ArticlePDF AvailableLiterature Review

Mesenchymal Stem Cells-Derived Exosomes: A Possible Therapeutic Strategy for Osteoporosis

Authors:
  • The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine

Abstract and Figures

Osteoporosis is a common age-related disorder characterized by low bone mass and deterioration in bone microarchitecture, leading to increased skeletal fragility and fracture risk. The pathophysiology of osteoporosis is multifactorial. It is related to the imbalance between osteoblasts and osteoclasts; reduced bone mass and increased adipogenesis in the bone marrow. Moreover, angiogenesis, inflammatory process and miRNAs have shown effects in the formation of osteoporosis. In the recent years, mesenchymal stem cells (MSCs) have been regarded as an excellent choice for cell-based tissue engineering therapy of osteoporosis. Growing evidence showed that, paracrine effect have been considered as the predominant mechanism for the role of MSCs in tissue repair. Recently, many studies have proposed that MSCs-derived exosomes are effective for a variety of diseases like cancer, cardiovascular diseases, etc. However, whether the MSCs-derived exosomes could serve as a novel therapeutic tool for osteoporosis has not clearly described. In this review, we summarize the MSCs-derived exosomes and the relationship with osteogenesis, osteoclast differentiation, angiogenesis, immune processes and miRNAs. Finally, we suggest that MSCs-derived exosomes might be a promising therapeutic method for osteoporosis in the future.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.ae
Current Stem Cell Research & Therapy, 2018, 13, 000-000 1
REVIEW ARTICLE
1574-888X/18 $58.00+.00 © 2018 Bentham Science Publishers
Mesenchymal Stem Cells-Derived Exosomes: A Possible Therapeutic
Strategy for Osteoporosis
Yue Li1, Daxiang Jin2,3,*, Weixing Xie2, Longfei Wen1, Weijian Chen1, Jixi Xu2, Jinyong Ding2 ,
Dongcheng Ren1 and Zenglin Xiao1
1Guangzhou University of Chinese Medicine, Guangzhou, China; 2The First Affiliated Hospital of Guangzhou Univer-
sity of Chinese Medicine, Guangzhou, China; 3Laboratory Affiliated to National Key Discipline of Orthopaedic and
Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
A R T I C L E H I S T O R Y
Received: October 28, 2017
Revised: March 24, 2017
Accepted: March 30, 2017
DOI:
10.2174/1574888X13666180403163456
Abstract: Osteoporosis is a common age-related disorder characterized by low bone mass and deterio-
ration in bone microarchitecture, leading to increased skeletal fragility and fracture risk. The patho-
physiology of osteoporosis is multifactorial. It is related to the imbalance between osteoblasts and
osteoclasts; reduced bone mass and increased adipogenesis in the bone marrow. Moreover, angiogene-
sis, inflammatory process and miRNAs have shown effects in the formation of osteoporosis. In the
recent years, mesenchymal stem cells (MSCs) have been regarded as an excellent choice for cell-based
tissue engineering therapy of osteoporosis. Growing evidence showed that paracrine effect has been
considered as the predominant mechanism for the role of MSCs in tissue repair. Recently, many stud-
ies have proposed that MSCs-derived exosomes are effective for a variety of diseases like cancer, car-
diovascular diseases, etc. However, whether the MSCs-derived exosomes could serve as a novel thera-
peutic tool for osteoporosis has not clearly described. In this review, we summarize the MSCs-derived
exosomes and the relationship with osteogenesis, osteoclast differentiation, angiogenesis, immune
processes and miRNAs. Finally, we suggest that MSCs-derived exosomes might be a promising thera-
peutic method for osteoporosis in the future.
Keywords: MSCs, exosomes, osteoporosis, therapeutic.
1. INTRODUCTION
Osteoporosis is a common disease characterized by a sys-
temic impairment of bone mass and microarchitecture, re-
sults in increasing the propensity of fragility fractures [1].
However, the etiology of osteoporosis is complex and not
completely known so far.
The pathogenesis of osteoporosis-related to the improper
balance between the activities of osteoblasts and osteoclasts
[2]. In addition, since osteoblasts and adipocytes share a
common precursor, during the process of aging, the cell line-
age commitment of MSCs shifts to adipocyte in bone mar-
row, resulting in osteoporosis [3]. Osteoporosis and angio-
genesis are also intimately related. Accumulating evidence
from studies in animals, cells and patients suggest that the
local blood supply or decreased angiogenesis contributes to
the process of osteoporosis [4]. Inflammation is a defensive
mechanism for pathogen clearance and maintaining tissue
homeostasis. In the skeletal system, inflammation is closely
*Address correspondence to this author at the Laboratory Affiliated to Na-
tional Key Discipline of Orthopaedic and Traumatology of Chinese Medi-
cine, Guangzhou University of Chinese Medicine, Guangzhou, China; E-
mail: 18620260809@163.com
related to many bone disorders including osteoporosis [5].
MiRNAs are a class of short non-coding RNA molecules that
regulate gene expression by targeting mRNAs [6]. They are
involved in various cellular and molecular activities and
played important roles in lots of biological and pathological
processes [7]. Recently, miRNAs have been proved to play
crucial roles in the etiology of various diseases such as os-
teoporosis [8].
MSCs have been proposed as promising candidates for a
variety of therapeutic applications, such as bone, and carti-
lage regeneration, acute renal failure and neurological dis-
eases, etc. [9]. Recently, it has been identified that MSCs
exert their therapeutical activity through a paracrine effect,
mediated by the release of small particles like growth factors
[10]. Extracellular vesicles (EVs) released from MSCs are
involved in tissue regeneration and contribute to the
paracrine effect of MSCs [11].
EVs are membrane-packed vesicles and come in several
different vesicular formats [12]. Mammalian cells can release
different types of EVs. The main types include apoptotic bod-
ies, microvesicles and exosomes [13]. The largest EVs (1000–
5000 nm) are apoptotic bodies, which result from the frac-
tionation of the cellular content of cells that die by apoptosis
2 Current Stem Cell Research & Therapy, 2018, Vol. 13, No. 00 Li et al.
[14]. Microvesicles are shed directly from the cell surface
membrane and have a diameter range of 500–1000 nm [15].
Exosomes comprise one of the main subclasses of EVs and
have an endosomal origin [16]. Exosomes are small (30-
100nm) extracellular nano-sized vesicles that originate as the
internal vesicles of multivesicular bodies in many cell types
[17], such as tumor cells, T-cells, mast cells and dendritic
cells, etc. [18]. Recent findings uncovered that exosomes have
a large variety of therapeutic applications in some disorders
such as inflammatory diseases, cardiovascular diseases, meta-
bolic and neurodegenerative disorders, etc. [19].
2. PROPERTIES OF OSTEOPOROSIS AND MSCs-
DERIVED EXOSOMES
2.1. Properties of Osteoporosis
Osteoporosis is a systemic skeletal disease, characterized
by low bone mineral density (BMD) and microarchitectural
deterioration of bone tissues, with a consequent increase in
susceptibility to fracture [20]. Owing to the changes of mi-
cro-architectural in trabecular and cortical skeleton, osteopo-
rosis is manifested by fractures and enhanced skeletal fragil-
ity [21]. Hip and vertebral fractures are the most devastating
consequences of osteoporosis and are closely related to the
increased morbidity and mortality [22]. It is a worldwide
health problem with serious consequences of personal suffer-
ing and economic costs [23]. The pathophysiology of osteo-
porosis is multifactorial. Such as the imbalance between os-
teoclastic bone resorption and osteoblastic bone formation
[24]; decreased osteogenic differentiation and increased adi-
pogenic differentiation [25]. At the same time, angiogenesis
and inflammation also affect the pathogenesis of osteoporo-
sis [26, 27]. Furthermore, deregulation of miRNAs mediated
mechanisms is also becoming an important pathological fac-
tor in bone-related diseases such as osteoporosis [10].
2.2. Properties of Exosomes Derived from MSCs
In the recent years, many studies indicate that paracrine
signaling by MSCs become a potential mechanism to explain
the effects of these cells on tissue regeneration [28]. The
paracrine effects were mediated by numerous factors such as
extracellular vesicles, including exosomes. Exosomes are the
smallest subset of EVs secreted by most cell types, such as
MSCs, dendritic cells, macrophages, epithelial cells, B cells
and T cells, etc. [29]. They can also be identified in the most
bodily fluids including blood, urine, amniotic fluid, serum, etc.
[30]. Similar to the cells, exosomes are composed of a lipid
bilayer and can contain many molecular constituents of a cell,
such as proteins, miRNAs, and DNA, etc. [31]. Tetraspanins
(e.g., CD9, CD63, CD81) have been used as characteristic
markers of exosomes [32]. Currently, the most common
methods to isolate exosomes include ultracentrifugation and
immune-bead isolation [33]. We can discover via the electron
microscopy that exosomes are vesicles with deflated football
or distinctive cup shaped morphology [34] (Fig. 1) [35, 36].
2.3. Genetic Modification and the Secretion of Exosomes
from MSCs
To secrete exosomes, several cellular steps need to be
completed. Firstly, the formation of intraluminal vesicles
(ILVs) in multivesicular bodies (MVBs); secondly, transport
Fig. (1). Exosome biogenesis: exosomes are incorporated into mul-
tivesicular bodies. They could be targeted for lysosomal degrada-
tion or can be fused to cell membrane.
the MVBs to plasma membrane; third, fusion MVBs with the
plasma membrane [37]. In this process, the endosomal sort-
ing complex required for transport (ESCRT) machinery is
very important [38]. ESCRTs consist of approximately
twenty proteins that assemble into four complexes (ESCRT-
0, -I, -II and -III) with associated proteins (VPS4, VTA1,
ALIX, CHMP4C) [39]. Via the thorough RNAi screen study,
seven ESCRT proteins can affect the secretion of exosomes
[37]. Depletion ESCRT-0 proteins Hrs, TSG101 and
ESCRT-I protein STAM1 could reduce exosomes secretion
[37, 39]. Genetic modification might enhance the secretion
of exosomes. For example, knockdown of the ESCRT-III
and associated proteins VPS4B, VTA1, ALIX and CHMP4C
could increase exosome secretion [37]. The ESCRT-0,
ESCRT-I, ESCRT-III and related proteins VPS4B etc. are
important for the secretion of exosomes. So we could modify
these genes in order to regulate the secretion of exosomes
from MSCs.
MSCs secrete and synthesize exosomes that are cholesterol
rich phospholipid vesicles [40]. Compare to transplantation of
exogenous MSCs, MSCs-derived exosomes are less immuno-
genic, easier to store and deliver than MSCs [41]. Searching
for MSCs-derived exosomes is an attractive scope of the in-
vestigation because they are involved in cell-to-cell interac-
tions [42]. Recent studies demonstrate that MSCs-derived
exosomes can modulate the immune response and regenerate
tissues such as heart and nerve in vivo [43].
3. ROLES OF MSCS-DERIVED EXOSOMES IN THE
THERAPY OF OSTEOPOROSIS
3.1. MSCs-derived Exosomes and Osteogenesis
Osteogenesis is a key process responsible for the patho-
genesis of osteoporosis, promoting osteogenesis is important
for the treatments for osteoporosis [44]. Qi showed that
Mesenchymal Stem Cells-Derived Exosomes Current Stem Cell Research & Therapy, 2018, Vol. 13, No. 00 3
MSCs-derived exosomes could enhance ALP activity and
up-regulated mRNA and protein expression of osteoblast
genes in rBMSCs-OVX in vitro. At the same time, MSCs-
derived exosomes can also stimulate bone regeneration in
critical-sized calvarial defects in ovariectomized rats [45].
Furthermore, MSCs-derived exosomes can bind to matrix
proteins such as type I collagen, fibronectin and induce os-
teoblastic differentiation in vitro and in vivo [43]. Bioactive
materials such as Tricalcium Phosphate (-TCP) provide an
alternative solution for the repair of bone defects recently
[46]. However, although -TCP is osteoconductive and bio-
compatible, further improvements to osteogenesis are needed
[47]. Interestingly, when combined with the -TCP scaffolds
besides MSCs-derived exosomes, osteogenesis activity of -
TCP can be enhanced through activating the PI3K/Akt sig-
naling pathway [28, 48]. Given the osteogenic capacity of
the MSCs-derived exosomes in vitro and in vivo, the
exosomes derived from MSCs might be promising vesicles
to improve bone formation in skeletal disorders such as os-
teoporosis [49].
3.2. MSCs-derived Exosomes and Osteoclast Differentia-
tion
Normally, osteoporosis results from an imbalance of
bone resorption and bone formation, where in the net activi-
ties of the osteoclasts supersede the osteoblasts [50]. Thus,
understanding the mechanisms of MSCs-derived exosomes
regulate osteoclast formation may be useful for the treatment
of osteoporosis. MSCs release exosomes which contain func-
tional miRNAs, and then be transferred from cell-to-cell by
exosomes uptake and release, resulting in cross-cellular
gene-regulation [51]. For example, human bone MSCs-
derived exosomal miR-148a has been shown to suppress V-
maf musculoaponeurotic fibrosarcoma oncogene homolog B
(MAFB) expression and promote osteoclastogenesis [52].
The finding suggests that MSCs-derived exosomes might be
an important component to regulate osteoclast differentiation
in the treatment of osteoporosis. However, due to researches
focus on MSCs-derived exosomes influence osteoclast dif-
ferentiation are few recently, so comprehensive studies are
necessary in the future.
3.3. MSCs-derived Exosomes and Angiogenesis
Angiogenesis is the process which new vasculature
sprouts from pre-existing blood vessels [53]. Mounting evi-
dence suggests that local blood supply or decreased angio-
genesis play important roles in osteoporosis [54]. Therefore,
basic strategy for enhancing osteoporotic bone regeneration
is to promote angiogenesis [55]. Various studies have dem-
onstrated the effect of MSCs-derived exosomes on key steps
in angiogenesis [56]. Zhang J et al. found that hiPSC-MSCs-
derived exosomes could promote angiogenesis and collagen
synthesis in fibroblasts in HUVECs directly [57]. Similarly,
another study concluded that iMSCs-derived exosomes could
activate angiogenesis-related molecule expression (such as
PGF, HIF-1, TGFB1, etc.) and promote HUVECs migra-
tion and tube formation [58]. Moreover, angiogenesis plays
crucial roles in numerous physiological processes, it requires
a tight interaction between endothelial cells and their sur-
rounding environment. MSCs-derived exosomes might re-
pressed the expression of the angiogenic inhibitor delta-like
4 (DLL4) by targeting its 3 untranslated region [58]. MSCs-
derived exosomes could modulate endothelial cell angio-
genesis by promoting the formation of endothelial tip cells.
In conclusion, MSCs-derived exosomes as a pro-angiogenic
factor might be a promising candidate for the treatment of
osteoporosis [59].
3.4. MSCs-derived Exosomes and Adipogenesis
In bone marrow, osteoblasts and adipocytes share a
common precursor MSCs [60, 61]. Accumulating informa-
tion show that osteoporosis shifts the MSCs fate to favor
adipocytes over osteoblasts [62], and decrease bone mass
with marrow fat accumulation [63]. Martin et al. found that
human mesenchymal stem cells (HMSCs) derived adipo-
cytes could secrete exosomes containing adipogenic specific
mRNAs (such as leptin, PPARg, CEBPa and CEBPd tran-
scripts) and antiosteogenic miRNAs (miR30c, miR-31,
miR125a, miR-125b, miR-138) that can be transferred to
MSCs-derived osteoblasts [64]. MiR-30c and miR-31, tar-
geting osteogenic transcripts RUNX2 and Osterix [65, 66],
regulate the osteogenic differentiation. MiR-125a and miR-
125b are significantly down-regulated during osteogenic
differentiation in human adipose-derived stem cells [67].
MiR-138 modulates osteogenic differentiation of hMSCs,
overexpression of miR-138 reduced bone formation [68].
MiR-30c, miR-31, miR-125a, miR-125b and miR-138 were
selected for their capacity to inhibit osteoblast gene expres-
sion, all of them showed an increase in their expression in
the research [64]. These results probably implicated that the
miRNAs in exosomes might repress osteogenic differentia-
tion. These findings indicate that MSCs-derived exosomes
might be a target component to regulate the relationship be-
tween osteoblasts and adipocytes in the treatment of osteopo-
rosis.
3.5. MSCs-Derived Exosomes and Inflammation
The process of bone repair requires inflammatory re-
sponse, as the immune system responds to a variety of cyto-
kines that recruit and activate several cell types, like MSCs,
to promote bone formation [69]. Exosomes secreted from
MSCs have been reported to contribute to the regulation of
the immune system [70]. Dysregulated inflammation leads to
the increased bone resorption and suppressed bone formation
[71]. T cells have been identified as key regulators of osteo-
clast and osteoblast formation and activity in different dis-
eases, such as osteoporosis [72]. MSCs-derived exosomes
have an inhibitory role in the differentiation, proliferation,
and activation of T cells [73]; MSCs-derived exosomes
might regulate the immune system and influence bone me-
tabolism, suggesting potential clinical application of MSCs-
derived exosomes in cell-free therapy of osteoporosis.
3.6. MSCs-derived Exosomes and miRNAs
There are growing evidence suggesting that a number of
exosomal-containing miRNAs obtained from MSCs can
regulate bone metabolism [74]. Using isolated MSCs-derived
exosomes, a number of miRNAs (for example, miR-135b,
miR-148a, miR-199b, miR-218 and miR-221) were identi-
fied to be up-regulated during MSCs culture, while some
miRNAs (such as miR-155, miR-181a, miR-221, miR-320c
4 Current Stem Cell Research & Therapy, 2018, Vol. 13, No. 00 Li et al.
and miR-885-5p) were shown to be down-regulated [52, 75].
MiRNA let-7 was reported to enhance osteogenesis while
repressing adipogenesis of HMSCs by regulating HMGA2
[76]. MiR-199b was shown to regulate Runx2 to control the
osteogenesis [77]. MiR-218 stimulates the Wnt pathway to
promote MSCs osteogenesis [78]. MiR-135b reduced the
expression of osterix and integrin binding sialoprotein
(IBSP), suppressed MSCs osteogenic differentiation [79].
Down-regulation of miR-221 was discovered influence
Runx2 expression and trigger osteogenesis in HMSCs [52].
Therefore, future investigation will be focussed on the poten-
tial function of miRNAs derived from MSCs exosomes in
bone metabolism (Table 1) (Fig. 2) [80].
4. THE ADVANTAGE AND CHALLENGE OF MSCs-
DERIVED EXOSOMES FOR OSTEOPOROSIS
THERAPY
In the past few years, MSCs-derived exosomes have
shown exciting promise and great ability to provide thera-
peutically benefits for diseases such as rheumatoid arthritis,
osteoarthritis, etc. [81]. Use of MSCs-derived exosomes has
several potential advantages. First, their usage avoids the
transfer of cells that might have mutated or damaged DNA
[82]; Second, the vesicles are small and circulate readily
whereas MSCs are too large to circulate [82]; Thirdly,
exosomes do not contain MHCI proteins and can overcome
some disadvantages of cell transplantation therapies. Re-
searchers have shown that application to xenogeneic animals
did not induce obvious immune reactions [45].
Moreover, compared to traditional MSCs therapies,
exosomes therapies might decrease injury from MSCs trans-
plantation surgery and the possibility of favoring tumor
growth by MSCs [40]. Furthermore, exosomes have advan-
tages over the corresponding MSCs. MSCs-derived
exosomes are less complex than cells, so they are easier to
produce and also have the potential to avoid some of the
regulatory issues that face in MSCs [83]. Therefore, MSCs-
derived exosomes might represent an ideal therapeutic tool
for bone diseases like osteoporosis in the near future.
However, despite the interesting possibilities for MSCs-
derived exosomes treatment, novel therapeutic approaches
still face some challenges. First, although remarkable pro-
gress have been made in the development of exosomes isola-
tion techniques, it has also been proven that it is challenging
to rapidly and efficiently isolate exosomes. Because of the
complexity of biological samples and the heterogeneity of
exosomes themselves [84]. Second, due to the complex
structure of MSCs-derived exosomes, it might be difficult to
characterize pharmaceutically [85]. Third, the question of
which cell type to be used for exosomes derivation still re-
Table 1. MSCs-derived exosomes miRNAs and their potential effects on bone metabolism.
MiRNA Regulate Gene Effects References
let-7 HMGA2 Enhance ostegenesis/repress adipogenesis [76]
miR-199b Runx2 Control osteogenic [77]
miR-218 Wnt Enhance osteogenesis [78]
miR-135b IBSP/ Osterix Repressing osteogenesis [79]
miR-221 Runx2 Rrepressing osteogenesis [52]
miR-148a MAFB Promote osteoclastogenesis [52]
inflammation
adipogenesisangiogenesis
osteogenesisosteoclastogenesis
osteogenesis
MSC
miR-148a miR-218
miR-135b miR-221
miR30c miR -31 miR125a
miR-138
miR-125b
exo
exo
Fig. (2). MSCs-derived exosomes and the relationship with osteogenesis, osteoclast differentiation, angiogenesis, immune processes and
miRNAs.
Mesenchymal Stem Cells-Derived Exosomes Current Stem Cell Research & Therapy, 2018, Vol. 13, No. 00 5
mains to be answered [86]. Hence, the novel therapeutic ap-
proaches still need comprehensive investigation in order to
bring them into osteoporosis applications.
CONCLUSION
Osteoporosis has become a serious threat to elderly peo-
ple both at the individual and society levels. The recent find-
ings suggest that MSCs-derived exosomes might one day be
able to provide an effective therapy for bone metabolism
disease like osteoporosis. The molecular mechanisms of the
effect by MSCs-derived exosomes on osteogenesis, osteo-
clast differentiation, angiogenesis, angiogenesis and immune
processes and miRNAs have been reported here, however,
the exact mechanism still unknown and has to be studied
further. Meanwhile, considering the majority of the MSCs-
derived exosomes studies are currently on pre-clinical stage
and the conventional methods for isolating and characteriz-
ing exosomes are not effective for clinical application.
Therefore, it is important to develop a large scale of
exosomes production, as well as isolation and purification
methods. Moreover, further investigations are required to
explore specific mechanisms of generation, secretion and
action of MSCs-derived exosomes. Finally, we believe that
exosomes could be a promising new therapeutic strategy for
osteoporosis although several challenges we might face.
CONSENT FOR PUBLICATION
Not applicable.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
This work was funded by the Science and Technology
Planning Project of Guangdong Province,
China.2016A020226007
REFERENCES
[1] Nih Consensus Development Panel on Osteoporosis Prevention D,
Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA
2001; 285: 785-95.
[2] Teitelbaum SL. Stem cells and osteoporosis therapy. Cell Stem
Cell 2010; 7: 553-4.
[3] Liao L, Yang X, Su X, et al. Redundant miR-3077-5p and miR-705
mediate the shift of mesenchymal stem cell lineage commitment to
adipocyte in osteoporosis bone marrow. Cell Death Dis 2013; 4:
e600.
[4] Peng J, Lai ZG, Fang ZL, et al. Dimethyloxalylglycine prevents
bone loss in ovariectomized C57BL/6J mice through enhanced an-
giogenesis and osteogenesis. PLoS One 2014; 9: e112744.
[5] Lin TH, Pajarinen J, Lu L, et al. NF-kappaB as a therapeutic target
in inflammatory-associated bone diseases. Adv Protein Chem
Struct Biol 2017; 107: 117-54.
[6] Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new
era for the management of cancer and other diseases. Nat Rev Drug
Discov 2017; 16: 203-22.
[7] Sun M, Zhou X, Chen L, et al. The Regulatory roles of micrornas
in bone remodeling and perspectives as biomarkers in osteoporosis.
Biomed Res Int 2016; 2016: 1652417.
[8] Cao Z, Moore BT, Wang Y, et al. MiR-422a as a potential cellular
microRNA biomarker for postmenopausal osteoporosis. PLoS One
2014; 9: e97098.
[9]
Zhang B, Wu X, Zhang X, et al. Human umbilical cord mesen-
chymal stem cell exosomes enhance angiogenesis through the
Wnt4/beta-catenin pathway. Stem Cells Transl Med 2015; 4: 513-
22.
[10]
Alcayaga-Miranda F, Varas-Godoy M, Khoury M. Harnessing the
angiogenic potential of stem cell-derived exosomes for vascular re-
generation. Stem Cells Int 2016; 2016: 3409169.
[11] Merino-Gonzalez C, Zuniga FA, Escudero C, et al. Mesenchymal
stem cell-derived extracellular vesicles promote angiogenesis: po-
tencial clinical application. Front Physiol 2016; 7: 24.
[12] Whiteside TL. Exosome and mesenchymal stem cell cross-talk in
the tumor microenvironment. Semin Immunol 2017.
[13] Vader P, Mol EA, Pasterkamp G, Schiffelers RM. Extracellular
vesicles for drug delivery. Adv Drug Deliv Rev 2016; 106: 148-56.
[14] Barile L, Vassalli G. Exosomes: Therapy delivery tools and bio-
markers of diseases. Pharmacol Ther 2017; 174: 63-78.
[15] Colao IL, Corteling R, Bracewell D, Wall I. Manufacturing
exosomes: a promising therapeutic platform. Trends Mol Med
2018; 24: 242-56.
[16] Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-
derived extracellular vesicles: novel frontiers in regenerative medi-
cine. Stem Cell Res Ther 2018; 9: 63.
[17] Ge M, Ke R, Cai T, Yang J, Mu X. Identification and proteomic
analysis of osteoblast-derived exosomes. Biochem Biophys Res
Commun 2015; 467: 27-32.
[18] Kishore R, Khan M. More than tiny sacks: stem cell exosomes as
cell-free modality for cardiac repair. Circ Res 2016; 118: 330-43.
[19] Nazimek K, Bryniarski K, Santocki M, Ptak W. Exosomes as me-
diators of intercellular communication: clinical implications. Pol
Arch Med Wewn 2015; 125: 370-80.
[20] Xu XH, Dong SS, Guo Y, et al. Molecular genetic studies of gene
identification for osteoporosis: the 2009 update. Endocr Rev 2010;
31: 447-505.
[21] Kawai M, Modder UI, Khosla S, Rosen CJ. Emerging therapeutic
opportunities for skeletal restoration. Nat Rev Drug Discov 2011;
10: 141-56.
[22] Ensrud KE, Crandall CJ. Osteoporosis. Ann Intern Med 2017; 167:
ITC17-ITC32.
[23] Silva I, Branco JC. Denosumab: recent update in postmenopausal
osteoporosis. Acta Reumatol Port 2012; 37: 302-13.
[24] Zhao XL, Chen JJ, Zhang GN, et al. Small molecule T63 sup-
presses osteoporosis by modulating osteoblast differentiation via
BMP and WNT signaling pathways. Sci Rep 2017; 7: 10397.
[25] Wang C, Meng H, Wang X, Zhao C, Peng J, Wang Y. Differentia-
tion of bone marrow mesenchymal stem cells in osteoblasts and
adipocytes and its role in treatment of osteoporosis. Med Sci Monit
2016; 22: 226-33.
[26] Zhao Q, Shen X, Zhang W, Zhu G, Qi J, Deng L. Mice with in-
creased angiogenesis and osteogenesis due to conditional activation
of HIF pathway in osteoblasts are protected from ovariectomy in-
duced bone loss. Bone 2012; 50: 763-70.
[27] Redlich K, Smolen JS. Inflammatory bone loss: pathogenesis and
therapeutic intervention. Nat Rev Drug Discov 2012; 11: 234-50.
[28] Furuta T, Miyaki S, Ishitobi H, et al. Mesenchymal Stem Cell-
Derived Exosomes Promote Fracture Healing in a Mouse Model.
Stem Cells Transl Med 2016; 5: 1620-30.
[29] Batrakova EV, Kim MS. Using exosomes, naturally-equipped
nanocarriers, for drug delivery. J Control Release 2015; 219: 396-
405.
[30] Beach A, Zhang HG, Ratajczak MZ, Kakar SS. Exosomes: an
overview of biogenesis, composition and role in ovarian cancer. J
Ovarian Res 2014; 7: 14.
[31] Kalluri R. The biology and function of exosomes in cancer. J Clin
Invest 2016; 126: 1208-15.
[32] Zhou Y, Xu H, Xu W, et al. Exosomes released by human umbili-
cal cord mesenchymal stem cells protect against cisplatin-induced
renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell
Res Ther 2013; 4: 34.
[33] Zhang X, Yuan X, Shi H, Wu L, Qian H, Xu W. Exosomes in
cancer: small particle, big player. J Hematol Oncol 2015; 8: 83.
[34] Masyuk AI, Masyuk TV, Larusso NF. Exosomes in the pathogene-
sis, diagnostics and therapeutics of liver diseases. J Hepatol 2013;
59: 621-5.
[35] Qin Y, Sun R, Wu C, Wang L, Zhang C. Exosome: A Novel Ap-
proach to Stimulate Bone Regeneration through Regulation of Os-
teogenesis and Angiogenesis. Int J Mol Sci 2016; 17.
6 Current Stem Cell Research & Therapy, 2018, Vol. 13, No. 00 Li et al.
[36]
Raab-Traub N, Dittmer DP. Viral effects on the content and func-
tion of extracellular vesicles. Nat Rev Microbiol 2017; 15: 559-72.
[37]
Hessvik NP, Llorente A. Current knowledge on exosome biogene-
sis and release. Cell Mol Life Sci 2018; 75: 193-208.
[38]
Dreyer F, Baur A. Biogenesis and Functions of Exosomes and
Extracellular Vesicles. Methods Mol Biol 2016; 1448: 201-16.
[39]
Colombo M, Moita C, van Niel G, et al. Analysis of ESCRT func-
tions in exosome biogenesis, composition and secretion highlights
the heterogeneity of extracellular vesicles. J Cell Sci 2013; 126:
5553-65.
[40] Huang L, Ma W, Ma Y, Feng D, Chen H, Cai B. Exosomes in
mesenchymal stem cells, a new therapeutic strategy for cardiovas-
cular diseases? Int J Biol Sci 2015; 11: 238-45.
[41] Zhang Y, Chopp M, Meng Y, et al. Effect of exosomes derived
from multipluripotent mesenchymal stromal cells on functional re-
covery and neurovascular plasticity in rats after traumatic brain in-
jury. J Neurosurg 2015; 122: 856-67.
[42] Pashoutan Sarvar D, Shamsasenjan K, Akbarzadehlaleh P. Mesen-
chymal stem cell-derived exosomes: new opportunity in cell-free
therapy. Adv Pharm Bull 2016; 6: 293-9.
[43] Narayanan R, Huang CC, Ravindran S. Hijacking the cellular mail:
exosome mediated differentiation of mesenchymal stem cells. Stem
Cells Int 2016; 2016: 3808674.
[44] Sun T, Li CT, Xiong L, et al. miR-375-3p negatively regulates
osteogenesis by targeting and decreasing the expression levels of
LRP5 and beta-catenin. PLoS One 2017; 12: e0171281.
[45] Qi X, Zhang J, Yuan H, et al. Exosomes secreted by human-
induced pluripotent stem cell-derived mesenchymal stem cells re-
pair critical-sized bone defects through enhanced angiogenesis and
osteogenesis in osteoporotic rats. Int J Biol Sci 2016; 12: 836-49.
[46] Wang Z, Guo Z, Bai H, et al. Clinical evaluation of beta-TCP in
the treatment of lacunar bone defects: a prospective, randomized
controlled study. Mater Sci Eng C Mater Biol Appl 2013; 33: 1894-
9.
[47] Deng Y, Jiang C, Li C, et al. 3D printed scaffolds of calcium sili-
cate-doped beta-TCP synergize with co-cultured endothelial and
stromal cells to promote vascularization and bone formation. Sci
Rep 2017; 7: 5588.
[48] Zhang J, Liu X, Li H, et al. Exosomes/tricalcium phosphate combi-
nation scaffolds can enhance bone regeneration by activating the
PI3K/Akt signaling pathway. Stem Cell Res Ther 2016; 7: 136.
[49] Liu J, Li D, Wu X, Dang L, Lu A, Zhang G. Bone-derived
exosomes. Curr Opin Pharmacol 2017; 34: 64-9.
[50] Wu X, Pang L, Lei W, et al. Inhibition of Sca-1-positive skeletal
stem cell recruitment by alendronate blunts the anabolic effects of
parathyroid hormone on bone remodeling. Cell Stem Cell 2010; 7:
571-80.
[51] Katakowski M, Buller B, Zheng X, et al. Exosomes from marrow
stromal cells expressing miR-146b inhibit glioma growth. Cancer
Lett 2013; 335: 201-4.
[52] Zhu S, Yao F, Qiu H, Zhang G, Xu H, Xu J. Coupling factors and
exosomal packaging microRNAs involved in the regulation of bone
remodelling. Biol Rev Camb Philos Soc 2018; 93: 469-80.
[53] Olsen JJ, Pohl SO, Deshmukh A, et al. The Role of Wnt signalling
in angiogenesis. Clin Biochem Rev 2017; 38: 131-42.
[54] Wang L, Jia P, Shan Y, et al. Synergistic protection of bone vascu-
lature and bone mass by desferrioxamine in osteoporotic mice. Mol
Med Rep 2017; 16: 6642-9.
[55] Jia P, Chen H, Kang H, et al. Deferoxamine released from
poly(lactic-co-glycolic acid) promotes healing of osteoporotic bone
defect via enhanced angiogenesis and osteogenesis. J Biomed Ma-
ter Res A 2016; 104: 2515-27.
[56] Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R. Mesen-
chymal Stem Cell Secretome: Toward Cell-Free Therapeutic
Strategies in Regenerative Medicine. Int J Mol Sci 2017; 18.
[57] Zhang J, Guan J, Niu X, et al. Exosomes released from human
induced pluripotent stem cells-derived MSCs facilitate cutaneous
wound healing by promoting collagen synthesis and angiogenesis. J
Transl Med 2015; 13: 49.
[58] Hu GW, Li Q, Niu X, et al. Exosomes secreted by human-induced
pluripotent stem cell-derived mesenchymal stem cells attenuate
limb ischemia by promoting angiogenesis in mice. Stem Cell Res
Ther 2015; 6: 10.
[59] Gong M, Yu B, Wang J, et al. Mesenchymal stem cells release
exosomes that transfer miRNAs to endothelial cells and promote
angiogenesis. Oncotarget 2017; 8: 45200-12.
[60]
Li J, Liu X, Zuo B, Zhang L. The Role of Bone Marrow Microen-
vironment in Governing the Balance between Osteoblastogenesis
and Adipogenesis. Aging Dis 2016; 7: 514-25.
[61]
Xu C, Wang J, Zhu T, et al. Cross-Talking Between PPAR and
WNT Signaling and its Regulation in Mesenchymal Stem Cell Dif-
ferentiation. Curr Stem Cell Res Ther 2016; 11: 247-54.
[62]
Yuan Z, Li Q, Luo S, et al. PPARgamma and Wnt Signaling in
Adipogenic and Osteogenic Differentiation of Mesenchymal Stem
Cells. Curr Stem Cell Res Ther 2016; 11: 216-25.
[63] Chen Q, Shou P, Zheng C, et al. Fate decision of mesenchymal
stem cells: adipocytes or osteoblasts? Cell Death Differ 2016; 23:
1128-39.
[64] Martin PJ, Haren N, Ghali O, et al. Adipogenic RNAs are trans-
ferred in osteoblasts via bone marrow adipocytes-derived extracel-
lular vesicles (EVs). BMC Cell Biol 2015; 16: 10.
[65] Zhang Y, Xie RL, Croce CM, et al. A program of microRNAs
controls osteogenic lineage progression by targeting transcription
factor Runx2. Proc Natl Acad Sci U S A 2011; 108: 9863-8.
[66] Baglio SR, Devescovi V, Granchi D, Baldini N. MicroRNA ex-
pression profiling of human bone marrow mesenchymal stem cells
during osteogenic differentiation reveals Osterix regulation by
miR-31. Gene 2013; 527: 321-31.
[67] Gong Y, Xu F, Zhang L, et al. MicroRNA expression signature for
Satb2-induced osteogenic differentiation in bone marrow stromal
cells. Mol Cell Biochem 2014; 387: 227-39.
[68] Eskildsen T, Taipaleenmaki H, Stenvang J, et al. MicroRNA-138
regulates osteogenic differentiation of human stromal (mesenchy-
mal) stem cells in vivo. Proc Natl Acad Sci U S A 2011; 108: 6139-
44.
[69] Kovach TK, Dighe AS, Lobo PI, Cui Q. Interactions between
MSCs and immune cells: implications for bone healing. J Immunol
Res 2015; 2015: 752510.
[70] Wang J, Faict S, Maes K, et al. Extracellular vesicle cross-talk in
the bone marrow microenvironment: implications in multiple mye-
loma. Oncotarget 2016; 7: 38927-45.
[71] Loi F, Cordova LA, Pajarinen J, Lin TH, Yao Z, Goodman SB.
Inflammation, fracture and bone repair. Bone 2016; 86: 119-30.
[72] Faienza MF, Ventura A, Marzano F, Cavallo L. Postmenopausal
osteoporosis: the role of immune system cells. Clin Dev Immunol
2013; 2013: 575936.
[73] Ti D, Hao H, Fu X, Han W. Mesenchymal stem cells-derived
exosomal microRNAs contribute to wound inflammation. Sci
China Life Sci 2016; 59: 1305-12.
[74] Xie Y, Chen Y, Zhang L, Ge W, Tang P. The roles of bone-derived
exosomes and exosomal microRNAs in regulating bone remodel-
ling. J Cell Mol Med 2017; 21: 1033-41.
[75] Xu JF, Yang GH, Pan XH, et al. Altered microRNA expression
profile in exosomes during osteogenic differentiation of human
bone marrow-derived mesenchymal stem cells. PLoS One 2014; 9:
e114627.
[76] Wei J, Li H, Wang S, et al. let-7 enhances osteogenesis and bone
formation while repressing adipogenesis of human stro-
mal/mesenchymal stem cells by regulating HMGA2. Stem Cells
Dev 2014; 23: 1452-63.
[77] Lauvrak SU, Munthe E, Kresse SH, et al. Functional characterisa-
tion of osteosarcoma cell lines and identification of mRNAs and
miRNAs associated with aggressive cancer phenotypes. Br J Can-
cer 2013; 109: 2228-36.
[78] Zhang WB, Zhong WJ, Wang L. A signal-amplification circuit
between miR-218 and Wnt/beta-catenin signal promotes human
adipose tissue-derived stem cells osteogenic differentiation. Bone
2014; 58: 59-66.
[79] Xu S, Cecilia Santini G, De Veirman K, et al. Upregulation of
miR-135b is involved in the impaired osteogenic differentiation of
mesenchymal stem cells derived from multiple myeloma patients.
PLoS One 2013; 8: e79752.
[80] Wu P, Zhang B, Shi H, Qian H, Xu W. MSC-exosome: A novel
cell-free therapy for cutaneous regeneration. Cytotherapy 2018; 20:
291-301.
[81] Burke J, Kolhe R, Hunter M, Isales C, Hamrick M, Fulzele S. Stem
Cell-Derived Exosomes: A Potential Alternative Therapeutic Agent
in Orthopaedics. Stem Cells Int 2016; 2016: 5802529.
[82] Phinney DG, Pittenger MF. Concise Review: MSC-Derived
Exosomes for Cell-Free Therapy. Stem Cells 2017; 35: 851-8.
Mesenchymal Stem Cells-Derived Exosomes Current Stem Cell Research & Therapy, 2018, Vol. 13, No. 00 7
[83]
Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell-derived
exosomes as a new therapeutic strategy for liver diseases. Exp Mol
Med 2017; 49: e346.
[84]
Li P, Kaslan M, Lee SH, Yao J, Gao Z. Progress in Exosome Isola-
tion Techniques. Theranostics 2017; 7: 789-804.
[85]
Emanueli C, Shearn AI, Angelini GD, Sahoo S. Exosomes and
exosomal miRNAs in cardiovascular protection and repair. Vascul
Pharmacol 2015; 71: 24-30.
[86]
Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D.
Engineering exosomes as refined biological nanoplatforms for drug
delivery. Acta Pharmacol Sin 2017; 38: 754-63.
... Many studies have focused on differentiating stem cells into particular cell types, such as adipocytes [9], chondrocytes [1, 10], hepatocytes [2], and osteoblasts [11]. Other studies focused on their advantageous immunomodulatory properties as they secrete intrinsic cytokines that have the potential to mitigate inflammations and attenuate fibrosis [12,13]. Consequently, stem cells are considered indispensable due to their unique combination of multipotency and immunomodulatory capabilities. ...
Article
Full-text available
In multicellular organisms, stem cells are impacted by microenvironmental resources such as nutrient availability and oxygen tension for their survival, growth, and differentiation. However, the accessibility of these resources in the pericellular environment greatly varies from organ to organ. This divergence in resource availability leads to variations in the potency and differentiation potential of stem cells. This study aimed to explore the distinct effects of glucose and fructose, as well as different oxygen tensions, on the growth dynamics, cytokine production, and differentiation of stem cells. We showed that replacing glucose with fructose subjected stem cells to stress, resulting in increased Hif1α expression and stability, which in turn led to a reduction in cell proliferation, and alterations in cytokine production. However, fructose failed to induce differentiation of human mesenchymal stem cells (hMSCs) as well as mouse fibroblasts into mature adipocytes compared to glucose, despite the upregulation of key markers of adipogenesis, including C/EBPβ, and PPARγ. Conversely, we showed that fructose induced undifferentiated mouse fibroblasts to release cytokines associated with senescence, including IL1α1, IL6, IL8, MCP1, and TNF1α, suggesting that these cells were undergoing lipolysis. Taken together, our results suggest that altering the culture conditions through changes in hexose levels and oxygen tension places considerable stress on stem cells. Additional research is required to further characterize the mechanisms governing stem cell response to their microenvironments.
... For instance, it has shown effectiveness in ameliorating bone loss in various animal models with minimal side effects [9,10]. The underlying mechanisms involve the paracrine action of cytokines, cell-to-cell contact, and the release of extracellular vesicles, which is currently a major research focus [11][12][13]. In our early studies, we utilized human bone marrow mesenchymal stem cells (BMMSCs) and exfoliated deciduous tooth stem cells (SHED) in the treatment of an osteoporosis mouse model induced by ovariectomy (OVX). ...
Preprint
Full-text available
Apoptotic vesicles (apoVs) play a vital role in various pathological conditions; however, we have yet to fully understand their precise biological effects in rescuing impaired mesenchymal stem cells (MSCs) and regulating tissue homeostasis. Here, we proved that systemic infusion of bone marrow MSCs derived from wild-type (WT) mice effectively improved the osteopenia phenotype and hyperimmune state in ovariectomized (OVX) mice. Importantly, the WT MSCs rescued the impairment of OVX MSCs both in vivo and in vitro , whereas OVX MSCs did not show the same efficacy. Interestingly, treatment with apoVs derived from WT MSCs (WT apoVs) restored the impaired biological function of OVX MSCs and their ability to improve osteoporosis. This effect was not observed with OVX MSCs-derived apoVs (OVX apoVs) treatment. Mechanistically, the reduced miR-145a-5p expression hindered the osteogenic differentiation and immunomodulatory capacity of OVX MSCs by affecting the TGF-β/Smad 2/3-Wnt/β-catenin signaling axis, resulting in the development of osteoporosis. WT apoVs directly transferred miR-145a-5p to OVX MSCs, which were then reused to restore their impaired biological functions. Conversely, treatment with OVX apoVs did not produce significant effects due to their limited expression of miR-145a-5p. Overall, our findings unveil the remarkable potential of apoVs in rescuing the biological function and therapeutic capability of MSCs derived from individuals with diseases. This discovery offers a new avenue for exploring apoVs-based MSC engineering and expands the application scope of stem cell therapy, contributing to the maintenance of bone homeostasis through a previously unrecognized mechanism.
... Terefore, although the current medications have exerted satisfactory results, their adverse efects have encouraged researchers to seek novel approaches for the treatment of osteoporosis and its related fractures. Since osteoporosis management should be based on tissue regeneration principles [18][19][20], there is growing evidence that the application of platelet-derived biomaterials (PDBs), socalled platelet concentrates, may support a treatment for osteoporosis-related injury [21][22][23][24]. ...
Article
Full-text available
Osteoporosis is a chronic multifactorial condition that affects the skeletal system, leading to the deterioration of bone microstructure and an increased risk of bone fracture. Platelet-derived biomaterials (PDBs), so-called platelet concentrates, such as platelet-rich plasma (PRP) and platelet-rich fibrin (PRF), have shown potential for improving bone healing by addressing microstructural impairment. While the administration of platelet concentrates has yielded positive results in bone regeneration, the optimal method for its administration in the clinical setting is still debatable. This comprehensive review aims to explore the systemic and local use of PRP/PRF for treating various bone defects and acute fractures in patients with osteoporosis. Furthermore, combining PRP/PRF with stem cells or osteoinductive and osteoconductive biomaterials has shown promise in restoring bone microstructural properties, treating bony defects, and improving implant osseointegration in osteoporotic animal models. Here, reviewing the results of in vitro and in vivo studies, this comprehensive evaluation provides a detailed mechanism for how platelet concentrates may support the healing process of osteoporotic bone fractures.
... Recently, a lot of studies have shown that MSC-derived exosomes are effective for the treatment of a variety of diseases like cancer, cardiovascular diseases, bone tissue regeneration, etc. Thus, the MSC-derived exosomes could serve as a novel therapeutic tool for osteoporosis in the future [64]. MiRNAs can regulate bone formation and/or resorption and mineralization. ...
Article
Aging is an inevitable process of our civilization. Since ancient times, scientists have tried to solve the mystery of aging. The research continues to this day. The most common diseases of old age are disorders of the musculoskeletal system. Among them, osteoporosis continues to occupy the third place in the structure of overall morbidity and mortality. Such statistical data make us think about the search for a possible cause of the disease at the molecular level. The purpose of this study was to analyze the literature data on modern directions of osteoporosis treatment, including stem-cells based bioregenerative medicine. An analytical review of literature data was conducted using the information analysis of Medline (PubMed), Web of Science and Scopus databases, Google Scholar and the Cochrane Central Register of Controlled Trials (CENTRAL) from 2018 to 2022 using the keywords “osteoporosis”, bioregenerative therapy”, “stem cell therapy”. Recent results of preclinical experimental studies have shown the effectiveness of the introduction of new bioregenerative technologies. In particular, the use of mesenchymal stem cells, exosomes and miRNAs. Preclinical studies on MSC transplantation in the treatment of osteoporosis indicate an increase in osteogenic differentiation, an increase in BMD. Exosomes also may play multiple roles in the treatment of osteoporosis: improving the disbalance between osteoclasts and osteoblasts, structural modification of exosomes and transmitters’ drug function. The promotion of bone regeneration of exosomes has been shown in animal models. Exosomes with active ingredients can treat a variety of skeletal disorders including osteoporosis and osteoporotic fractures. The results of recent research of the bone disorder treatment based on stem cells therapy have shown convincing prospects for new approaches.
Article
Full-text available
Skeletal diseases impose a considerable burden on society. The clinical and tissue-engineering therapies applied to alleviate such diseases frequently result in complications and are inadequately effective. Research has shifted from conventional therapies based on mesenchymal stem cells (MSCs) to exosomes derived from MSCs. Exosomes are natural nanocarriers of endogenous DNA, RNA, proteins, and lipids and have a low immune clearance rate and good barrier penetration and allow targeted delivery of therapeutics. MSC-derived exosomes (MSC-exosomes) have the characteristics of both MSCs and exosomes, and so they can have both immunosuppressive and tissue-regenerative effects. Despite advances in our knowledge of MSC-exosomes, their regulatory mechanisms and functionalities are unclear. Here we review the therapeutic potential of MSC-exosomes for skeletal diseases.
Article
Full-text available
Osteoporotic vertebral compression fractures (OVCFs) significantly increase morbidity and mortality, presenting a formidable challenge in healthcare. Traditional interventions such as vertebroplasty and kyphoplasty, despite their widespread use, are limited in addressing the secondary effects of vertebral fractures in adjacent areas and do not facilitate bone regeneration. This review paper explores the emerging domain of regenerative therapies, spotlighting stem cell therapy’s transformative potential in OVCF treatment. It thoroughly describes the therapeutic possibilities and mechanisms of action of mesenchymal stem cells against OVCFs, relying on recent clinical trials and preclinical studies for efficacy assessment. Our findings reveal that stem cell therapy, particularly in combination with scaffolding materials, holds substantial promise for bone regeneration, spinal stability improvement, and pain mitigation. This integration of stem cell-based methods with conventional treatments may herald a new era in OVCF management, potentially improving patient outcomes. This review advocates for accelerated research and collaborative efforts to translate laboratory breakthroughs into clinical practice, emphasizing the revolutionary impact of regenerative therapies on OVCF management. In summary, this paper positions stem cell therapy at the forefront of innovation for OVCF treatment, stressing the importance of ongoing research and cross-disciplinary collaboration to unlock its full clinical potential.
Article
Full-text available
Osteoporotic tendon‐to‐bone healing (TBH) after rotator cuff repair (RCR) is a significant orthopedic challenge. Considering the aligned architecture of the tendon, inflammatory microenvironment at the injury site, and the need for endogenous cell/tissue infiltration, there is an imminent need for an ideal scaffold to promote TBH that has aligned architecture, ability to modulate inflammation, and macroporous structure. Herein, a novel macroporous hydrogel comprising sodium alginate/hyaluronic acid/small extracellular vesicles from adipose‐derived stem cells (sEVs) (MHA‐sEVs) with aligned architecture and immunomodulatory ability is fabricated. When implanted subcutaneously, MHA‐sEVs significantly improve cell infiltration and tissue integration through its macroporous structure. When applied to the osteoporotic RCR model, MHA‐sEVs promote TBH by improving tendon repair through macroporous aligned architecture while enhancing bone regeneration by modulating inflammation. Notably, the biomechanical strength of MHA‐sEVs is approximately two times higher than the control group, indicating great potential in reducing postoperative retear rates. Further cell‐hydrogel interaction studies reveal that the alignment of microfiber gels in MHA‐sEVs induces tenogenic differentiation of tendon‐derived stem cells, while sEVs improve mitochondrial dysfunction in M1 macrophages (Mφ) and inhibit Mφ polarization toward M1 via nuclear factor‐kappaB (NF‐κb) signaling pathway. Taken together, MHA‐sEVs provide a promising strategy for future clinical application in promoting osteoporotic TBH.
Article
Full-text available
Mesenchymal stem cells (MSCs) are multipotent stem cells that have gained significant attention in the field of regenerative medicine. The differentiation potential along with paracrine properties of MSCs have made them a key option for tissue repair. The paracrine functions of MSCs are applied through secreting soluble factors and releasing extracellular vesicles like exosomes and microvesicles. Extracellular vesicles are predominantly endosomal in origin and contain a cargo of miRNA, mRNA, and proteins that are transferred from their original cells to target cells. Recently it has emerged that extracellular vesicles alone are responsible for the therapeutic effect of MSCs in plenty of animal diseases models. Hence, MSC-derived extracellular vesicles may be used as an alternative MSC-based therapy in regenerative medicine. In this review we discuss MSC-derived extracellular vesicles and their therapeutic potential in various diseases.
Article
Full-text available
Extracellular vesicles, in particular the subclass exosomes, are rapidly emerging as a novel therapeutic platform. However, currently very few clinical validation studies and no clearly defined manufacturing process exist. As exosomes progress towards the clinic for treatment of a vast array of diseases, it is important to define the engineering basis for their manufacture early in the development cycle to ensure they can be produced cost-effectively at the appropriate scale. We hypothesize that transitioning to defined manufacturing platforms will increase consistency of the exosome product and improve their clinical advancement as a new therapeutic tool. We present manufacturing technologies and strategies that are being implemented and consider their application for the transition from bench-scale to clinical production of exosomes.
Article
Full-text available
Angiogenesis is a normal biological process wherein new blood vessels form from the growth of pre-existing blood vessels. Preventing angiogenesis in solid tumours by targeting pro-angiogenic factors including vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1), basic fibroblast growth factor (bFGF), hepatocyte growth factor, and platelet-derived growth factor (PDGF) is currently under investigation for cancer treatment. Concurrently targeting the cell signalling pathways involved in the transcriptional and post-translational regulation of these factors may provide positive therapeutic results. One such pathway is the Wnt signalling pathway. Wnt was first discovered in mice infected with mouse mammary tumour virus, and has been crucial in improving our understanding of oncogenesis and development. In this review, we summarise molecular and cellular aspects of the importance of Wnt signalling to angiogenesis, including β-catenin-dependent mechanisms of angiogenic promotion, as well as the study of Wnt antagonists, such as the secreted frizzled-related protein family (SFRPs) which have been shown to inhibit angiogenesis. The growing understanding of the underlying complexity of the biochemical pathways mediating angiogenesis is critical to the identification of new molecular targets for therapeutic applications.
Article
Full-text available
It has previously been demonstrated that impaired angiogenesis is associated with metabolic abnormalities in bone in addition to osteoporosis (including postmenopausal osteoporosis). Enhancing vessel formation in bone is therefore a potential clinical therapy for osteoporosis. The present study conducted an in‑depth investigation using desferrioxamine (DFO) in an ovariectomy (OVX)‑induced osteoporotic mouse model in order to determine the time frame of alteration of bone characteristics and the therapeutic effect of DFO. It was demonstrated that OVX induced instant bone mass loss 1 week following surgery, as expected. In contrast, DFO treatment protected the mice against OVX‑induced osteoporosis during the first week, however failed to achieve long‑term protection at a later stage. A parallel alteration for cluster of differentiation 31/endomucin double positive vessels (type H vessels) was observed, which have previously been reported to be associated with osteogenesis. DFO administration not only partially prevented bone loss and maintained trabecular bone microarchitecture, however additionally enhanced the type H vessels during the first week post‑OVX. The molecular mechanism of how DFO influences type H vessels to regulate bone metabolism needs to be further investigated. However, the findings of the present study provide preliminary evidence to support combined vascular and osseous therapies for osteoporotic patients. Pharmacotherapy may offer a novel target for improving osteoporosis by promoting type H vessel formation, which indicates potential clinical significance in the field of bone metabolism.
Article
Full-text available
Osteoporosis results from the imbalance between bone resorption and bone formation, and restoring the normal balance of bone remodeling is highly desirable for identification of better treatment. In this study, using a cell-based high-throughput screening model representing Runt-related transcription factor 2 (RUNX2) transcriptional activity, we identified a novel small-molecular-weight compound, T63, as an efficient up-regulator of osteogenesis. T63 increased the alkaline phosphatase (ALPL) activity and mineralization as well as gene expression of Alpl and other osteogenic marker genes in mouse osteoblasts and mesenchymal stem cell-like cells. Upon induction of osteoblast differentiation, T63 inhibited adipogenic differentiation in the pluripotent mesenchymal cells. Consistently, T63 up-regulated RUNX2 mRNA and protein levels, and knockdown of RUNX2 reduced the osteogenic role of T63. Mechanistically, T63 activated both BMPs and WNT/β-catenin signaling pathways. Inhibition of either signaling pathway with specific inhibitor suppressed T63-induced RUNX2 expression and the osteogenic phenotypes. Moreover, T63 markedly protected against bone mass loss in the ovariectomized and dexamethasone treated rat osteoporosis model. Collectively, our data demonstrate that T63 could be a promising drug candidate and deserves further development for potential therapeutics in osteoporosis.
Article
Full-text available
Earlier research primarily attributed the effects of mesenchymal stem cell (MSC) therapies to their capacity for local engrafting and differentiating intomultiple tissue types.However, recent studies have revealed that implanted cells do not survive for long, and that the benefits of MSC therapy could be due to the vast array of bioactive factors they produce, which play an important role in the regulation of key biologic processes. Secretome derivatives, such as conditioned media or exosomes, may present considerable advantages over cells for manufacturing, storage, handling, product shelf life and their potential as a ready-to-go biologic product. Nevertheless, regulatory requirements for manufacturing and quality control will be necessary to establish the safety and efficacy profile of these products. Among MSCs, human uterine cervical stem cells (hUCESCs) may be a good candidate for obtaining secretome-derived products. hUCESCs are obtained by Pap cervical smear, which is a less invasive and painful method than those used for obtaining other MSCs (for example, from bone marrow or adipose tissue). Moreover, due to easy isolation and a high proliferative rate, it is possible to obtain large amounts of hUCESCs or secretome-derived products for research and clinical use.
Article
Full-text available
Bone remodelling is a continuous process by which bone resorption by osteoclasts is followed by bone formation by osteoblasts to maintain skeletal homeostasis. These two forces must be tightly coordinated not only quantitatively, but also in time and space, and its malfunction leads to diseases such as osteoporosis. Recent research focusing on the cross-talk and coupling mechanisms associated with the sequential recruitment of osteoblasts to areas where osteoclasts have removed bone matrix have identified a number of osteogenic factors produced by the osteoclasts themselves. Osteoclast-derived factors and exosomal-containing microRNA (miRNA) can either enhance or inhibit osteoblast differentiation through paracrine and juxtacrine mechanisms, and therefore may have a central coupling role in bone formation. Entwined with angiocrine factors released by vessel-specific endothelial cells and perivascular cells or pericytes, these factors play a critical role in angiogenesis–osteogenesis coupling essential in bone remodelling.
Article
Cutaneous regeneration is a dynamic and complex process that requires a series of coordinated interactions involving epidermal cells, dermal cells, growth factors, the extracellular matrix (ECM), nerves and blood vessels at a damaged site. Mesenchymal stromal cells (MSCs) have been reported to participate in all afore-mentioned stages. Exosomes are one of the key secretory products of MSCs, resembling the effect of parental MSCs. They can shuttle various proteins, messenger RNA (mRNA) and microRNAs (miRNAs) to modulate the activity of recipient cells, and play important roles in cutaneous wound healing. Compared with MSCs, exosomes are more convenient to store and transport. Moreover, they avoid many risks associated with cell transplantation. Therefore, MSC-exosome-mediated therapy may be more safe and efficient. In this review, we summarize the latest studies and observations on the role of MSC-exosome in the acute and chronic wound model and provide a comprehensive understanding of the role of exosomes in wound healing. This review can assist investigators in exploring new therapeutic strategies for enhancing the efficacy of MSC-exosome for cutaneous repair and regeneration.
Article
Mesenchymal stem cells (MSCs) are a major component of the tumor microenvironment (TME) and play a key role in promoting tumor progression. The tumor uses exosomes to co-opt MSCs and re-program their functional profile from normally trophic to pro-tumorigenic. These tumor-derived small vesicles called "TEX" carry and deliver a cargo rich in proteins and nucleic acids to MSCs. Upon interactions with surface receptors on MSCs and uptake of the exosome cargo by MSCs, molecular, transcriptional and translational changes occur that convert MSCs into producers of factors that are necessary for tumor growth and that also alter functions of non-tumor cells in the TME. The MSCs re-programmed by TEX become avid producers of their own exosomes that carry and deliver mRNA and miRNA species as well as molecular signals not only back to tumor cells, directly enhancing their growth, but also horizontally to fibroblasts, endothelial cells and immune cells in the TME, indirectly enhancing their pro-tumor functions. TEX-driven cross-talk of MSCs with immune cells blocks their anti-tumor activity and/or converts them into suppressor cells. MSCs re-programmed by TEX mediate pro-angiogenic activity and convert stromal cells into cancer-associated fibroblasts (CAFs). Although MSCs have a potential to exert anti-tumor activities, they largely provide service to the tumor using the multidirectional communication system established by exosomes in the TME. Future therapeutic options consider disruption of this complex vicious cycle by either molecular or gene-regulated silencing of pro-tumor effects mediated by MSCs in the TME.
Article
Exosomes are endosomal-derived vesicles secreted by a wide range of cell types for transporting functional proteins, microRNAs (miRNAs) and messenger RNAs (mRNA) from their parental cells to affect the recipient cells. Emerging evidence indicates that exosomes also play an important role in the intercellular communication among the bone cells, that is, osteoblasts, osteoclasts and bone marrow stromal cells, within bone microenvironment. Here, we reviewed the local and systemic effects of bone-derived exosomes, and discussed the potential of exosomes as targeted delivery systems for molecular therapy in skeletal disorders.