differentiation by suppression of exosomal microRNAs.
Tissue Eng. Part A 25, 11461154. [PubMed: 1146]. doi:10.1089/ten.TEA.2018.0257
Colao, I. L., Corteling, R., Bracewell, D., and Wall, I. (2018). Manufacturing
exosomes: A promising therapeutic platform. Trends Mol. Med. 24 (3), 242256.
[PMID: 29449149]. doi:10.1016/j.molmed.2018.01.006
Colombo, M., Raposo, G., and Thery, C. (2014). Biogenesis, secretion, and
intercellular interactions of exosomes and other extracellular vesicles. Annu.
Rev. Cell Dev. Biol. 30, 255289. [PubMed: 25288114]. doi:10.1146/annurev-
cellbio-101512-122326
Crick, F. (1970)., 227. PubMed, 561561561563. doi:10.1038/227561a0Central
dogma of molecular biologyNature
Cui, L., Zhang, J., Zou, J., Yang, X., Guo, H., Tian, H., et al. (2020). Electroactive
composite scaffold with locally expressed osteoinductive factor for synergistic bone
repair upon electrical stimulation. Biomaterials 230, 119617. [PubMed: 31771859.
doi:10.1016/j.biomaterials.2019.119617
Cui, Y., Fu, S., Sun, D., Xing, J., Hou, T., and Wu, X. (2019)., 23. PubMed,
3843384338433854. doi:10.1111/jcmm.14228EPC-derived exosomes promote
osteoclastogenesis through LncRNA-MALAT1J. Cell. Mol. Med.
Dai, T., Tanaka, M., Huang, Y. Y., and Hamblin, M. R. (2011). Chitosan
preparations for wounds and burns: Antimicrobial and wound-healing effects.
Expert Rev. Anti-infective Ther. 9, 857879. [PubMed: 21810057]. doi:10.1586/eri.
11.59
De Luca, L., Trino, S., Laurenzana, I., Lamorte, D., Caivano, A., Del Vecchio, L.,
et al. (2017). Mesenchymal stem cell derived extracellular vesicles: A role in
hematopoietic transplantation? Int. J. Mol. Sci. 18, 1022. [PubMed: 1022. doi:10.
3390/ijms18051022
Donoso-Quezada, J., Ayala-Mar, S., and González-Valdez, J. (2020). State-of-the-
art exosome loading and functionalization techniques for enhanced therapeutics: A
review. Crit. Rev. Biotechnol. 40, 804820. [PubMed: 804]. doi:10.1080/07388551.
2020.1785385
Du, Z., Yuan, J., Wu, Z., Chen, Q., Liu, X., and Jia, J. (2022). Circulating exosomal
circRNA_0063476 impairs expression of markers of bone growth via the miR-518c-
3p/DDX6 Axis in ISS. Endocrinology 163, bqac138. [PubMed: 35974445]. doi:10.
1210/endocr/bqac138
Elefteriou, F. (2018). Impact of the autonomic nervous system on the skeleton.
Physiol. Rev. 98, 10831112. [PubMed: 1083]. doi:10.1152/physrev.00014.2017
Fan, J., Lee, C. S., Kim, S., Chen, C., Aghaloo, T., and Lee, M. (2020). Generation
of small rna-modulated exosome mimetics for bone regeneration. ACS Nano 14,
1197311984. [PubMed: 11973]. doi:10.1021/acsnano.0c05122
Fang, S., He, T., Jiang, J., Li, Y., and Chen, P. (2020). <p&gt;Osteogenic effect of
tsRNA-10277-loaded exosome derived from bone mesenchymal stem cells on
steroid-induced osteonecrosis of the femoral head</p&gt;. Drug Des. Devel Ther.
14, 45794591. [PubMed: 4579]. doi:10.2147/dddt.S258024
Feng, W., Jin, Q., Ming-Yu, Y., Yang, H., Xu, T., You-Xing, S., et al. (2021). MiR-
6924-5p-rich exosomes derived from genetically modied Scleraxis-overexpressing
PDGFRα(+) BMMSCs as novel nanotherapeutics for treating osteolysis during
tendon-bone healing and improving healing strength. Biomaterials 279, 121242.
[PubMed: 34768151]. doi:10.1016/j.biomaterials.2021.121242
Ferrer, R. A., Wobus, M., List, C., Wehner, R., Schönefeldt, C., Brocard, B., et al.
(2013). Mesenchymal stromal cells from patients with myelodyplastic syndrome
display distinct functional alterations that are modulated by lenalidomide.
Haematologica 98, 16771685. [PubMed: 1677]. doi:10.3324/haematol.2013.083972
Fu, S., Ni, P., Wang, B., Chu, B., Zheng, L., Luo, F., et al. (2012). Injectable and
thermo-sensitive PEG-PCL-PEG copolymer/collagen/n-HA hydrogel composite
for guided bone regeneration. Biomaterials 33, 48014809. [PubMed: 4801].
doi:10.1016/j.biomaterials.2012.03.040
Furuta, T., Miyaki, S., Ishitobi, H., Ogura, T., Kato, Y., Kamei, N., et al. (2016).
Mesenchymal stem cell-derived exosomes promote fracture healing in a mouse
model. Stem Cells Transl. Med. 5, 16201630. [PubMed: 27460850]. doi:10.5966/
sctm.2015-0285
Gao, X., Ran, N., Dong, X., Zuo, B., Yang, R., Zhou, Q., et al. (2018a). Anchor
peptide captures, targets, and loads exosomes of diverse origins for diagnostics and
therapy. Sci. Transl. Med. 10, eaat0195. [PubMed: 29875202]. doi:10.1126/
scitranslmed.aat0195
Gao, Z., Duan, L., Yang, Y., Hu, W., and Gao, G. (2018b). Mussel-inspired tough
hydrogels with self-repairing and tissue adhesion. Appl. Surf. Sci. 427, 7482. doi:10.
1016/j.apsusc.2017.08.157
Frontiers in Bioengineering and Biotechnology frontiersin.org12
Ma et al. 10.3389/fbioe.2022.1091360
Ge, Y., and Wang, X. (2021). The role and mechanism of exosomes from
umbilical cord mesenchymal stem cells in inducing osteogenesis and preventing
osteoporosis. Cell Transpl. 30, 096368972110574. [PubMed: 34814742]. doi:10.
1177/09636897211057465
Gilligan, K. E., and Dwyer, R. M. (2017). Engineering exosomes for cancer
therapy. Int. J. Mol. Sci. 18, 1122. [PubMed: 28538671]. doi:10.3390/ijms18061122
Guo, J., Zhou, F., Liu, Z., Cao, Y., Zhao, W., Zhang, Z., et al. (2022). Exosome-
shuttled mitochondrial transcription factor A mRNA promotes the osteogenesis of
dental pulp stem cells through mitochondrial oxidative phosphorylation activation.
Cell Prolif., e13324. [PubMed: 36054692]. doi:10.1111/cpr.13324
Ha, M., and Kim, V. N. (2014). Regulation of microRNA biogenesis. Nat. Rev.
Mol. Cell Biol. 15, 509524. [PubMed: 25027649]. doi:10.1038/nrm3838
Halder, G., Dupont, S., and Piccolo, S. (2012). Transduction of mechanical and
cytoskeletal cues by YAP and TAZ. Nat. Rev. Mol. Cell Biol. 13, 591600. [PubMed:
591. doi:10.1038/nrm3416
Han, L., Liu, H., Fu, H., Hu, Y., Fang, W., and Liu, J. (2022a). Exosome-delivered
BMP-2 and polyaspartic acid promotes tendon bone healing in rotator cuff tear via
Smad/RUNX2 signaling pathway. Bioengineered 13, 14591475. [PubMed: 1459].
doi:10.1080/21655979.2021.2019871
Han, M., Yang, H., Lu, X., Li, Y., Liu, Z., Li, F., et al. (2022b). Three-dimensional-
cultured MSC-derived exosome-hydrogel hybrid microneedle array patch for spinal
cord repair. Nano Lett. 22, 63916401. [PubMed: 35876503]. doi:10.1021/acs.
nanolett.2c02259
Haney, M. J., Klyachko, N. L., Zhao, Y., Gupta, R., Plotnikova, E. G., He, Z., et al.
(2015)., 207. PubMed, 18181830. doi:10.1016/j.jconrel.2015.03.033Exosomes as
drug delivery vehicles for Parkinsons disease therapyJ. Control. Release
Hasani-Sadrabadi, M. M., Sarrion, P., Pouraghaei, S., Chau, Y., Ansari, S., Li, S.,
et al. (2020). An engineered cell-laden adhesive hydrogel promotes craniofacial
bone tissue regeneration in rats. Sci. Transl. Med. 12, eaay6853. [PubMed:
eaay6853]. doi:10.1126/scitranslmed.aay6853
Hashimoto, K., Ochi, H., Sunamura, S., Kosaka, N., Mabuchi, Y., Fukuda, T., et al.
(2018). Cancer-secreted hsa-miR-940 induces an osteoblastic phenotype in the
bone metastatic microenvironment via targeting ARHGAP1 and FAM134A. Proc.
Natl. Acad. Sci. U. S. A. 115, 22042209. [PubMed: 29440427]. doi:10.1073/pnas.
1717363115
Hergenreider, E., Heydt, S., Tréguer, K., Boettger, T., Horrevoets, A. J., Zeiher, A.
M., et al. (2012). Atheroprotective communication between endothelial cells and
smooth muscle cells through miRNAs. Nat. Cell Biol. 14, 249256. [PubMed: 249].
doi:10.1038/ncb2441
Hernigou, P., Auregan, J. C., Dubory, A., Flouzat-Lachaniette, C. H., Chevallier,
N., and Rouard, H. (2018a). Subchondral stem cell therapy versus contralateral total
knee arthroplasty for osteoarthritis following secondary osteonecrosis of the knee.
Int. Orthop. 42, 25632571. [PubMed: 29589086]. doi:10.1007/s00264-018-3916-9
Hernigou, P., Dubory, A., Homma, Y., Guissou, I., Flouzat Lachaniette, C. H.,
Chevallier, N., et al. (2018b). Cell therapy versus simultaneous contralateral
decompression in symptomatic corticosteroid osteonecrosis: A thirty year
follow-up prospective randomized study of one hundred and twenty ve adult
patients. Int. Orthop. 42, 16391649. [PubMed: 29744647]. doi:10.1007/s00264-
018-3941-8
Holkar, K., Kale, V., and Ingavle, G. (2021). Hydrogel-Assisted 3D model to
investigate the osteoinductive potential of mc3t3-derived extracellular vesicles. ACS
Biomater. Sci. Eng. 7, 26872700. [PubMed: 34018721]. doi:10.1021/
acsbiomaterials.1c00386
Hoshino, A., Costa-Silva, B., Shen, T. L., Rodrigues, G., Hashimoto, A., Tesic
Mark, M., et al. (2015). Tumour exosome integrins determine organotropic
metastasis. Nature 527, 329335. [PubMed: 26524530]. doi:10.1038/nature15756
Hu, C., and Li, L. (2018). Preconditioning inuences mesenchymal stem cell
properties in vitro and in vivo.J. Cell. Mol. Med. 22, 14281442. [PubMed: 1428 ].
doi:10.1111/jcmm.13492
Hu, H., Dong, L., Bu, Z., Shen, Y., Luo, J., Zhang, H., et al. (2020). miR-23a-3p-
abundant small extracellular vesicles released from Gelma/nanoclay hydrogel for
cartilage regeneration. J. Extracell. Vesicles 9, 1778883. [PubMed: 32939233]. doi:10.
1080/20013078.2020.1778883
Huang,C.C.,Kang,M.,Lu,Y.,Shirazi,S.,Diaz,J.I.,Cooper,L.F.,etal.
(2020). Functionally engineered extracellular vesicles improve bone
regeneration. Acta Biomater. 109, 182194. [PubMed: 182]. doi:10.1016/j.
actbio.2020.04.017
Ishikawa, M., Ito, H., Kitaori, T., Murata, K., Shibuya, H., Furu, M., et al. (2014).
MCP/CCR2 signaling is essential for recruitment of mesenchymal progenitor cells
during the early phase of fracture healing. PLoS One 9, e104954. [PubMed:
25133509][. doi:10.1371/journal.pone.0104954
Jeck, W. R., and Sharpless, N. E. (2014). Detecting and characterizing circular
RNAs. Nat. Biotechnol. 32, 453461. [PubMed: 453]. doi:10.1038/nbt.2890
Jia, Y., Zhu, Y., Qiu, S., Xu, J., and Chai, Y. (2019). Exosomes secreted by
endothelial progenitor cells accelerate bone regeneration during distraction
osteogenesis by stimulating angiogenesis. Stem Cell Res. Ther. 10, 12. [PubMed:
30635031]. doi:10.1186/s13287-018-1115-7
Jin, Y., Xu, M., Zhu, H., Dong, C., Ji, J., Liu, Y., et al. (2021). Therapeutic effects of
bone marrow mesenchymal stem cells-derived exosomes on osteoarthritis. J. Cell.
Mol. Med. 25, 92819294. [PubMed: 34448527]. doi:10.1111/jcmm.16860
Jing, X., Wang, S., Tang, H., Li, D., Zhou, F., Xin, L., et al. (2022). Dynamically
bioresponsive DNA hydrogel incorporated with dual-functional stem cells from
apical papilla-derived exosomes promotes diabetic bone regeneration. ACS Appl.
Mat. Interfaces 14, 1608216099. [PubMed: 35344325]. doi:10.1021/acsami.
2c02278
Kalluri, R., and LeBleu, V. S. (2020). The biology, function, and biomedical
applications of exosomes. Science 367. [PubMed: eaau6977]. doi:10.1126/science.
aau6977
Kang, M., Huang, C. C., Lu, Y., Shirazi, S., Gajendrareddy, P., Ravindran, S., et al.
(2020). Bone regeneration is mediated by macrophage extracellular vesicles. Bone
141, 115627. [PubMed: 32891867]. doi:10.1016/j.bone.2020.115627
Kheirandish, M., Gavgani, S. P., and Samiee, S. (2017). The effect of hypoxia
preconditioning on the neural and stemness genes expression proling in human
umbilical cord blood mesenchymal stem cells. Transfus. Apher. Sci. 56, 392399.
[PubMed: 392]. doi:10.1016/j.transci.2017.03.015
Kim, M. H., Kim, B. S., Park, H., Lee, J., and Park, W. H. (2018). Injectable
methylcellulose hydrogel containing calcium phosphate nanoparticles for bone
regeneration. Int. J. Biol. Macromol. 109, 5764. [PubMed: 57]. doi:10.1016/j.
ijbiomac.2017.12.068
Kim, M. S., Haney, M. J., Zhao, Y., Mahajan, V., Deygen, I., Klyachko, N. L., et al.
(2016). Development of exosome-encapsulated paclitaxel to overcome MDR in
cancer cells. Nanomedicine Nanotechnol. Biol. Med. 12, 655664. [PubMed: 655].
doi:10.1016/j.nano.2015.10.012
Kim, Y. G., Choi, J., and Kim, K. (2020). Mesenchymal stem cell-derived
exosomes for effective cartilage tissue repair and treatment of osteoarthritis.
Biotechnol. J. 15, 2000082. [PubMed: 32559340]. doi:10.1002/biot.202000082
Kitaori, T., Ito, H., Schwarz, E. M., Tsutsumi, R., Yoshitomi, H., Oishi, S., et al.
(2009). Stromal cell-derived factor 1/CXCR4 signaling is critical for the recruitment
of mesenchymal stem cells to the fracture site during skeletal repair in a mouse
model. Arthritis Rheum. 60, 813823. [PubMed: 19248097]. doi:10.1002/art.24330
Kojima, R., Bojar, D., Rizzi, G., Hamri, G. C., El-Baba, M. D., Saxena, P., et al.
(2018). Designer exosomes produced by implanted cells intracerebrally deliver
therapeutic cargo for Parkinsons disease treatment. Nat. Commun. 9, 1305.
[PubMed:29610454]. doi:10.1038/s41467-018-03733-8
Kopp, F., and Mendell, J. T. (2018). Functional classication and experimental
dissection of long noncoding RNAs. Cell 172, 393407. [PubMed: 393]. doi:10.1016/
j.cell.2018.01.011
Kuang, M. J., Huang, Y., Zhao, X. G., Zhang, R., Ma, J. X., Wang, D. C., et al.
(2019). Exosomes derived from Whartons jelly of human umbilical cord
mesenchymal stem cells reduce osteocyte apoptosis in glucocorticoid-induced
osteonecrosis of the femoral head in rats via the miR-21-PTEN-AKT signalling
pathway. Int. J. Biol. Sci. 15, 18611871. [PubMed: 1861]. doi:10.7150/ijbs.32262
Lai, R. C., Tan, S. S., Teh, B. J., Sze, S. K., Arslan, F., de Kleijn, D. P., et al. (2012).
Proteolytic potential of the msc exosome proteome: Implications for an exosome-
mediated delivery of therapeutic proteasome. Int. J. Proteomics 2012, 114.
[PubMed: 22852084]. doi:10.1155/2012/971907
Lamichhane, T. N., Raiker, R. S., and Jay, S. M. (2015). Exogenous DNA loading
into extracellular vesicles via electroporation is size-dependent and enables limited
gene delivery. Mol. Pharm. 12, 36503657. [PubMed: 3650]. doi:10.1021/acs.
molpharmaceut.5b00364
Lander, E. S., Linton, L. M., Birren, B., Nusbaum, C., Zody, M. C., Baldwin, J., et al.
(2001). Initial sequencing and analysis of the human genome. Nature 409, 860921.
[PubMed: 860]. doi:10.1038/35057062
Lathwal, S., Yerneni, S. S., Boye, S., Muza, U. L., Takahashi, S., Sugimoto, N., et al.
(2021). Engineering exosome polymer hybrids by atom transfer radical
polymerization. Proc. Natl. Acad. Sci. U. S. A. 118, e2020241118. [PubMed:
33384328]. doi:10.1073/pnas.2020241118
Lei, F., Li, M., Lin, T., Zhou, H., Wang, F., and Su, X. (2022). Treatment of
inammatory bone loss in periodontitis by stem cell-derived exosomes. Acta
Biomater. 141, 333343. [PubMed: 333]. doi:10.1016/j.actbio.2021.12.035
Lener, T., Gimona, M., Aigner, L., Börger, V., Buzas, E., Camussi, G., et al. (2015).
Applying extracellular vesicles based therapeutics in clinical trials - an ISEV
position paper. J. Extracell. Vesicles 4, 30087. [PubMed: 26725829]. doi:10.3402/
jev.v4.30087
Li, H., Liu, D., Li, C., Zhou, S., Tian, D., Xiao, D., et al. (2017). Exosomes secreted
from mutant-HIF-1α-modied bone-marrow-derived mesenchymal stem cells
Frontiers in Bioengineering and Biotechnology frontiersin.org13
Ma et al. 10.3389/fbioe.2022.1091360
attenuate early steroid-induced avascular necrosis of femoral head in rabbit. Cell
Biol. Int. 41, 13791390. [PubMed:28877384]. doi:10.1002/cbin.10869
Li, L., Zhang, Y., Mu, J., Chen, J., Zhang, C., Cao, H., et al. (2020). Transplantation
of human mesenchymal stem-cell-derived exosomes immobilized in an adhesive
hydrogel for effective treatment of spinal cord injury. Nano Lett. 20, 42984305.
[PubMed: 4298]. doi:10.1021/acs.nanolett.0c00929
Li, R., Li, D., Wang, H., Chen, K., Wang, S., Xu, J., et al. (2022). Exosomes from
adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to
promote bone healing via miR-451a/MIF. Stem Cell Res. Ther. 13, 149. [PubMed:
35395782]. doi:10.1186/s13287-022-02823-1
Liang, B., Liang, J. M., Ding, J. N., Xu, J., Xu, J. G., and Chai, Y. M. (2019).
Dimethyloxaloylglycine-stimulated human bone marrow mesenchymal stem cell-
derived exosomes enhance bone regeneration through angiogenesis by targeting the
AKT/mTOR pathway. Stem Cell Res. Ther. 10, 335. [PubMed: 31747933]. doi:10.
1186/s13287-019-1410-y
Liang, X., Ding, Y., Zhang, Y., Tse, H. F., and Lian, Q. (2014). Paracrine
mechanisms of mesenchymal stem cell-based therapy: Current status and
perspectives. Cell Transpl. 23, 10451059. [PubMed: 23676629]. doi:10.3727/
096368913x667709
Liang, Y., Duan, L., Lu, J., and Xia, J. (2021). Engineering exosomes for targeted
drug delivery. Theranostics 11, 31833195. [PubMed: 3183]. doi:10.7150/thno.
52570
Liao,Q.,Li,B.J.,Li,Y.,Xiao,Y.,Zeng,H.,Liu,J.M.,etal.(2021).Low-
intensity pulsed ultrasound promotes osteoarthritic cartilage regeneration by
BMSC-derived exosomes via modulating the NF-κB signaling pathway. Int.
Immunopharmacol. 97, 107824. PubMed: 34102487]. doi:10.1016/j.intimp.
2021.107824
Liao, W., Ning, Y., Xu, H. J., Zou, W. Z., Hu, J., Liu, X. Z., et al. (2019). BMSC-
derived exosomes carrying microRNA-122-5p promote proliferation of osteoblasts
in osteonecrosis of the femoral head. Clin. Sci. (Lond) 133, 19551975. [PubMed:
1955]. doi:10.1042/cs20181064
Liebergall, M., Schroeder, J., Mosheiff, R., Gazit, Z., Yoram, Z., Rasooly, L., et al.
(2013). Stem cell-based therapy for prevention of delayed fracture union: A
randomized and prospective preliminary study. Mol. Ther. 21, 16311638.
[PubMed: 23732992]. doi:10.1038/mt.2013.109
Lin, Z., Xiong, Y., Meng, W., Hu, Y., Chen, L., Chen, L., et al. (2022)., 13. PubMed,
300300300311. doi:10.1016/j.bioactmat.2021.10.042Exosomal PD-L1 induces
osteogenic differentiation and promotes fracture healing by acting as an
immunosuppressantBioact. Mater.
Liu, A., Lin, D., Zhao, H., Chen, L., Cai, B., Lin, K., et al. (2021a). Optimized
BMSC-derived osteoinductive exosomes immobilized in hierarchical scaffold via
lyophilization for bone repair through Bmpr2/Acvr2b competitive receptor-
activated Smad pathway. Biomaterials 272, 120718. [PubMed: 33838528]. doi:10.
1016/j.biomaterials.2021.120718
Liu, C. X., and Chen, L. L. (2022). Circular RNAs: Characterization, cellular roles,
and applications. Cell 185, 20162034. [PubMed: 2016]. doi:10.1016/j.cell.2022.
04.021
Liu, L., Guo, S., Shi, W., Liu, Q., Huo, F., Wu, Y., et al. (2021b). Bone marrow
mesenchymal stem cell-derived small extracellular vesicles promote periodontal
regeneration. Tissue Eng. Part A 27, 962976. [PubMed: 962]. doi:10.1089/ten.TEA.
2020.0141
Liu, L., Yu, F., Li, L., Zhou, L., Zhou, T., Xu, Y., et al. (2021c). Bone marrow
stromal cells stimulated by strontium-substituted calcium silicate ceramics: Release
of exosomal miR-146a regulates osteogenesis and angiogenesis. Acta Biomater. 119,
444457. [PubMed: 444]. doi:10.1016/j.actbio.2020.10.038
Liu, P., Xiong, Y., Chen, L., Lin, C., Yang, Y., Lin, Z., et al. (2022). Angiogenesis-
based diabetic skin reconstruction through multifunctional hydrogel with sustained
releasing of M2 Macrophage-derived exosome. Chem. Eng. J. 431, 132413. doi:10.
1016/j.cej.2021.132413
Liu, W., Yu, M., Chen, F., Wang, L., Ye, C., Chen, Q., et al. (2021d). A novel
delivery nanobiotechnology: Engineered miR-181b exosomes improved
osteointegration by regulating macrophage polarization. J. Nanobiotechnol. 19,
269. [PubMed: 34493305]. doi:10.1186/s12951-021-01015-y
Liu, X., Duan, B., Cheng, Z., Jia, X., Mao, L., Fu, H., et al. (2011). SDF-1/
CXCR4 axis modulates bone marrow mesenchymal stem cell apoptosis, migration
and cytokine secretion. Protein Cell 2, 845854. [PubMed: 22058039]. doi:10.1007/
s13238-011-1097-z
Liu, Z., Li, C., Huang, P., Hu, F., Jiang, M., Xu, X., et al. (2020).
CircHmbox1 targeting miRNA-1247-5p is involved in the regulation of bone
metabolism by TNF-αin postmenopausal osteoporosis. Front. Cell Dev. Biol. 8,
594785. [PubMed: 33425899]. doi:10.3389/fcell.2020.594785
López-Noriega, A., Ruiz-Hernández, E., Quinlan, E., Storm, G., Hennink, W. E.,
and OBrien, F. J. (2014). Thermally triggered release of a pro-osteogenic peptide
from a functionalized collagen-based scaffold using thermosensitive liposomes.
J. Control. Release 187, 158166. [PubMed: 158]. doi:10.1016/j.jconrel.2014.05.043
Lu, G. D., Cheng, P., Liu, T., and Wang, Z. (2020). BMSC-de rived exosomal miR-
29a promotes angiogenesis and osteogenesis. Front. Cell Dev. Biol. 8, 608521.
[PubMed: 33363169]. doi:10.3389/fcell.2020.608521
Lu, Z., Chen, Y., Dunstan, C., Roohani-Esfahani, S., and Zreiqat, H. (2017).
Priming adipose stem cells with tumor necrosis factor-alpha preconditioning
potentiates their exosome efcacy for bone regeneration. Tissue Eng. Part A 23,
12121220. [PubMed: 1212]. doi:10.1089/ten.tea.2016.0548
Lv, P. Y., Gao, P. F., Tian, G. J., Yang, Y. Y., Mo, F. F., Wang, Z. H., et al. (2020).
Osteocyte-derived exosomes induced by mechanical strain promote human
periodontal ligament stem cell proliferation and osteogenic differentiation via
the miR-181b-5p/PTEN/AKT signaling pathway. Stem Cell Res. Ther. 11, 295.
[PubMed: 32680565]. doi:10.1186/s13287-020-01815-3
Ma, S., Niu, M., Hao, Z., Liu, M., Tong, C., and Zhao, X. (2021). Selective
packaged circular RNAs in milk extracellular vesicles during Staphylococcus aureus
infection may have potential against bacterial infection. RNA Biol. 18, 818831.
[PubMed: 818]. doi:10.1080/15476286.2020.1853975
Ma, S., Wu, J., Hu, H., Mu, Y., Zhang, L., Zhao, Y., et al. (2022). Novel fusion
peptides deliver exosomes to modify injectable thermo-sensitive hydrogels for bone
regeneration. Mater. Today Bio 13, 100195. [PubMed: 35024598]. doi:10.1016/j.
mtbio.2021.100195
Mao, G., Xu, Y., Long, D., Sun, H., Li, H., Xin, R., et al. (2021). Exosome-
transported circRNA_0001236 enhances chondrogenesis and suppress cartilage
degradation via the miR-3677-3p/Sox9 axis. Stem Cell Res. Ther. 12, 389. [PubMed:
34256841]. doi:10.1186/s13287-021-02431-5
Matos, S., Guerra, F., Krauser, J. T., Figueiredo, H., Marcelino, J. P., and Sanz, M.
(2012). Evaluation of an anorganic bovine-derived mineral with P-15 hydrogel bone
graft: Preliminary study in a rabbit cranial bone model. Clin. Oral Implants Res. 23,
698705. [PubMed: 698]. doi:10.1111/j.1600-0501.2011.02179.x
Mauffrey, C., Barlow, B. T., and Smith, W. (2015). Management of segmental
bone defects. J. Am. Acad. Orthop. Surg. 23, 143153. [PubMed: 25716002]. doi:10.
5435/jaaos-d-14-00018
Meng, W., Hao, Y., He, C., Li, L., and Zhu, G. (2019). Exosome-orchestrated
hypoxic tumor microenvironment. Mol. Cancer 18, 57. [PubMed: 30925935].
doi:10.1186/s12943-019-0982-6
Mi, B., Chen, L., Xiong, Y., Yang, Y., Panayi, A. C., Xue, H., et al. (2022).
Osteoblast/osteoclast and immune cocktail therapy of an exosome/drug delivery
multifunctional hydrogel accelerates fracture repair. ACS Nano 16, 771782.
[PubMed: 771]782. doi:10.1021/acsnano.1c08284
Mo, C., Huang, B., Zhuang, J., Jiang, S., Guo, S., and Mao, X. (2021). LncRNA
nuclear-enriched abundant transcript 1 shuttled by prostate cancer cells-secreted
exosomes initiates osteoblastic phenotypes in the bone metastatic
microenvironment via miR-205-5p/runt-related transcription factor 2/splicing
factor proline- and glutamine-rich/polypyrimidine tract-binding protein 2 axis.
Clin. Transl. Med. 11, e493. [PubMed: 34459124]. doi:10.1002/ctm2.493
Nair, L., Chung, H., and Basu, U. (2020). Regulation of long non-coding RNAs
and genome dynamics by the RNA surveillance machinery. Nat. Rev. Mol. Cell Biol.
21, 123136. [PubMed: 32020081]. doi:10.1038/s41580-019-0209-0
Nan, K., Zhang, Y., Zhang, X., Li, D., Zhao, Y., Jing, Z., et al. (2021). Exosomes
from miRNA-378-modied adipose-derived stem cells prevent glucocorticoid-
induced osteonecrosis of the femoral head by enhancing angiogenesis and
osteogenesis via targeting miR-378 negatively regulated suppressor of fused
(Sufu). Stem Cell Res. Ther. 12, 331. [PubMed: 34099038]. doi:10.1186/s13287-
021-02390-x
Ni, P., Ding, Q., Fan, M., Liao, J., Qian, Z., Luo, J., et al. (2014). Injectable
thermosensitive PEG-PCL-PEG hydrogel/acellular bone matrix composite for bone
regeneration in cranial defects. Biomaterials 35, 236248. [PubMed: 236]. doi:10.
1016/j.biomaterials.2013.10.016
Nielsen, A. F., Bindereif, A., Bozzoni, I., Hanan, M., Hansen, T. B., Irimia, M.,
et al. (2022). Best practice standards for circular RNA research. Nat. Methods 19,
12081220. [PubMed: 35618955]. doi:10.1038/s41592-022-01487-2
Nikhil, A., and Kumar, A. (2022). Evaluating potential of tissue-engineered
cryogels and chondrocyte derived exosomes in articular cartilage repair. Biotech.
Bioeng. 119, 605625. [PubMed: 34723385]. doi:10.1002/bit.27982
Nunes, A., Ribeiro, D. R., Marques, M., Santos, M. A. S., Ribeiro, D., and Soares,
A. R. (2020). Emerging roles of tRNAs in RNA virus infections. Trends Biochem. Sci.
45, 794805. [PubMed: 794]. doi:10.1016/j.tibs.2020.05.007
Ocansey, D. K. W., Zhang, L., Wang, Y., Yan, Y., Qian, H., Zhang, X., et al. (2020).
Exosome-mediated effects and applications in inammatory bowel disease. Biol.
Rev. 95, 12871307. [PubMed: 1287]. doi:10.1111/brv.12608
Patel, G. K., Khan, M. A., Zubair, H., Srivastava, S. K., Khushman, M., Singh, S.,
et al. (2019). Comparative analysis of exosome isolation methods using culture
Frontiers in Bioengineering and Biotechnology frontiersin.org14
Ma et al. 10.3389/fbioe.2022.1091360
supernatant for optimum yield, purity and downstream applications. Sci. Rep. 9 (1),
5335. [PMID: 30926864]. doi:10.1038/s41598-019-41800-2
Pishavar, E., Luo, H., Naserifar, M., Hashemi, M., Toosi, S., Atala, A., et al. (2021).
Advanced hydrogels as exosome delivery systems for osteogenic differentiation of
MSCs: Application in bone regeneration. Int. J. Mol. Sci. 22. [PubMed: 6203]. doi:10.
3390/ijms22126203
Prattichizzo, F., Matacchione, G., Giuliani, A., Sabbatinelli, J., Olivieri, F., de
Candia, P., et al. (2021). Extracellular vesicle-shuttled miRNAs: A critical appraisal
of their potential as nano-diagnostics and nano-therapeutics in type 2 diabetes
mellitus and its cardiovascular complications. Theranostics 11, 10311045.
[PubMed: 1031]. doi:10.7150/thno.51605
Ren, B., Wang, X., Duan, J., and Ma, J. (2019). Rhizobial tRNA-derived small
RNAs are signal molecules regulating plant nodulation. Science 365, 919922.
[PubMed: 919]. doi:10.1126/science.aav8907
Riau, A. K., Ong, H. S., Yam, G. H. F., and Mehta, J. S. (2019). Sustained delivery
system for stem cell-derived exosomes. Front. Pharmacol. 10, 1368. [PubMed:
1368]. doi:10.3389/fphar.2019.01368
Salmena, L., Poliseno, L., Tay, Y., Kats, L., and Pandol, P. P. (2011). A ceRNA
hypothesis: The rosetta stone of a hidden RNA language? Cell 146, 353358.
[PubMed: 353]. doi:10.1016/j.cell.2011.07.014
Sang, X., Zhao, X., Yan, L., Jin, X., Wang, X., Wang, J., et al. (2022).
Thermosensitive hydrogel loaded with primary chondrocyte-derived exosomes
promotes cartilage repair by regulating macrophage polarization in
osteoarthritis. Tissue Eng. Regen. Med. 19, 629642. [PubMed: 35435577].
doi:10.1007/s13770-022-00437-5
Shang,F.,Yu,Y.,Liu,S.,Ming,L.,Zhang,Y.,Zhou,Z.,etal.(2021).
Advancing application of mesenchymal stem cell-based bone tissue
regeneration. Bioact. Mater. 6, 666683. [PubMed: 33005830]. doi:10.1016/j.
bioactmat.2020.08.014
Shao, L. T., Luo, L., Qiu, J. H., and Deng, D. Y. B. (2022). PTH (1-34) enhances the
therapeutic effect of bone marrow mesenchymal stem cell-derived exosomes by
inhibiting proinammatory cytokines expression on OA chondrocyte repair
in vitro.Arthritis Res. Ther. 24, 96. [PubMed: 35488245]. doi:10.1186/s13075-
022-02778-x
Shen, K., Duan, A., Cheng, J., Yuan, T., Zhou, J., Song, H., et al. (2022). Exosomes
derived from hypoxia preconditioned mesenchymal stem cells laden in a silk
hydrogel promote cartilage regeneration via the miR-205-5p/PTEN/AKT
pathway. Acta Biomater. 143, 173188. [PubMed:35202856]. doi:10.1016/j.
actbio.2022.02.026
Shen, Z., Kuang, S., Zhang, Y., Yang, M., Qin, W., Shi, X., et al. (2020). Chitosan
hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates
periodontitis in mice via a macrophage-dependent mechanism. Bioact. Mater. 5,
11131126. [PubMed: 32743122]. doi:10.1016/j.bioactmat.2020.07.002
Silván, U., Díez-Torre, A., Arluzea, J., Andrade, R., Silió, M., and Aréchaga, J.
(2009). Hypoxia and pluripotency in embryonic and embryonal carcinoma stem
cell biology. Differentiation 78, 159168. [PubMed: 159]. doi:10.1016/j.diff.2009.
06.002
Song, H., Chen, X., Hao, Y., Wang, J., Xie, Q., and Wang, X. (2022).
Nanoengineering facilitating the target mission: Targeted extracellular vesicles
delivery systems design. J. Nanobiotechnol. 20, 431. [PubMed: 36175866][.
doi:10.1186/s12951-022-01638-9
Song, H., Li, X., Zhao, Z., Qian, J., Wang, Y., Cui, J., et al. (2019). Reversal of
osteoporotic activity by endothelial cell-secreted bone targeting and biocompatible
exosomes. Nano Lett. 19, 30403048. [PubMed: 3040]. doi:10.1021/acs.nanolett.
9b00287
Sun, J., Yin, Z., Wang, X., and Su, J. (2022). Exosome-laden hydrogels: A novel
cell-free strategy for in-situ bone tissue regeneration. Front. Bioeng. Biotechnol. 10,
866208. [PubMed: 35433664]. doi:10.3389/fbioe.2022.866208
Tan, L., Fu, J., Feng, F., Liu, X., Cui, Z., Li, B., et al. (2020a). Engineered probiotics
biolm enhances osseointegration via immunoregulation and anti-infection. Sci.
Adv. 6. [PubMed: eaba5723]. doi:10.1126/sciadv.aba5723
Tan, S. H. S., Wong, J. R. Y., Sim, S. J. Y., Tjio, C. K. E., Wong, K. L., Chew, J. R. J.,
et al. (2020b). Mesenchymal stem cell exosomes in bone regenerative strategies-a
systematic review of preclinical studies. Mater. Today Bio 7, 100067. [PubMed:
32695985]. doi:10.1016/j.mtbio.2020.100067
Tan, S. S. H., Tjio, C. K. E., Wong, J. R. Y., Wong, K. L., Chew, J. R. J., Hui, J. H. P.,
et al. (2021). Mesenchymal stem cell exosomes for cartilage regeneration: A
systematic review of preclinical in vivo studies. Tissue Eng. Part B Rev. 27, 113.
[PubMed: 1]. doi:10.1089/ten.TEB.2019.0326
Tao, S. C., Huang, J. Y., Gao, Y., Li, Z. X., Wei, Z. Y., Dawes, H., et al. (2021). Small
extracellular vesicles in combination with sleep-related circRNA3503: A targeted
therapeutic agent with injectable thermosensitive hydrogel to prevent osteoarthritis.
Bioact. Mater. 6, 44554469. [PubMed: 4455]. doi:10.1016/j.bioactmat.2021.04.031
Teng, Z., Zhu, Y., Zhang, X., Teng, Y., and Lu, S. (2020). Osteoporosis is
characterized by altered expression of exosomal long non-coding RNAs. Front.
Genet. 11, 566959. [PubMed: 33281871]. doi:10.3389/fgene.2020.566959
Tian, Y., Li, S., Song, J., Ji, T., Zhu, M., Anderson, G. J., et al. (2014). A doxorubicin
delivery platform using engineered natural membrane vesicle exoso mes for targeted
tumor therapy. Biomaterials 35, 23832390. [PubMed: 2383]. doi:10.1016/j.
biomaterials.2013.11.083
Tibbitt, M. W., and Anseth, K. S. (2009). Hydrogels as extracellular matrix mimics
for 3D cell culture. Biotechnol. Bioeng. 103, 655663. [PubMed: 19472329]. doi:10.
1002/bit.22361
Tkach, M., and Thery, C. (2016). Communication by extracellular vesicles: Where
we are and where we need to go. Cell 164, 12261232. [PubMed: 1226]. doi:10.1016/
j.cell.2016.01.043
Toupadakis, C. A., Wong, A., Genetos, D. C., Chung, D. J., Murugesh, D.,
Anderson, M. J., et al. (2012). Long-term administration of AMD3100, an
antagonist of SDF-1/CXCR4 signaling, alters fracture repair. J. Orthop. Res. 30,
18531859. [PubMed: 1853]. doi:10.1002/jor.22145
Valadi, H., Ekström, K., Bossios, A., Sjöstrand, M., Lee, J. J., and Lötvall, J. O.
(2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel
mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654659.
[PubMed: 654]. doi:10.1038/ncb1596
van der Meel, R., Fens, M. H., Vader, P., van Solinge, W. W., Eniola-Adefeso, O.,
and Schiffelers, R. M. (2014). Extracellular vesicles as drug delivery systems: Lessons
from the liposome eld. J. Control. Release 195, 7285. [PubMed: 72]. doi:10.1016/j.
jconrel.2014.07.049
Wang, C., Wang, M., Xu, T., Zhang, X., Lin, C., Gao, W., et al. (2019a).
Engineering bioactive self-healing antibacterial exosomes hydrogel for
promoting chronic diabetic wound healing and complete skin regeneration.
Theranostics 9, 6576. [PubMed: 65]. doi:10.7150/thno.29766
Wang, D., Cao, H., Hua, W., Gao, L., Yuan, Y., Zhou, X., et al. (2022a).
Mesenchymal stem cell-derived extracellular vesicles for bone defect repair.
Membr. (Basel) 12, 716. [PubMed:35877919]. doi:10.3390/membranes12070716
Wang, L., Wang, J., Zhou, X., Sun, J., Zhu, B., Duan, C., et al. (2020). A new self-
healing hydrogel containing hucmsc-derived exosomes promotes bone
regeneration. Front. Bioeng. Biotechnol. 8, 564731. [PubMed: 33042966]. doi:10.
3389/fbioe.2020.564731
Wang, N., Liu, X., Tang, Z., Wei, X., Dong, H., Liu, Y., et al. (2022b). Increased
BMSC exosomal miR-140-3p alleviates bone degradation and promotes bone
restoration by targeting Plxnb1 in diabetic rats. J. Nanobiotechnol. 20, 97.
[PubMed: 35236339]. doi:10.1186/s12951-022-01267-2
Wang, P., Wang, H., Huang, Q., Peng, C., Yao, L., Chen, H., et al. (2019b).
Exosomes from M1-polarized macrophages enhance paclitaxel antitumor activity
by activating macrophages-mediated inammation. Theranostics 9, 17141727.
[PubMed: 1714]. doi:10.7150/thno.30716
Wang, Q. S., Xu, B. X., Fan, K. J., Fan, Y. S., Teng, H., and Wang, T. Y. (2021a).
Dexamethasone-loaded thermo-sensitive hydrogel attenuates osteoarthritis by
protecting cartilage and providing effective pain relief. Ann. Transl. Med. 9,
1120. [PubMed:34430561]. doi:10.21037/atm-21-684
Wang, W., Qiao, S. C., Wu, X. B., Sun, B., Yang, J. G., Li, X., et al. (2021b).
Circ_0008542 in osteoblast exosomes promotes osteoclast-induced bone resorption
through m6A methylation. Cell Death Dis. 12, 628. [PubMed: 34145224]. doi:10.
1038/s41419-021-03915-1
Wang, X., Li, Z., Cui, Y., Cui, X., Chen, C., and Wang, Z. (2021c). Exosomes
isolated from bone marrow mesenchymal stem cells exert a protective effect on
osteoarthritis via lncRNA LYRM4-AS1-GRPR-miR-6515-5p. Front. Cell Dev. Biol.
9, 644380. [PubMed: 34124036]. doi:10.3389/fcell.2021.644380
Wang, Y., Chen, W., Zhao, L., Li, Y., Liu, Z., Gao, H., et al. (2021d). Obesity
regulates miR-467/HoxA10 axis on osteogenic differentiation and fracture healing
by BMSC-derived exosome LncRNA H19. J. Cell. Mol. Med. 25, 17121724.
[PubMed: 1712]. doi:10.1111/jcmm.16273
Wang, Y., He, S. H., Liang, X., Zhang, X. X., Li, S. S., and Li, T. F. (2021e)., 73.
PubMed, 146146146158. doi:10.1002/iub.2414ATF4-modied serum exosomes
derived from osteoarthritic mice inhibit osteoarthritis by inducing
autophagyIUBMB Life
Wang, Y., Liu, J., Ma, J., Sun, T., Zhou, Q., Wang, W., et al. (2019c). Exosomal
circRNAs: Biogenesis, effect and application in human diseases. Mol. Cancer 18,
116. [PubMed: 31277663]. doi:10.1186/s12943-019-1041-z
Wang, Z., Le, H., Wang, Y., Liu, H., Li, Z., Yang, X., et al. (2022c). Instructive
cartilage regeneration modalities with advanced therapeutic implantations under
abnormal conditions. Bioact. Mater. 11, 317338. [PubMed: 34977434]. doi:10.
1016/j.bioactmat.2021.10.002
Wang, Z., Zhao, F., Zhao, Y., Bai, L., and Hang, R. (2022d). Simultaneously
enhanced osteogenesis and angiogenesis via macrophage-derived exosomes upon
Frontiers in Bioengineering and Biotechnology frontiersin.org15
Ma et al. 10.3389/fbioe.2022.1091360
stimulation with titania nanotubes. Biomater. Adv. 134, 112708. [PubMed:
35581093]. doi:10.1016/j.msec.2022.112708
Wei, F., Li, M., Crawford, R., Zhou, Y., and Xiao, Y. (2019). Exosome-integrated
titanium oxide nanotubes for targeted bone regeneration. Acta Biomater. 86,
480492. [PubMed: 480]. doi:10.1016/j.actbio.2019.01.006
Wei, Z., Batagov, A. O., Schinelli, S., Wang, J., Wang, Y., El Fatimy, R., et al.
(2017). Coding and noncoding landscape of extracellular RNA released by human
glioma stem cells. Nat. Commun. 8, 1145. [PubMed: 29074968]. doi:10.1038/
s41467-017-01196-x
Wu, D., Chang, X., Tian, J., Kang, L., Wu, Y., Liu, J., et al. (2021a). Bone
mesenchymal stem cells stimulation by magnetic nanoparticles and a static
magnetic eld: Release of exosomal miR-1260a improves osteogenesis and
angiogenesis. J. Nanobiotechnol. 19, 209. [PubMed: 34256779]. doi:10.1186/
s12951-021-00958-6
Wu, D., Qin, H., Wang, Z., Yu, M., Liu, Z., Peng, H., et al. (2021b). Bone
mesenchymal stem cell-derived sEV-encapsulated thermosensitive hydrogels
accelerate osteogenesis and angiogenesis by release of exosomal miR-21. Front.
Bioeng. Biotechnol. 9, 829136. [PubMed: 35127680]. doi:10.3389/fbioe.2021.829136
Wu, J., Kuang, L., Chen, C., Yang, J., Zeng, W. N., Li, T., et al. (2019). miR-100-5p-
abundant exosomes derived from infrapatellar fat pad MSCs protect articular
cartilage and ameliorate gait abnormalities via inhibition of mTOR in
osteoarthritis. Biomaterials 206, 87100. [PubMed: 87]. doi:10.1016/j.
biomaterials.2019.03.022
Xia, P., Wang, Q., Song, J., Wang, X., Wang, X., Lin, Q., et al. (2022). Low-
intensity pulsed ultrasound enhances the efcacy of bone marrow-derived MSCs in
osteoarthritis cartilage repair by regulating autophagy-mediated exosome release.
Cartilage 13, 194760352210930. [PubMed: 35438034]. doi:10.1177/
19476035221093060
Xie, L., Guan, Z., Zhang, M., Lyu, S., Thuaksuban, N., Kamolmattayakul, S., et al.
(2020). Exosomal circLPAR1 promoted osteogenic differentiation of homotypic
dental pulp stem cells by competitively binding to hsa-miR-31. BioMed Res. Int.
2020, 113. [PubMed:33062690]. doi:10.1155/2020/6319395
Xing, H., Zhang, Z., Mao, Q., Wang, C., Zhou, Y., Zhou, X., et al. (2021). Injectable
exosome-functionalized extracellular matrix hydrogel for metabolism balance and
pyroptosis regulation in intervertebral disc degeneration. J. Nanobiotechnol. 19, 264.
[PubMed:34488795]. doi:10.1186/s12951-021-00991-5
Xing, Y., Yerneni, S. S., Wang, W., Taylor, R. E., Campbell, P. G., and Ren, X.
(2022). Engineering pro-angiogenic biomaterials via chemoselective extracellular
vesicle immobilization. Biomaterials 281, 121357. [PubMed: 34999538]. doi:10.
1016/j.biomaterials.2021.121357
Xu, X. Y., Tian, B. M., Xia, Y., Xia, Y. L., Li, X., Zhou, H., et al. (2020). Exosomes
derived from P2X7 receptor gene-modied cells rescue inammation-
compromised periodontal ligament stem cells from dysfunction. Stem Cells
Transl. Med. 9, 14141430. [PubMed:32597574]. doi:10.1002/sctm.19-0418
Yang, D., Zhang, W., Zhang, H., Zhang, F., Chen, L., Ma, L., et al. (2020a).
Progress, opportunity, and perspective on exosome isolation - efforts for efcient
exosome-based theranostics. Theranostics 1910 (8), 36843707. [PMID: 32206116].
doi:10.7150/thno.41580
Yang, F., Ning, Z., Ma, L., Liu, W., Shao, C., Shu, Y., et al. (2017). Exosomal
miRNAs and miRNA dysregulation in cancer-associated broblasts. Mol. Cancer
16, 148. [PubMed: 28851377]. doi:10.1186/s12943-017-0718-4
Yang, S., Zhu, B., Yin, P., Zhao, L., Wang, Y., Fu, Z., et al. (2020b). Integration of
human umbilical cord mesenchymal stem cells-derived exosomes with
hydroxyapatite-embedded hyaluronic acid-alginate hydrogel for bone
regeneration. ACS Biomater. Sci. Eng. 6, 15901602. [PubMed: 33455380].
doi:10.1021/acsbiomaterials.9b01363
Yang, X., Yang, J., Lei, P., and Wen, T. (2019). LncRNA MALAT1 shuttled by
bone marrow-derived mesenchymal stem cells-secreted exosomes alleviates
osteoporosis through mediating microRNA-34c/SATB2 axis. Aging (Albany NY)
11, 87778791. [PubMed: 8777]. doi:10.18632/aging.102264
Yang, Y., Choi, H., Seon, M., Cho, D., and Bang, S. I. (2016). LL-37 stimulates the
functions of adipose-derived stromal/stem cells via early growth response 1 and the
MAPK pathway. Stem Cell Res. Ther. 7, 58. [PubMed: 27095351]. doi:10.1186/
s13287-016-0313-4
Yang, Z., Shi, J., Xie, J., Wang, Y., Sun, J., Liu, T., et al. (2020c). Large-scale
generation of functional mRNA-encapsulating exosomes via cellular nanoporation.
Nat. Biomed. Eng. 4, 6983. [PubMed: 31844155]. doi:10.1038/s41551-019-0485-1
Yerneni, S. S., Adamik, J., Weiss, L. E., and Campbell, P. G. (2021). Cell trafcking
and regulation of osteoblastogenesis by extracellular vesicle associated bone
morphogenetic protein 2. J. Extracell. Vesicles 10, e12155. [PubMed: 34669267].
doi:10.1002/jev2.12155
Yerneni, S. S., Lathwal, S., Cuthbert, J., Kapil, K., Szczepaniak, G., Jeong, J., et al.
(2022). Controlled release of exosomes using atom transfer radical polymerization-
based hydrogels. Biomacromolecules 23, 17131722. [PubMed: 35302760]. doi:10.
1021/acs.biomac.1c01636
Yerneni, S. S., Lathwal, S., Shrestha, P., Shirwan, H., Matyjaszewski, K., Weiss, L.,
et al. (2019). Rapid on-demand extracellular vesicle augmentation with versatile
oligonucleotide tethers. ACS Nano 13, 1055510565. [PubMed: 10555]. doi:10.
1021/acsnano.9b04651
Yin, K., Zhu, R., Wang, S., and Zhao, R. C. (2017). Low-level laser effect on
proliferation, migration, and antiapoptosis of mesenchymal stem cells. Stem Cells
Dev. 26, 762775. [PubMed: 762]. doi:10.1089/scd.2016.0332
Yu, C., Chen, L., Zhou, W., Hu, L., Xie, X., Lin, Z., et al. (2022a). Injectable
bacteria-sensitive hydrogel promotes repair of infected fractures via sustained
release of miRNA antagonist. ACS Appl. Mat. Interfaces 14, 3442734442.
[PubMed:35866896]. doi:10.1021/acsami.2c08491
Yu, P., Xie, J., Chen, Y., Liu, J., Liu, Y., Bi, B., et al. (2020). A thermo-sensitive
injectable hydroxypropyl chitin hydrogel for sustained salmon calcitonin release
with enhanced osteogenesis and hypocalcemic effects. J. Mat. Chem. B 8, 270281.
[PubMed:31802093]. doi:10.1039/c9tb02049g
Yu, W., Li, S., Guan, X., Zhang, N., Xie, X., Zhang, K., et al. (2022b). Higher yield
and enhanced therapeutic effects of exosomes derived from MSCs in hydrogel-
assisted 3D culture system for bone regeneration. Biomater. Adv. 133, 112646.
[PubMed: 35067433]. doi:10.1016/j.msec.2022.112646
Zeng, Y., Qiu, Y., Jiang, W., Shen, J., Yao, X., He, X., et al. (2022). Biological
features of extracellular vesicles and challenges. Front. Cell Dev. Biol. 10, 816698.
[PMID: 35813192]. doi:10.3389/fcell.2022.816698
Zha, Y., Li, Y., Lin, T., Chen, J., Zhang, S., and Wang, J. (2021). Progenitor cell-
derived exosomes endowed with VEGF plasmids enhance osteogenic induction and
vascular remodeling in large segmental bone defects. Theranostics 11, 397409.
[PubMed: 397]. doi:10.7150/thno.50741
Zhang, B., Huang, J., Liu, J., Lin, F., Ding, Z., and Xu, J. (2021a). Injectable
composite hydrogel promotes osteogenesis and angiogenesis in spinal fusion by
optimizing the bone marrow mesenchymal stem cell microenvironment and
exosomes secretion. Mater. Sci. Eng. C 123, 111782. [PubMed: 33812569].
doi:10.1016/j.msec.2020.111782
Zhang, F. X., Liu, P., Ding, W., Meng, Q. B., Su, D. H., Zhang, Q. C., et al. (2021b).
Injectable Mussel-Inspired highly adhesive hydrogel with exosomes for endogenous
cell recruitment and cartilage defect regeneration. Biomaterials 278, 121169.
[PubMed: 34626937][. doi:10.1016/j.biomaterials.2021.121169
Zhang, J., Liu, X., Li, H., Chen, C., Hu, B., Niu, X., et al. (2016). Exosomes/
tricalcium phosphate combination scaffolds can enhance bone regeneration by
activating the PI3K/Akt signaling pathway. Stem Cell Res. Ther. 7, 136. [PubMed:
27650895]. doi:10.1186/s13287-016-0391-3
Zhang, J., Tong, D., Song, H., Ruan, R., Sun, Y., Lin, Y., et al. (2022).
Osteoimmunity-regulating biomimetically hierarchical scaffold for augmented
bone regeneration. Adv. Mater. 34, e2202044. [PubMed: 35785450]. doi:10.1002/
adma.202202044
Zhang, L., and Yu, D. (2019). Exosomes in cancer development, metastasis, and
immunity. Biochimica Biophysica Acta - Rev. Cancer 1871, 455468. [PubMed:
455]. doi:10.1016/j.bbcan.2019.04.004
Zhang, L., Zhang, S., Yao, J., Lowery, F. J., Zhang, Q., Huang, W. C., et al. (2015).
Microenvironment-induced PTEN loss by exosomal microRNA primes brain
metastasis outgrowth. Nature 527, 100104. [PubMed: 100]. doi:10.1038/
nature15376
Zhang, S., Teo, K. Y. W., Chuah, S. J., Lai, R. C., Lim, S. K., and Toh, W. S. (2019).
MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating
inammation and restoring matrix homeostasis. Biomaterials 200, 3547.
[PubMed: 35]. doi:10.1016/j.biomaterials.2019.02.006
Zhang, X., Trebak, F., Souza, L. A. C., Shi, J., Zhou, T., Kehoe, P. G., et al. (2020).
Small RNA modications in Alzheimers disease. Neurobiol. Dis. 145, 105058.
[PubMed:32835860]. doi:10.1016/j.nbd.2020.105058
Zhang, Y., Kim, M. S., Jia, B., Yan, J., Zuniga-Hertz, J. P., Han, C., et al. (2017).
Hypothalamic stem cells control ageing speed partly through exosomal miRNAs.
Nature 548, 5257. [PubMed: 28746310]. doi:10.1038/nature23282
Zhang, Y., Xie, Y., Hao, Z., Zhou, P., Wang, P., Fang, S., et al. (2021c). Umbilical
mesenchymal stem cell-derived exosome-encapsulated hydrogels accelerate bone
repair by enhancing angiogenesis. ACS Appl. Mat. Interfaces 13, 1847218487.
[PubMed:33856781]. doi:10.1021/acsami.0c22671
Zhang, Y., Zhang, X., Shi, J., Tuorto, F., Li, X., Liu, Y., et al. (2018a).
Dnmt2 mediates intergenerational transmission of paternally acquired metabolic
disorders through sperm small non-coding RNAs. Nat. Cell Biol. 20, 535540.
[PubMed: 29695786]. doi:10.1038/s41556-018-0087-2
Zhang, Z., Wang, X., Wang, Y., and Hao, J. (2018b). Rapid-forming and self-
healing agarose-based hydrogels for tissue adhesives and potential wound dressings.
Biomacromolecules 19, 980988. [PubMed: 980]. doi:10.1021/acs.biomac.7b01764
Frontiers in Bioengineering and Biotechnology frontiersin.org16
Ma et al. 10.3389/fbioe.2022.1091360
Zhao, D., Zhu, T., Li, J., Cui, L., Zhang, Z., Zhuang, X., et al. (2021a). Poly(lactic-
co-glycolic acid)-based composite bone-substitute materials. Bioact. Mater. 6,
346360. [PubMed: 346]. doi:10.1016/j.bioactmat.2020.08.016
Zhao, J., Li, X., Hu, J., Chen, F., Qiao, S., Sun, X., et al. (2019). Mesenchymal
stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury
through miR-182-regulated macrophage polarization. Cardiovasc Res. 115,
12051216. [PubMed:30753344]. doi:10.1093/cvr/cvz040
Zhao, W., Hu, J., and He, Q. (2021b). The effect of the WKYMVm peptide on
promoting mBMSC secretion of exosomes to induce M2 macrophage polarization
through the FPR2 pathway. J. Orthop. Surg. Res. 16, 171. [PubMed: 33658070].
doi:10.1186/s13018-021-02321-9
Zhao, Y., Chen, Y., Wang, Z., Xu, C., Qiao, S., Liu, T., et al. (2022). Bone
marrow mesenchymal stem cell exosome attenuates inammasome-related
pyroptosis via delivering circ_003564 to improve the recovery of spinal cord
injury. Mol. Neurobiol. 59, 67716789. [PubMed: 36038697]. doi:10.1007/
s12035-022-03006-y
Zhi, F., Ding, Y., Wang, R., Yang, Y., Luo, K., and Hua, F. (2021). Exosomal
hsa_circ_0006859 is a potential biomarker for postmenopausal osteoporosis and
enhances adipogenic versus osteogenic differentiation in human bone marrow
mesenchymal stem cells by sponging miR-431-5p. Stem Cell Res. Ther. 12, 157.
[PubMed: 33648601]. doi:10.1186/s13287-021-02214-y
Zhou, W., Zhou, Y., Chen, X., Ning, T., Chen, H., Guo, Q., et al. (2021). Pancreatic
cancer-targeting exosomes for enhancing immunotherapy and reprogramming
tumor microenvironment. Biomaterials 268, 120546. [PubMed: 33253966].
doi:10.1016/j.biomaterials.2020.120546
Zhu, L., Li, J., Gong, Y., Wu, Q., Tan, S., Sun, D., et al. (2019). Exosomal tRNA-
derived small RNA as a promising biomarker for cancer diagnosis. Mol. Cancer 18,
74. [PubMed: 30940133]. doi:10.1186/s12943-019-1000-8
Zhu, L., Liu, X., Pu, W., and Peng, Y. (2018). tRNA-derived small non-coding
RNAs in human disease. Cancer Lett. 419, 17. [PubMed: 1]. doi:10.1016/j.canlet.
2018.01.015
Zhu, L., Sun, H. T., Wang, S., Huang, S. L., Zheng, Y., Wang, C. Q., et al. (2020).
Isolation and characterization of exosomes for cancer research. J. Hematol. Oncol.
13, 152. [PubMed: 33168028]. doi:10.1186/s13045-020-00987-y
Zhu, T., Jiang, M., Zhang, M., Cui, L., Yang, X., Wang, X., et al. (2022a).
Biofunctionalized composite scaffold to potentiate osteoconduction,
angiogenesis, and favorable metabolic microenvironment for osteonecrosis
therapy. Bioact. Mater. 9, 446460. [PubMed: 34820582]. doi:10.1016/j.
bioactmat.2021.08.005
Zhu, T., Jiang, M., Zhang, M., Cui, L., Yang, X., Wang, X., et al. (2022b).
Construction and validation of steroid-induced rabbit osteonecrosis model.
MethodsX 9, 101713. [PubMed: 35601954]. doi:10.1016/j.mex.2022.101713
Zoulikha, M., Xiao, Q., Boafo, G. F., Sallam, M. A., Chen, Z., and He, W. (2022).
Pulmonary delivery of siRNA against acute lung injury/acute respiratory distress
syndrome. Acta Pharm. Sin. B 12, 600620. [PubMed: 600]. doi:10.1016/j.apsb.
2021.08.009
Frontiers in Bioengineering and Biotechnology frontiersin.org17
Ma et al. 10.3389/fbioe.2022.1091360
... Endogenous and exogenous strategies have demonstrated effectiveness in selectively loading EVs, increasing payload, and improving homing efficiency to bone defects [128][129][130][131]. Endogenous engineering is the enhancement of parent cells to produce desired EV phenotypes, while exogenous engineering is a direct manipulation of EVs. Engineering techniques to optimize EV delivery of growth factors include gene transduction, electroporation, sonication, preconditioning, surface modification, and mechanical manipulation [132][133][134]. Despite ongoing improvements in selective cargo loading, significant variability in preclinical dosing paradigms hinders clinical translation [135]. ...
Article
Full-text available
The management of critical-sized bone defects caused by nonunion, trauma, infection, malignancy, pseudoarthrosis, and osteolysis poses complex reconstruction challenges for orthopedic surgeons. Current treatment modalities, including autograft, allograft, and distraction osteogenesis, are insufficient for the diverse range of pathology encountered in clinical practice, with significant complications associated with each. Therefore, there is significant interest in the development of delivery vehicles for growth factors to aid in bone repair in these settings. This article reviews innovative strategies for the management of critical-sized bone loss, including novel scaffolds designed for controlled release of rhBMP, bioengineered extracellular vesicles for delivery of intracellular signaling molecules, and advances in regional gene therapy for sustained signaling strategies. Improvement in the delivery of growth factors to areas of significant bone loss has the potential to revolutionize current treatment for this complex clinical challenge.
Article
Full-text available
Canine mesenchymal stromal cells (MSCs) possess the capacity to differentiate into a variety of cell types and secrete a wide range of bioactive molecules in the form of soluble and membrane-bound exosomes. Extracellular vesicles/exosomes are nano-sized vesicles that carry proteins, lipids, and nucleic acids and can modulate recipient cell response in various ways. The process of exosome formation is a physiological interaction between cells. With a significant increase in basic research over the last two decades, there has been a tremendous expansion in research in MSC exosomes and their potential applications in canine disease models. The characterization of exosomes has demonstrated considerable variations in terms of source, culture conditions of MSCs, and the inclusion of fetal bovine serum or platelet lysate in the cell cultures. Furthermore, the amalgamation of exosomes with various nano-materials has become a novel approach to the fabrication of nano-exosomes. The fabrication of exosomes necessitates the elimination of extrinsic proteins, thus enhancing their potential therapeutic uses in a variety of disease models, including spinal cord injury, osteoarthritis, and inflammatory bowel disease. This review summarizes current knowledge on the characteristics, biological functions, and clinical relevance of canine MSC exosomes and their potential use in human and canine research. As discussed, exosomes have the ability to control lethal vertebrate diseases by administration directly at the injury site or through specific drug delivery mechanisms.
Article
Full-text available
Extracellular vesicles (EVs) can deliver various bioactive molecules among cells, making them promising diagnostic and therapeutic alternatives in diseases. Mesenchymal stem cell-derived EVs (MSC-EVs) have shown therapeutic potential similar to MSCs but with drawbacks such as lower yield, reduced biological activities, off-target effects, and shorter half-lives. Improving strategies utilizing biotechniques to pretreat MSCs and enhance the properties of released EVs, as well as modifying MSC-EVs to enhance targeting abilities and achieve controlled release, shows potential for overcoming application limitations and enhancing therapeutic effects in treating bone-related diseases. This review focuses on recent advances in functionalizing MSC-EVs to treat bone-related diseases. Firstly, we underscore the significance of MSC-EVs in facilitating crosstalk between cells within the skeletal environment. Secondly, we highlight strategies of functional-modified EVs for treating bone-related diseases. We explore the pretreatment of stem cells using various biotechniques to enhance the properties of resulting EVs, as well as diverse approaches to modify MSC-EVs for targeted delivery and controlled release. Finally, we address the challenges and opportunities for further research on MSC-EVs in bone-related diseases.
Article
BACKGROUND Mesenchymal stem cells (MSCs) modulated by various exogenous signals have been applied extensively in regenerative medicine research. Notably, nanosecond pulsed electric fields (nsPEFs), characterized by short duration and high strength, significantly influence cell phenotypes and regulate MSCs differentiation via multiple pathways. Consequently, we used transcriptomics to study changes in messenger RNA (mRNA), long noncoding RNA (lncRNA), microRNA (miRNA), and circular RNA expression during nsPEFs application. AIM To explore gene expression profiles and potential transcriptional regulatory mechanisms in MSCs pretreated with nsPEFs. METHODS The impact of nsPEFs on the MSCs transcriptome was investigated through whole transcriptome sequencing. MSCs were pretreated with 5-pulse nsPEFs (100 ns at 10 kV/cm, 1 Hz), followed by total RNA isolation. Each transcript was normalized by fragments per kilobase per million. Fold change and difference significance were applied to screen the differentially expressed genes (DEGs). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed to elucidate gene functions, complemented by quantitative polymerase chain reaction verification. RESULTS In total, 263 DEGs were discovered, with 92 upregulated and 171 downregulated. DEGs were predominantly enriched in epithelial cell proliferation, osteoblast differentiation, mesenchymal cell differentiation, nuclear division, and wound healing. Regarding cellular components, DEGs are primarily involved in condensed chromosome, chromosomal region, actin cytoskeleton, and kinetochore. From aspect of molecular functions, DEGs are mainly involved in glycosaminoglycan binding, integrin binding, nuclear steroid receptor activity, cytoskeletal motor activity, and steroid binding. Quantitative real-time polymerase chain reaction confirmed targeted transcript regulation. CONCLUSION Our systematic investigation of the wide-ranging transcriptional pattern modulated by nsPEFs revealed the differential expression of 263 mRNAs, 2 miRNAs, and 65 lncRNAs. Our study demonstrates that nsPEFs may affect stem cells through several signaling pathways, which are involved in vesicular transport, calcium ion transport, cytoskeleton, and cell differentiation.
Article
Exosomes, nanoparticles secreted by various cells, composed of a bilayer lipid membrane, and containing bioactive substances such as proteins, nucleic acids, metabolites, etc., have been intensively investigated in tissue engineering owing to their high biocompatibility and versatile biofunction. However, there is still a lack of a high‐quality review on bone defect regeneration potentiated by exosomes. In this review, we will first introduce the biogenesis and isolation methods of exosomes, More importantly, the engineered exosomes of the current state of knowledge will be discussed intensively in this review. Afterward, the biomaterial carriers of exosomes and the mechanisms of bone repair elucidated by compelling evidence will be presented. Thus, future perspectives and concerns are revealed to help devise advanced modalities based on exosomes to overcome the challenges of bone regeneration. We totally believe this review will attract special attention from clinicians and provide promising ideas for their future works. This article is protected by copyright. All rights reserved
Article
Bone defects are often difficult to treat due to their complexity and specificity, and therefore pose a serious threat to human life and health. Currently, the clinical treatment of bone defects is mainly surgical. However, this treatment is often more harmful to patients and there is a potential risk of rejection and infection. Hydrogels have a unique three-dimensional structure that can accommodate a variety of materials, including particles, polymers and small molecules, making them ideal for treating bone defects. Therefore, emerging composite hydrogels are considered one of the most promising candidates for the treatment of bone defects. This review describes the use of different types of composite hydrogel in the treatment of bone defects. We present the basic concepts of hydrogels, different preparation techniques (including chemical and physical crosslinking), and the clinical requirements for hydrogels used to treat bone defects. In addition, a review of numerous promising designs of different types of hydrogel doped with different materials (e.g., nanoparticles, polymers, carbon materials, drugs, and active factors) is also highlighted. Finally, the current challenges and prospects of composite hydrogels for the treatment of bone defects are presented. This review will stimulate research efforts in this field and promote the application of new methods and innovative ideas in the clinical field of composite hydrogels.
Article
Full-text available
Precision medicine has put forward the proposition of "precision targeting" for modern drug delivery systems. Inspired by techniques from biology, pharmaceutical sciences, and nanoengineering, numerous targeted drug delivery systems have been developed in recent decades. But the large-scale applications of these systems are limited due to unsatisfactory targeting efficiency, cytotoxicity, easy removability, and instability. As such, the natural endogenous cargo delivery vehicle—extracellular vesicles (EVs)—have sparked significant interest for its unique inherent targeting properties, biocompatibility, transmembrane ability, and circulatory stability. The membranes of EVs are enriched for receptors or ligands that interact with target cells, which endows them with inherent targeting mission. However, most of the natural therapeutic EVs face the fate of being cleared by macrophages, resulting in off-target. Therefore, the specificity of natural EVs delivery systems urgently needs to be further improved. In this review, we comprehensively summarize the inherent homing mechanisms of EVs and the effects of the donor cell source and administration route on targeting specificity. We then go over nanoengineering techniques that modify EVs for improving specific targeting, such as source cell alteration and modification of EVs surface. We also highlight the auxiliary strategies to enhance specificity by changing the external environment, such as magnetic and photothermal. Furthermore, contemporary issues such as the lack of a gold standard for assessing targeting efficiency are discussed. This review will provide new insights into the development of precision medicine delivery systems. Graphical Abstract
Article
Full-text available
Bone marrow mesenchymal stem cell (BMSC) is previously reported to present a certain effect on treating spinal cord injury (SCI), while the underlying mechanism is largely uncovered. Therefore, the current study aimed to investigate the involvement of exosome-delivered circRNA profile in the BMSC’s effect on pyroptosis for SCI treatment. H2O2 treated rat primary neurons were cultured with normal medium, BMSC, BMSC plus GW4869, and BMSC-derived exosome, respectively, then inflammasome-related pyroptosis markers, and circRNA profiles were detected. Subsequently, circ_003564-knockdown BMSC exosome was transfected into H2O2 treated rat primary neurons and NGF-stimulated PC-12 cells. Furthermore, in vivo validation was conducted. BMSC and BMSC-derived exosome both decreased inflammasome-related pyroptosis markers including cleaved caspase-1, GSDMD, NLRP3, IL-1β, and IL-18 in H2O2-treated neurons, while exosome-free BMSC (BMSC plus GW4869) did not obviously reduce these factors. Microarray assay revealed that BMSC (vs. exosome-free BMSC) and BMSC-derived exosome (vs. normal medium) greatly regulated circRNA profiles, which were enriched in neuroinflammation pathways (such as neurotrophin, apoptosis, and TNF). Among three functional candidate circRNAs (circ_015525, circ_008876, and circ_003564), circ_003564 was most effective to regulate inflammasome-related pyroptosis. Interestingly, circ_003564-knockdown BMSC exosome showed higher expression of inflammasome-related pyroptosis markers compared to negative-control-knockdown BMSC exosome in H2O2 treated primary neurons/NGF-stimulated PC-12 cells. In vivo, BMSC exosome improved the function recovery and decreased tissue injury and inflammasome-related pyroptosis in SCI rats, whose effect was attenuated by circ_003564 knockdown transfection. BMSC exosome attenuates inflammasome-related pyroptosis via delivering circ_003564, contributing to its treatment efficacy for SCI.
Article
Full-text available
Objectives: The treatment of bone defects by stem cells (MSCs) has achieved limited success over the recent few decades. The emergence of exosomes provides a new strategy for bone regeneration. Here, we aimed to investigate the effect and mechanisms of exosomes combined with dental pulp stem cells (DPSCs) on bone regeneration. Materials and methods: We isolated exosomes from stem cells from human exfoliated deciduous teeth (SHED) aggregates and evaluated the efficacy of exosomes combined with DPSCs in a cranial bone defect model. The potential mechanisms were further investigated. Results: The effect of exosomes combined with DPSCs was remarkable on bone regeneration in vivo and exosomes promoted osteogenic differentiation of DPSCs in vitro. Mechanistically, exosomes increased the expression of mitochondrial transcription factor A (TFAM) in DPSCs by transferring TFAM mRNA. Moreover, highly expressed TFAM in DPSCs enhanced glutamate metabolism and oxidative phosphorylation (OXPHOS) activity. Conclusions: Consequently, exosomes strengthened bone regeneration of DPSCs through the activation of mitochondrial aerobic metabolism. Our study provides a new potential strategy to improve DPSC-based bone regenerative treatment.
Article
Full-text available
Objectives Idiopathic short stature (ISS), a disorder of unknown cause, accounts for approximately 80% of the clinical diagnoses of children with short stature. Exosomal circular RNA in plasma has been implicated in various disease processes. However, the role of exosome-derived circRNA in ISS has not been elucidated yet. Methods Plasma exosomes of ISS and normal children were cocultured with human chondrocytes. Microarray analysis and RT-PCR identified the differential expression of circRNA in exosomes between ISS and normal children. Hsa_circ_0063476 was upregulated or downregulated in human chondrocytes. Subsequently, overexpression rats of hsa_circ_0063476 was constructed via adenovirial vector to further validate the role of hsa_circ_0063476 on longitudinal bone growth via in vivo experiment. Results The plasma exosome of ISS children suppressed the expression of markers of chondrocyte hypertrophy and endochondral ossification. Subsequently, upregulation of hsa_circ_0063476 in ISS exosome was identified. In vitro experiment demonstrated that chondrocyte proliferation, cell cycle and endochondral ossification were suppressed, and apoptosis was increased following hsa_circ_0063476 overexpression in human chondrocyte. Conversely, silencing hsa_circ_0063476 in human chondrocyte can show opposite outcomes. Our study further revealed hsa_circ_0063476 overexpression in vitro can enhance chondrocyte apoptosis and inhibit the expression of markers of chondrocyte proliferation and endochondral ossification via miR-518c-3p/DDX6 axis. Additionally, the rats of hsa_circ_0063476 overexpression showed a short stature phenotype. Conclusions The authors identified a novel pathogenesis in ISS that exosome-derived hsa_circ_ 0063476 retards the expression of markers of endochondral ossification and impairs longitudinal bone growth via miR-518c-3p/DDX6 axis, which may provide a unique therapeutic avenue for ISS.
Article
Full-text available
Fracture nonunion can result in considerable physical harm and limitation of quality of life in patients, exerting an extensive economic burden to the society. Nonunion largely results from unresolved inflammation and impaired osteogenesis. Despite advancements in surgical techniques, the indispensable treatment for nonunion is robust anti-inflammation therapy and the promotion of osteogenic differentiation. Herein, we report that plasma exosomes derived from infected fracture nonunion patients (Non-Exos) delayed fracture repair in mice by inhibiting the osteogenic differentiation of bone marrow stromal cells in vivo and in vitro. Unique molecular identifier microRNA-sequencing (UID miRNA-seq) suggested that microRNA-708-5p (miR-708-5p) was overexpressed in Non-Exos. Mechanistically, miR-708-5p targeted structure-specific recognition protein 1, thereby suppressing the Wnt/β-catenin signaling pathway, which, in turn, impaired osteogenic differentiation. AntagomicroRNA-708-5p (antagomiR-708-5p) could partly reverse the above process. A bacteria-sensitive natural polymer hyaluronic-acid-based hydrogel (HA hydrogel) loaded with antagomiR-708-5p exhibited promising effects in an in vivo study through antibacterial and pro-osteogenic differentiation functions in infected fractures. Overall, the effectiveness and reliability of an injectable bacteria-sensitive hydrogel with sustained release of agents represent a promising approach for infected fractures.
Article
Full-text available
The repair of critical bone defects is a hotspot of orthopedic research. With the development of bone tissue engineering (BTE), there is increasing evidence showing that the combined application of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) (MSC-EVs), especially exosomes, with hydrogels, scaffolds, and other bioactive materials has made great progress, exhibiting a good potential for bone regeneration. Recent studies have found that miRNAs, proteins, and other cargo loaded in EVs are key factors in promoting osteogenesis and angiogenesis. In BTE, the expression profile of the intrinsic cargo of EVs can be changed by modifying the gene expression of MSCs to obtain EVs with enhanced osteogenic activity and ultimately enhance the osteoinductive ability of bone graft materials. However, the current research on MSC-EVs for repairing bone defects is still in its infancy, and the underlying mechanism remains unclear. Therefore, in this review, the effect of bioactive materials such as hydrogels and scaffolds combined with MSC-EVs in repairing bone defects is summarized, and the mechanism of MSC-EVs promoting bone defect repair by delivering active molecules such as internal miRNAs is further elucidated, which provides a theoretical basis and reference for the clinical application of MSC-EVs in repairing bone defects.
Article
Full-text available
Engineering a proper immune response following biomaterial implantation is essential to bone tissue regeneration. Herein, a biomimetically hierarchical scaffold composed of deferoxamine@poly(ε‐caprolactone) (DFO@PCL) nanoparticle, manganese carbonyl (MnCO) nanosheet, gelatin methacryloyl hydrogel, and polylactide/hydroxyapatite (PLA/HA) matrix was fabricated to augment bone repair by facilitating the balance of immune system and bone metabolism. First, a 3D printed stiff scaffold with a well‐organized gradient structure mimicked the cortical and cancellous bone tissues; meanwhile, an inside infusion of a soft hydrogel further endowed the scaffold characteristics of extracellular matrices. A Fenton‐like reaction between MnCO and endogenous hydrogen peroxide generated at implant−tissue site triggered continuous release of carbon monoxide and Mn2+, thus significantly lessening inflammatory response by up‐regulating M2 phenotype of macrophages, which also secreted vascular endothelial growth factor to induce vascular formation. Through activating the hypoxia‐inducible factor‐1α pathway, Mn2+ and DFO@PCL further promoted angiogenesis. Moreover, DFO inhibited osteoclast differentiation and synergistically collaborated with the osteoinductive activity of HA. Based on amounts of in vitro and in vivo data, strong immunomodulatory, intensive angiogenic, weak osteoclastogenic, and superior osteogenic abilities of such osteoimmunity‐regulating scaffold presented a profound effect on improving bone regeneration, which puts forward a worthy base and positive enlightenment for large‐scale bone defect repair. This article is protected by copyright. All rights reserved
Article
Full-text available
Extracellular vesicles (EVs) are vesicles with a lipid bilayer membrane on the outside, which are widely found in various body fluids and contain biological macromolecules such as DNA, RNA, lipids and proteins on the inside. EVs were once thought to be vesicles for the removal of waste materials, but are now known to be involved in a variety of pathophysiological processes in many diseases. This study examines the advantage of EVs and the challenges associated with their application. A more rational use of the advantageous properties of EVs such as composition specificity, specific targeting, circulatory stability, active penetration of biological barriers, high efficient drug delivery vehicles and anticancer vaccines, oxidative phosphorylation activity and enzymatic activity, and the resolution of shortcomings such as isolation and purification methods, storage conditions and pharmacokinetics and biodistribution patterns during drug delivery will facilitate the clinical application of EVs.
Article
Purpose: To seek out the bone regeneration effect of human umbilical cord-mesenchymal stem cell (hUC-MSC)-derived exosomes of loaded chitosan/hydroxyapatite (CS/HA) scaffold in a rat calvarium bone regeneration model. Materials and methods: The hUC-MSC exosomes were purified and characterized. The scaffolds were prepared by a freeze-drying method. Animals were divided into five groups, and the CS/HA/exosome (CS/HA/Exo) scaffolds were transplanted to 5 × 2-mm critical-sized calvarial bone defects for repair in rats. All animals were sacrificed at the postoperative sixth week. Immunohistochemical and histologic analyses were performed. Results: Scanning electron microscopy (SEM) images showed that the exosomes were round-shaped vesicles with bounded membrane, and the diameter of the exosomes was 83.728 ± 27.269 nm. Histologic analysis showed that mean new bone volumes were statistically significantly higher in the CS/HA/Exo group (1.83 ± 0.54, PCS/Exo-CS/HA/Exo = .000), and other new bone volumes in the other groups were statistically significant compared with the control (CS/Exo 1.50 ± 0.14 mm3; CS 1.20 ± 0.43 mm3; control 1.06 ± 0.10 mm3; and CS/HA 1.43 ± 0.66 mm3). Conclusion: The CS/HA/Exo combination is a novel treatment for bone defect repair to induce bone formation. The CS scaffold can significantly promote bone regeneration compared with the control. Moreover, the combination with HA and exosomes is promising for applications in bone tissue regeneration.
Article
Exosomes derived from mesenchymal stem cells (MSCs) have been proven to exhibit great potentials in spinal cord injury (SCI) therapy. However, conventional two-dimensional (2D) culture will inevitably lead to the loss of stemness of MSCs, which substantially limits the therapeutic potency of MSCs exosomes (2D-Exo). Exosomes derived from three-dimensional culture (3D-Exo) possess higher therapeutic efficiency which have wide applications in spinal cord therapy. Typically, conventional exosome therapy that relies on local repeated injection results in secondary injury and low efficiency. It is urgent to develop a more reliable, convenient, and effective exosome delivery method to achieve constant in situ exosomes release. Herein, we proposed a controlled 3D-exohydrogel hybrid microneedle array patch to achieve SCI repair in situ. Our studies suggested that MSCs with 3D-culturing could maintain their stemness, and consequently, 3D-Exo effectively reduced SCI-induced inflammation and glial scarring. Thus, it is a promising therapeutic strategy for the treatment of SCI.