ArticlePDF AvailableLiterature Review

Exosomes Immunity Strategy: A Novel Approach for Ameliorating Intervertebral Disc Degeneration

Frontiers
Frontiers in Cell and Developmental Biology
Authors:

Abstract and Figures

Low back pain (LBP), which is one of the most severe medical and social problems globally, has affected nearly 80% of the population worldwide, and intervertebral disc degeneration (IDD) is a common musculoskeletal disorder that happens to be the primary trigger of LBP. The pathology of IDD is based on the impaired homeostasis of catabolism and anabolism in the extracellular matrix (ECM), uncontrolled activation of immunologic cascades, dysfunction, and loss of nucleus pulposus (NP) cells in addition to dynamic cellular and biochemical alterations in the microenvironment of intervertebral disc (IVD). Currently, the main therapeutic approach regarding IDD is surgical intervention, but it could not considerably cure IDD. Exosomes, extracellular vesicles with a diameter of 30–150 nm, are secreted by various kinds of cell types like stem cells, tumor cells, immune cells, and endothelial cells; the lipid bilayer of the exosomes protects them from ribonuclease degradation and helps improve their biological efficiency in recipient cells. Increasing lines of evidence have reported the promising applications of exosomes in immunological diseases, and regarded exosomes as a potential therapeutic source for IDD. This review focuses on clarifying novel therapies based on exosomes derived from different cell sources and the essential roles of exosomes in regulating IDD, especially the immunologic strategy.
Content may be subject to copyright.
Exosomes Immunity Strategy: A Novel
Approach for Ameliorating
Intervertebral Disc Degeneration
Weihang Li
1
, Shilei Zhang
1
, Dong Wang
1
,
2
, Huan Zhang
1
, Quan Shi
1
, Yuyuan Zhang
3
,
Mo Wang
4
, Ziyi Ding
1
, Songjie Xu
5
*, Bo Gao
1
* and Ming Yan
1
*
1
Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xian, China,
2
Department of Orthopaedics,
Afliated Hospital of Yanan University, Yanan, China,
3
Department of Critical Care Medicine, Xijing Hospital, Air Force Medical
University, Xian, China,
4
The First Brigade of Basic Medical College, Air Force Military Medical University, Xian, China,
5
Beijing
Luhe Hospital, Capital Medical University, Beijing, China
Low back pain (LBP), which is one of the most severe medical and social problems
globally, has affected nearly 80% of the population worldwide, and intervertebral disc
degeneration (IDD) is a common musculoskeletal disorder that happens to be the primary
trigger of LBP. The pathology of IDD is based on the impaired homeostasis of catabolism
and anabolism in the extracellular matrix (ECM), uncontrolled activation of immunologic
cascades, dysfunction, and loss of nucleus pulposus (NP) cells in addition to dynamic
cellular and biochemical alterations in the microenvironment of intervertebral disc (IVD).
Currently, the main therapeutic approach regarding IDD is surgical intervention, but it could
not considerably cure IDD. Exosomes, extracellular vesicles with a diameter of 30150 nm,
are secreted by various kinds of cell types like stem cells, tumor cells, immune cells, and
endothelial cells; the lipid bilayer of the exosomes protects them from ribonuclease
degradation and helps improve their biological efciency in recipient cells. Increasing
lines of evidence have reported the promising applications of exosomes in immunological
diseases, and regarded exosomes as a potential therapeutic source for IDD. This review
focuses on clarifying novel therapies based on exosomes derived from different cell
sources and the essential roles of exosomes in regulating IDD, especially the
immunologic strategy.
Keywords: exosome, intervertebral disc degeneration, immunologic therapy, vascularization, low back pain
INTRODUCTION
Low back pain (LBP), one of the most severe medical and social problems globally, together with the
causes of complete disability in middle-aged or older adults, has affected nearly 80% of the
population worldwide. It is the most common cause of limited activity in patients younger than
45 (Taylor et al., 1994;Andersson, 1999;Millecamps et al., 2015). Dieleman et al. (2020) reported that
the total cost of back pain, neck pain, and other musculoskeletal disorders comprised a great
proportion of expenditures from 1996 to 2016approximately $264 billion, which leads to
considerable nancial burdens on both society and families of affected individuals. IDD
(intervertebral disc degeneration) is the main cause of LBP, and it occurs frequently in adults. It
is a common musculoskeletal disorder; the progression results in disc herniation, spinal canal
stenosis, and degenerative spondylolisthesis (Jin et al., 2013). While the exact etiology and
degenerative mechanisms remain delineated, existing studies have discovered several factors
Edited by:
Zhongyang Liu,
Chinese PLA General Hospital, China
Reviewed by:
Qizhao Huang,
Southern Medical University, China
Yonggao Mou,
Sun Yat-sen University Cancer Center,
China
Lei Ma,
Third Hospital of Hebei Medical
University, China
Qicong Shen,
Naval Medical Universtity, China
*Correspondence:
Songjie Xu
stu_xusj@163.com
Bo Gao
gaobofmmu@hotmail.com
Ming Yan
yanming_spine@163.com
These authors have contributed
equally to this work and share rst
authorship
Specialty section:
This article was submitted to
Molecular and Cellular Pathology,
a section of the journal
Frontiers in Cell and Developmental
Biology
Received: 25 November 2021
Accepted: 21 December 2021
Published: 10 February 2022
Citation:
Li W, Zhang S, Wang D, Zhang H,
Shi Q, Zhang Y, Wang M, Ding Z, Xu S,
Gao B and Yan M (2022) Exosomes
Immunity Strategy: A Novel Approach
for Ameliorating Intervertebral
Disc Degeneration.
Front. Cell Dev. Biol. 9:822149.
doi: 10.3389/fcell.2021.822149
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221491
REVIEW
published: 10 February 2022
doi: 10.3389/fcell.2021.822149
involved in the initiation and progression of IDD, including
aging, loading changes, poor nutrient supply, smoking, and
hereditary aspects (Adams et al., 2000;Mannion et al., 2000;
Freemont et al., 2001;Horner and Urban, 2001;Vogt et al., 2002;
Bibby and Urban, 2004;Wang et al., 2012). Although etiology is
likely multifactorial, mounting lines of evidence have pointed that
genetic factor is considered as a pivotal risk of IDD, which
accounts for more than 70%, with smaller contributions from
environmental factors (Battié et al., 1995;Bijkerk et al., 1999;
Sambrook et al., 1999;Kepler et al., 2013). The pathological basis
of IDD includes the disorders of catabolism and anabolism in the
extracellular matrix (ECM), continuous decrease in nucleus
pulposus (NP) cells, and cellular and biochemical alterations
in the microenvironment of intervertebral disc (IVD) (Wang
et al., 2020a;Binch et al., 2021).
IVD, located between adjacent vertebrae, are
brocartilaginous tissues and allow motion between vertebral
bodies; they provide load support, exibility, energy storage, and
consumption in the spine (Ji et al., 2018). It is a complex avascular
organ consisting of the central NP, peripheral annulus brosus
(AF), which envelops the NP, and the upper and lower cartilage
endplates (EP) (Le Maitre et al., 2007). A healthy NP is gelatinous
and is primarily made of proteoglycans (glycosaminoglycan),
type II collagen, and NP cells, while the peripheral AF is a
thick, dense structure, and it is composed of type I collagen
and AF cells; EPs seal the disc, which are cartilaginous structures
that resemble the hyaline cartilage (Kadow et al., 2015). These
distinct anatomical areas formed a special complex structure,
giving it unique biomechanical properties to maintain spinal
exibility and mechanical stability (Lam et al., 2011). Mature
IVD is made up of avascular tissues, which are inundated with
extensive ECM; the blood supply through peripheral capillaries is
rather limited, and nutrient supply could only be received from
passive diffusion from the EPs (Boubriak et al., 2013). So, it is
easily understood that IVD is prone to degeneration.
Currently, the therapeutic approaches regarding IDD mainly
include conservative treatment and surgical intervention.
Conservative treatment may not alleviate the patientspain
immediately; patients could only maintain a normal life
mainly through oral painkillers. Surgical interventions attempt
to relieve symptoms rather than restore inherent structure and
function. Discectomy is the most common spinal surgical
treatment of IDD and typically performed in young patients
about 2540 years old, while the impact of the alteration in
biomechanics and long-term sequelae may be signicant
(Hermantin et al., 1999;Yorimitsu et al., 2001). Brinckmann
et al. found that loss of disc tissue resulted in a decrease in disc
height and intradiscal pressure, and an increase in radial disc
bulge (Brinckmann and Grootenboer, 1991). Scoville and Corkill
(1973) reported a 50% incidence of narrowing after disc surgery
at a 3-month follow-up. Tibrewal and Pearcy (1985) also found a
signicant narrowing of disc space following disc surgery,
compared to non-operated controls. Patients suffer relapse
after treatment, and the disk degeneration may even accelerate
the degeneration of adjacent segments (Hashimoto et al., 2019),
so recurrent disc herniation might be an inevitable issue for
surgeons and patients; both oral pills and surgical methods could
not considerably cure IDD. Consequently, further explorations
about more effective IDD treatment approaches are of great
signicance. Studies have reported that the ideal strategy for
disc regeneration is to restore the functions as well as the integrity
of disc, including biomaterials therapy, native matrices
supplement, mesenchymal stem cell (MSC) therapy, growth
factors therapy, tissue engineering technology,
immunotherapy, and exosome therapy (Longo et al., 2012;Jin
et al., 2013;Richardson et al., 2016;Bowles and Setton, 2017;
Cheng et al., 2018;Du et al., 2019;Sun et al., 2020a); these
methods are currently the most compelling research avenues for
IDD treatment.
Cellcell communication is an essential way to exchange
information between cells; paracrine signaling is the primary
means of cellular communication, while exosome secretion is a
special mechanism of paracrine regulation, which has been widely
considered for IDD therapy (Lu et al., 2017). In this review, we
focus on the novel approach of exosome therapy, to clarify the
roles in regulating the immunological and inammatory
pathological process of IDD. This review provides a reference
for elucidating the molecular mechanisms of exosomes in the
treatment of IDD as well as its application prospects.
EXOSOMES
Exosomes were rstly discovered in sheep reticulocytes by Pan
and Johnstone in 1893. During the maturation of sheep
reticulocytes, the release of transferrin receptors into ECM was
correlated with a type of small vesicle (Pan and Johnstone, 1983;
Pan and Johnstone, 1984); such extracellular vesicles (EVs) were
dened as exosomes in 1989 (Johnstone et al., 1989). In the last
decades, a series of EVs have been described, while the denition
of EVs remains confusing among different reports (Hunter et al.,
2008;Skog et al., 2008;Cocucci et al., 2009;Silva et al., 2011). Up
to now, the different kinds of EVs are differentiated based on their
size, content, and formation mechanisms; EVs mainly include
apoptotic bodies, microvesicles, and exosomes (Al-Nedawi et al.,
2009;Raposo and Stoorvogel, 2013), among which, apoptotic
bodies and microvesicles are generated from plasma membrane,
with a diameter of 8005,000 nm and 2001,000 nm, respectively
(Camussi et al., 2011;Livshits et al., 2015), and exosomes are
endogenous vesicles with a diameter of 30150 nm (Mathivanan
et al., 2010;Sampey et al., 2014;Zhang et al., 2019a). Since
exosomes have been rstly discovered, they have been thought of
as a cellular waste product, while in recent years, it has been found
that tiny membrane vesicles contain cell-specic proteins, lipids,
and nucleic acids, which can be delivered to other cells as signal
molecules to change functions or other cells. These ndings have
sparked interest in cell secretory vesicles.
The Formation of Exosomes
Exosomes, membrane-bound vesicles, with a diameter of
30150 nm, are presented in nearly all kinds of biological
uids; the existence of exosomes has already been found in
saliva, urine, semen, plasma, cerebral spinal uid, bronchial
uid, serum, amniotic uid, breast milk, bile, synovial uid,
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221492
Li et al. Different Exosomes for IDD Therapy
tears, lymph, and gastric acid (Caby et al., 2005;Akers et al., 2013;
Vojtech et al., 2014;Yoshida et al., 2014;Shi et al., 2015;
Zlotogorski-Hurvitz et al., 2015;Milasan et al., 2016;Yoon
and Chang, 2017;Li et al., 2018a;Dixon et al., 2018;Goto
et al., 2018;Yuan et al., 2018). Exosomes are initially formed
by endocytosis; the cell membrane is internalized to generate
endosomes and then many small vesicles are formed inside the
endosome by invaginating parts of the endosome membrane;
such vesicles are called multivesicular bodies (MVBs). Ultimately,
these MVBs fuse with the cell membrane, releasing the
intraluminal endosomal vesicles into extracellular space by
exocytosis to become exosomes (Heijnen et al., 1999;
Gruenberg and van der Goot, 2006). They are also usually
dened as intercellular communication vectors containing
bioactive substances, including cytokines, proteins, lipids,
mRNAs, miRNAs, non-coding RNAs, and ribosomal RNAs;
the lipid bilayer of the exosomes protect them from
ribonuclease degradation and help improve their biological
efciency in recipient cells.
The Identication of Exosomes
Generally, the existence of exosomes is authenticated by a series
of identications to conrm; authentication methods vary from
physical characteristics to surface molecular markers, including
transmission electron microscopy (TEM), nanoparticle tracking
analysis (NTA), and Western blot for molecular marker detection
(Dragovic et al., 2011;Kowal et al., 2017;Jung and Mun, 2018).
Through TEM detection, exosomes are visualized by electron
microscopy after negative staining; exosomes usually appear as
cup-shaped entities by transmission electron microscopy, but as
hemisphere-shaped entities by cryoelectronic microscopy. There
is a high degree of morphological diversity among exosomes
isolated from different kinds of body uids (Sahoo et al., 2011;
Höög and Lötvall, 2015). Using the NTA method, the Brownian
motion of individual vesicles is tracked, and their size and total
concentrations are calculated using NTA software. NTA could
measure cellular vesicles as small as 50 nm, which is far more
sensitive than conventional ow cytometry (lower limit is
300 nm). Besides, their phenotype could be quickly determined
by combining NTA with uorescence measurement (Dragovic
et al., 2011). From Western blotting analysis, exosome marker
proteins include a family of four-transmembrane proteins, such
as CD9, CD63, and CD81; cytoplasmic proteins like actin and
annexins; and molecules involved in biological functions,
including apoptotic transfer gene 2 interacting protein X
(Alix), tumor susceptibility gene 101 protein (TSG101), heat
shock protein (HSP70 and HSP90), and cell-secreted specic
proteins, among which CD9, CD63, HSP70, and TSG101 are
commonly used identication proteins for exosomes (Su et al.,
2019).
Communication of Exosomes
Cellcell communication is an essential way to exchange
information between cells, and exosomes play a pivotal role as
vehicle for carrying information. When exosomes are secreted
from host cells into recipient cells, they could regulate the
biological activities of recipient cells by transferring proteins,
nucleic acids, and lipids. Basically, exosome-mediated
intercellular communications mainly occur in three
mechanisms: First, the exosome membrane proteins could
interact with the receptors, to activate intracellular signaling
pathways of target cells (Munich et al., 2012). Second, in
ECM, exosome membrane proteins could be cleaved by
proteases, and the spliced fragments could act as ligands to
bind to receptors on the cell membrane, thus activating
intracellular signaling pathways (Tian et al., 2013). Third,
exosome membranes directly fuse with recipient cell
membranes, releasing their content such as proteins, mRNA,
and microRNA into the cytosol (Mulcahy et al., 2014).
Compared to traditional gene therapy vectors, exosomes could
protect and transfer bio factors as natural nanocarriers (van
Dommelen et al., 2012). In the clinic, the applications of
MSCs have been predominant due to their stronger functions
than other cells, such as proliferation and differentiation ability
in vitro (Yeo et al., 2013), and MSCs also generate a large amount
of exosomes, which have low immunogenic properties. Several
studies have shown that MSC exosomes may be more appropriate
than MSCs in stem cell-based therapies (Yuan et al., 2020;Krut
et al., 2021;Xing et al., 2021). Currently, numerous studies have
reported the signicance of exosomes in the treatment of IDD;
the biological composition and functions of exosomes are based
on different types of cells (Wang et al., 2021).
MicroRNAs (miRNAs) are a class of non-coding single-
stranded RNA with a length of less than 22 nucleotides.
Various studies have reported the associations between
miRNA levels and IDD, including proliferation and apoptosis
of NP cells, ECM regeneration, and inammation response (Jing
and Jiang, 2015;Li et al., 2015;Wang et al., 2015), and the
dysregulated miRNA expression is widely observed in IDD (Zhao
et al., 2014), which has essential roles in the progression of IDD,
and it attracts much attention in delivering exosome-derived
miRNAs in ameliorating IDD (Saravanan et al., 2019).
MECHANISMS OF EXOSOMES IN THE
TREATMENT OF IDD
In disc degeneration, the main pathological changes are
excessive degradation of ECM and reduction in AF and NP
cells. Since the intervertebral disc is a closed, avascular structure,
intervertebral injection is suggested to be an ideal method for
the treatment of IDD (Hu et al., 2020a). Exosomes have been
demonstrated to affect catabolism and anabolism of ECM by
inhibiting MMPs, and most exosomes play roles in IDD mainly
through releasing miRNAs (Zhang et al., 2019b); the
internalization of PKH67 (most detected)-labeled exosomes
into targeted cells indicates the involvement of exosomes in
modulating these changes. Thus, exosome therapy is a
promising therapeutic approach for IDD, which achieves its
therapeutic effects through continuous release of miRNAs,
proteins, and transcription factors that regulate metabolic
disorders, microenvironment, and cell homeostasis (Wang
et al., 2021). Existing studies have reported the application of
different resources of exosomes in the treatment of IDD; here,
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221493
Li et al. Different Exosomes for IDD Therapy
their main therapeutic mechanisms are illustrated, as shown in
Figure 1.
By Improving Immune Microenvironment
and Inammation Reactions
As the largest avascular organ in the body, IVDs are located
between vertebras, responsible for the sustainability, durability,
and exibility of the spine (Ji et al., 2018). NP cells are surrounded
by AF and EPs, and they are trapped in the IVD since the
formation of NP cells; this unique structure isolates NP tissues
from the immune system of the host, and thus, IVDs are
identied as an immune privileged organ (Sun et al., 2020a).
Studies have found that various ingredients of NP induce auto-
immune and inammation responses after exposure to the host
immune system during IDD (Capossela et al., 2014;Wang and
Samartzis, 2014), and antigenantibody complexes are
commonly present in herniated NP tissue (Satoh et al., 1999).
The recruitment of immunocytes may lead to deterioration of
IDD, through cellcell communication and cytokine secretion. As
for immunocyte types, activated T and B cells have been found to
be elevated by autologous NP subcutaneously in a pig model
(Geiss et al., 2007). Murai et al. (2010) have reported that
macrophages and NK cells may recognize autologous NP cells
and display positive cytotoxic effects, according to a comparison
between wild-type mice and immune-decient mice, and
plasmacytoid dendritic cells, along with few macrophages and
memory T cells, are also found in isolated and extruded discs
(Geiss et al., 2014). Capossela et al. (2014) have also provided
direct evidence of auto-immune response, showing that IgGs are
found specic to collagen type I, II, and V and aggrecan in human
degenerative IVD samples; these ndings suggest that
complicated immunocytes participate in auto-immune
response of NP tissues in different stages. Moreover,
inammatory factors are also increased in the development of
IDD. In an autograft model, Takada et al. (2012) have detected
the high expression of TNF-α, IL-6, IL-8, cyclooxygenase 2, and
macrophage inltration. Consequently, these reports elucidate
that with the damage of immune privilege in IVD, exposed NP
tissues could promote auto-immune response, which nally
leadstotheactivationofimmunocytesaswellasthe
inltration of inammatory factors. Previous research has
also reported the association between autophagy and
miRNAs, showing that dysregulation of the relationship
between autophagy and miRNAs may accelerate the aging
and apoptosis of NP cells (Akkoc and Gozuacik, 2020;Lan
et al., 2021). NP cells rstly activate or repress the expression of
specic miRNAs under stress condition, and then miRNAs
regulate autophagy level by directly targeting ATG and
signaling pathways to meet cellular demands (Zhou et al.,
2016). More studies need to be further conducted, including
the associations between exosomes and immunocytes, and
FIGURE 1 | Mechanisms of exosomes in the treatment of IDD.
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221494
Li et al. Different Exosomes for IDD Therapy
whether these immunocytes could secrete exosomes and play a
role in the treatment of IDD.
By Suppressing Vascularization
Vascularization is widely observed in IDD, which is considered to
play essential roles in the progression of IDD. Blood vessel invasion
would cause activation of immunocytes and inammatory factors,
increase neuralization, and thereby damage the dynamic balance in
IVD (Sun et al., 2020b). Existing studies have reported the pivotal
roles of exosomes in inhibiting the growth of blood vessels; Cornejo
et al. (2015) and Kwon et al. (2017) have found that soluble factors
from notochordal cellscould suppress endothelial cell invasion and
vessel formation by inhibiting the VEGF signaling pathway;
meanwhile, Sun et al. (2020b) have conrmed the nding that
notochordal cell-derived exosomes could suppress proliferation of
HUVECs; another research has also discovered the regulatory roles
of AF exosomes, showing that degenerative AF exosomes promote
migration of HUVECs and upregulate the expression of
inammatory factors (Sun et al., 2021). Furthermore, Liang
et al. (2017) have reported that the co-culture system between
PMSCs and endothelial progenitor cells (EPCs) enhances the
angiogenic potential of EPCs through PDGF and the Notch
signaling pathway (Komaki et al., 2017). Collectively, exosomes
play an important role in the regulation of vascularization, different
sources and cells may result in different outcomes, and detailed
information about vascularization is fully discussed below.
By Promoting Synthesis and Inhibiting
Decomposition of ECM
Due to the special structure of AF that consists of 99% ECM and
1% AF cells, the ECM is of great signicance to maintain the
avascular structure and homeostasis of IVD. Focal proteoglycan
loss has been noticed to cause alteration of ECM, which facilitates
the growth of nerves and blood vessels (Stefanakis et al., 2012).
Degenerative IVD possesses the common feature of signicant
loss of ECM, including COL2 and aggrecan. MMPs are chief
catabolic factors responsible for these pathological changes, and
the dysregulated expression of MMPs is widely observed and
could enhance deterioration in IDD progression, including
MMP-1, MMP-3, MMP-9, MMP-13, ADAMTS-4, and
ADAMTS-5 (Zhang et al., 2021a). Exosomes have been
demonstrated to affect the catabolism of ECM by inhibiting
MMPs (Cabral et al., 2018;Zhang et al., 2019b). Almost all
mechanisms ultimately promote regeneration of ECM and
inhibit ECM degradation for IVD regeneration. Different
nucleic acids transferred from exosomes exhibit their roles
through multiple transductions, aiming at MMPs to modulate
expression of ECM.
POTENTIAL SOURCES OF APPLICATIONS
BY EXOSOMES DURING THE
PROGRESSION AND TREATMENT OF IDD
A number of exosomes from different sources have been
reported, among which exosomes could be divided into two
major parts based on their properties: stem cell-derived
exosomes and non-stem cell-derived exosomes. They could be
further classied according to source, such as BMSC, PMSC,
USC, and ADSC exosomes from stem cell-derived exosomes, and
AF, NP, and NC exosomes from non-stem cell-derived exosomes.
This review aims to focus on the different sources of exosomes to
elucidate the detailed mechanisms in the treatment of IDD; the
whole mechanism is illustrated in Figure 2.
Stem Cell-Derived Exosomes
As a worldwide issue, the treatment of IDD based on stem cells
has been widely studied; the rationales of stem cell therapy are
considered as replenishing disc cells through multipotent
differentiation, promoting proliferation of NP cells, enhancing
immune privilege, and reducing apoptosis and anti-inammation
(Ma et al., 2015). Studies have already conrmed the potential
therapeutic efcacy of MSC transplantation in the treatment of
IDD, and by inheriting the characteristics of MSCs, exosomes
could also treat IDD through protecting NP cells from apoptosis,
mitigating the inammatory responses of disc, and promoting the
synthesis of ECM (Lu et al., 2017;Xing et al., 2021).
The usage of exosomes as a cell-free product has
continuously been regarded as substitute therapy against
stem cell transplantation; the applications of exosomes rather
than stem cells have advantages including low risk of
tumorigenesis, malformations, and microinfarctions;
convenience in collection and storage; increase of sustained
biological activity; stability; and minimal immunogenicity when
transplantation (Tao et al., 2018). Exosomes have been
demonstrated to affect the catabolism of ECM by inhibiting
MMPs (Cabral et al., 2018;Zhang et al., 2019b). Consequently,
exosome therapy is a novel promising therapeutic approach for
IDD; the efcacies are achieved from the continuous release of
microRNAs, proteins, and transcriptome factors to regulate
metabolic disorders, microenvironment, and cell homeostasis
(Xing et al., 2021). Different stem cell-derived exosomes are
highly specic in treating IDD; compared to traditional gene
therapy vector, exosomes could be considered as nanocarriers to
transfer specic molecules including miRNA, siRNA, or
AntagomiR to recipient cells through endocytosis and
membrane fusion (Zhou et al., 2016;van den Boorn et al.,
2011). The detailed mechanisms of MSC exosomes in the
treatment of IDD are discussed as follows:
Exosomes Derived From Bone Marrow Mesenchymal
Stem Cells
BMSCs derived from mesoderm are a type of stem cell with
multidirectional differentiation potential. BMSC transplantation,
which serves as a representative cell therapy, is becoming
prevalent in the eld of bone regeneration, cartilage repair,
spinal cord injury, and IDD (Sakai and Andersson, 2015;
Hejazi et al., 2021;Jiang et al., 2021;Maldonado-Lasunción
et al., 2021). However, stem cell therapy still has many
limitations such as difculty in obtaining cells, susceptibility to
aging, potential tumorigenesis, and immune rejection, which
greatly limit the application in the eld of regenerative
medicine (Dou et al., 2021).
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221495
Li et al. Different Exosomes for IDD Therapy
BMSC exosomes have advantages of low immunogenicity and
suitability for the IVD microenvironment, and it could provide
cell-free therapy instead of the traditional BMSC therapy. BMSC
exosomes have similar functions to BMSC, such as restoring
tissue damage, inhibiting inammation, and regulating immune
microenvironment. Existing research has reported that BMSC
FIGURE 2 | Potential sources of applications by exosomes in the treatment of IDD.
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221496
Li et al. Different Exosomes for IDD Therapy
exosomes could modulate the degenerative NP gene toward a
healthy NP gene phenotype (Lu et al., 2017). High levels of
inammatory factors, like TNF-α, have been demonstrated to
result in excessive apoptosis of NP cells and cause IDD
progression (Risbud and Shapiro, 2014), and growing lines of
evidence have indicated the roles of miRNAs in the progression of
IDD, showing that exosomes transport these miRNAs into IVD
(Lan et al., 2016). Cheng et al. (2018) showed that in an IDD rat
model, BMSC exosomes inhibited TNF-α-induced NP cell
apoptosis, and further analysis suggested that exosomes were
rich in miR-21 and inhibited NP cell apoptosis by specically
targeting PTEN through transporting miR-21 into NP cells. In a
study of degenerated and normal NP cells, Xia et al. (2019) have
found various proteins related to inammatory responses in IVD;
the exosomes suppress the progression of IDD by targeting the
activation of inammatory mediators NLRP3 in NP cells, which
is an effective therapeutic target of IDD. Zhu et al. (2020a) have
suggested highly expressed miR-532-5p in BMSC exosomes,
which could suppress the TNF-α-induced apoptosis, ECM
degradation, and brosis deposition in NP cells by delivering
miR-532-5p via targeting RASSF5. RASSF5, a major member of
RASSF5 family, could selectively trigger RAS and function as a
tumor suppressor, which is highly correlated with cell
proliferation, apoptosis, and tumorigenicity among many
neoplasms (Antoniou et al., 1996;Suryaraja et al., 2013).
MMP-13, a regulatory gene of ECM degradation in IDD, has
also been proven to be complementary with miR-532-5p
(Mohanakrishnan et al., 2018). They revealed two therapeutic
targets to attenuate IDD: miR-532-5p in BMSC exosomes could
decrease the apoptosis of NP cells by targeting RASSF5 and
inhibit ECM degradation by targeting MMP-13 in NP cells.
Autophagy is tightly connected to aging as well as apoptosis in
the pathogenesis of disorders, including cancer, osteoarthritis,
and degenerative diseases. Autophagy exists in degenerative disc
and is an effective approach in regulating ECM metabolism in
IVD (Zhang et al., 2021b). Related studies have elucidated that
BMSC exosomes promote proliferation of AF cells by inhibiting
expression of inammatory cytokines stimulated by IL-1β. The
PI3K/AKT/mTOR signaling pathway is activated by BMSC
exosomes, which is an essential signaling pathway regulating
autophagy (Li et al., 2020), and a previous study has also
conrmed the regulation of exosomes in the PI3K/AKT/
mTOR signaling pathway and that human umbilical cord
mesenchymal stem cell-derived exosomes inhibited apoptosis
of H9C2 cells via autophagy regulation (Liu et al., 2019).
Unfolded protein response (UPR) is an evolutionarily
conservative reaction induced by endoplasmic reticulum (ER)
stress; it is the most typical response among ER stress. When any
abnormal reaction of ER occurs, ER stress could be triggered to
protect the homeostasis of ER (Hetz, 2012). However, severe or
prolonged ER stress could hyperactivate UPR, which results in
excessive degradation of cellular proteins, and eventually leads to
cell demise (Kim et al., 2008). AGEs (advanced glycation end
products), also called the Millard reaction, are non-reducible
substances generated by the polymerization of sugars and
proteins through a series of reactions (Chan et al., 2016).
AGEs commonly accumulate in aging and degenerative
diseases, and are closely related to inammation, metabolic
dysfunction, and ER stress (Chiang et al., 2016). Both AGEs
and ER stress are highly correlated with IDD; higher levels of
AGEs are detected in degenerative disc, and AGEs are also
connected to Prrmann grades of IVD (Fields et al., 2015;
Song et al., 2018). Liao et al. (2019) have demonstrated that
according to an in vitro and rat model, BMSC exosomes reduce
AGE-induced ER stress, inhibit the activation of UPR, and
ameliorate NP cell apoptosis. AGE accumulations are reported
to induce ER stress, leading to cell apoptosis by induction of
prolonged UPR, and the accumulation of CHOP could promote
the cleavage of caspase-3 and caspase-12, resulting in cell
apoptosis (Adamopoulos et al., 2016;Go et al., 2017). Through
detecting the related signaling pathway AKT, ERK, and ER-
related CHOP protein, they elucidated that BMSC exosomes
signicantly activate AKT and ERK signaling, which reduced
expression of CHOP, then further inhibited caspase-3 and
caspase-12, and ultimately decreased the apoptosis of NP cells
and catabolism of ECM, thus preventing IDD.
The mitogen-activated protein kinase (MAPK) family consists
of ERK1/2, JNK, and p38 MAPK proteins, and studies show that
ERK is mediated to cell proliferation and inammation (Sawe
et al., 2008); p38 MAPKs are a type of proinammatory mediator,
and p38 MAPKs and JNK both modulate cell apoptosis and
inammation (Yang et al., 2016;Lai et al., 2019). Increasing lines
of evidence have shown that the activation of MAPK transduction
is activated and tightly correlated to ECM degradation, cell aging,
apoptosis, and inammatory reactions in IDD (Zhang et al.,
2021b). MLK3 belongs to serine/threonine MAPK kinase
(MAP3K) and is aberrantly expressed in mammals, which
mediates cell migration and invasion in various diseases
(Zhang et al., 2014a;Lan et al., 2017;Misek et al., 2017). Zhu
et al. (2020b) have revealed the relationships between miR-142-
3p and IDD, that miR-142-3p is overexpressed in BMSC
exosomes and miR-142-3p excreted from BMSC exosomes
could ameliorate NP cell injury via targeting MLK3, which
further inhibits the activation of the MAPK signaling pathway.
They have proved that exosomal miR-142-3p and the subsequent
MLK3/MAPK cascade transduction could be as an effective
approach in attenuating the progression of IDD.
Exosomes From Urine-Derived Stem Cells
Stem cells are regarded as ideal cells for IVD degeneration, since
they could prevent IVD tissues from aging and apoptosis. The
applications of exosomes from BMSC are widely used in various
diseases including IDD, while the limited sources of BMSC,
expensive costs to obtain BMSC, and the physical trauma in
the acquisition process (Sakai and Andersson, 2015) prompt
researchers to nd safer, less expensive, and more effective
exosomes from other sources. Studies have discovered that a
subpopulation of cells isolated from urine have similar
characteristics to BMSCs, such as multidirectional
differentiation capacity, clonogenicity, expansion patterns, self-
renewal capacity, and paracrine properties (Bodin et al., 2010;Wu
et al., 2011a;Bharadwaj et al., 2011;Wu et al., 2011b). These cells
are thereby named urine-derived cells or USCs. Compared to
BMSCs, USCs possess several merits: rst, they could be collected
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221497
Li et al. Different Exosomes for IDD Therapy
through a simple, safe, low-cost, and non-invasive manner;
second, the acquisition procedures do not violate ethics; last,
USCs have a low cost of culture and a faster proliferative rate;
besides, they could avoid immunological rejection when using
autologous therapy (Qin et al., 2014;Pavathuparambil Abdul
Manaph et al., 2018). Therefore, USCs, as a novel cell source, may
provide a more effective way in the amelioration of IDD.
IVD tissues are subjected to different levels of mechanical
stress in daily work and life, which have essential roles in spinal
biomechanics. Wang et al. (2013) and Gawri et al. (2014) have
discovered that excessive mechanical stress induces cell apoptosis,
promotes ECM-degrading enzymes, and nally leads to IDD. As
mentioned above, certain levels of UPR in ER stress could prevent
cells from external stimulus and restore cell homeostasis. Proper
biomechanical loads play pivotal roles in the structure and
function of articular cartilage, while abnormal and continuous
biomechanical stimulation could lead to accumulation of
misfolded proteins in ER lumen, resulting in continuous ER
stress and then cell apoptosis (Wang and Kaufman, 2016;
Hunt et al., 2020;Yang et al., 2020). Xiang et al. (2020) have
discovered the therapeutic application of USC exosomes in IDD
under high mechanical loads. They have elucidated that USC
exosomes suppress excessive activation of UPR, cell apoptosis,
and disc degeneration through AKT and ERK signaling pathways,
which is consistent with the above nding that AKT/ERK
transduction is activated in BMSC exosomes and behave in
AGE-induced ER stress (Liao et al., 2019). These ndings
imply that AKT/ERK signaling pathways mediated by
exosomes play active roles in AGE-induced or biomechanical
load-induced IDD, leading to caspase reduction by inhibiting
CHOP protein, and thereby reducing ECM degradation and NP
cell apoptosis. Consequently, inhibiting ER stress induced by
AGEs via BMSC or USC exosomes may be a potential therapeutic
target in the treatment of IDD. However, the type of nucleic acids
exosomes carry to mediate the AKT/ERK signaling pathway still
needs to be further analyzed.
COL2 and proteoglycan (chiey aggrecan, namely, ACAN) are
considered as crucial ECMs for discs to maintain their proper
functions, especially for NP cells (Vo et al., 2013). Guo et al.
(2021) have suggested that the methods of rebalancing disordered
COL2 and ACAN and increasing the synthesis is regarded as a
major point for ameliorating IDD progression. They have found
that USC exosomes could induce cell proliferation of NP cells and
ECM synthesis. TGF-βis a multifunctional cytokine, which
modulates cell fate and plasticity, and MATN3 could be
directly bound to specic integrins, promoting dissociation
and activation of TGF-βand affecting downstream gene
activation (Pullig et al., 2002). Lines of evidence have
demonstrated that multiple cellular reactions induced by TGF-
βare modulated via a canonical SMAD pathway and
noncanonical pathways like PI3K/AKT transduction (Goc
et al., 2011;Hamidi et al., 2017). Guo et al. (2021) have
conrmed that USC exosomes are found to be rich in
MATN3 protein, and exosomal MATN3 could ameliorate IDD
progression by activating the TGF-β/SMAD pathway to promote
the expression of COL2 and ACAN in ECM and NP cells, and by
triggering the TGF-β/PI3K/AKT pathway to function in cell
proliferation and antisenescence (Feng and Qiu, 2018).
Exosomes Derived From Human Placental
Mesenchymal Stem Cells
As mentioned above, although BMSCs are regarded as a gold
standard among other MSCs, the difculty and efcacies in
obtaining BMSCs have forced scientists to nd novel and
multifunctional sources. Among them, the placenta has
attracted much attention as an alternative source of BMSCs,
due to its abundance and ease of availability (Mathew et al., 2020).
It has been reported that a large amount of PMSC could be
collected from a small tissue chunk (In t Anker et al., 2004;
Parolini et al., 2008), making the acquisition of PMSC easier,
together with its exosomes. As a transient materno-fetal organ,
the placenta is disposed of after delivery and involves non-
invasive procedures, making it a more convenient source
(Mathew et al., 2020;Gorodetsky and Aicher, 2021). The
applications of PMSC exosomes have been reported to affect
osteogenic and adipogenic differentiation by regulating OCT4
and NANOG in dermal broblasts (Tooi et al., 2016). The main
advantage of PMSC exosome-based therapies appears to be the
secretion of a wide range of anti-inammatory and pro-
regenerative factors, which makes it possible for PMSC
exosomes to treat IDD.
Yuan et al. (2020) have discovered the applications of PMSC
exosomes in IDD; they have found that miR-4450 is highly
expressed in degeneration disc and PMSC exosomes have
therapeutic effects on NP cells, and by inhibiting miR-4450
and thus upregulating downstream gene ZNF121 to alleviate
apoptosis, inammation, and necrosis of NP cells, it could
even improve gait abnormality in vivo caused by IDD.
ZNF121 has been demonstrated to be the target of miR-4450;
as one of the biggest families of regulatory proteins in human
cells, ZNF121 plays an essential role in the development and
differentiation of various diseases (Ladomery and Dellaire, 2002).
It is modulated by multi-miRNAs and causes diseases including
breast cancer, cell proliferation of childhood neuroblastoma
(miR-1427), and invasion of gastric cancer (miR-204-5p) (Luo
et al., 2016;Wu et al., 2018;Huan et al., 2019). In addition to
nding that PMSC exosomes could directly inhibit miR-4450 and
then upregulate ZNF121, and thus ameliorate the apoptosis of NP
cells and IDD progression, Yuan et al. also successfully delivered
AntagomiR-4450 into exosomes, which made it more effective
than exosomes, further verifying the efcacy of PMSC exosomes
in treating IDD alone and their natural merits as an
oligonucleotide carrier. The method of using the direct role of
siRNA has been considered as an excellent therapeutic approach,
while its low bioavailability limits the full realization of its clinical
potential (El Andaloussi et al., 2013). These ndings imply that
the containing of oligonucleotide-based vesicles or drugs opens a
novel avenue for target therapy and that PMSC exosomes could
be regarded as nanocarriers to deliver specic molecules, like
siRNA or AntagomiR, and then transport the contents to target
cells through membrane fusion or endocytosis (Zhou et al., 2016;
Yuan et al., 2020).
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221498
Li et al. Different Exosomes for IDD Therapy
Although PMSC exosomes could be regarded as a vector that
delivers miRNAs, its vascularization characteristic is not suitable
for treating IDD, due to the avascular structure of IVD. The
detailed mechanisms between neovascularization and exosomes
would be discussed later.
Exosomes From Adipose-Derived Mesenchymal Stem
Cells
Like USCs and PMSCs, ADSCs also come from a wide range of
sources and the damage to people is negligible when collected
from the human body (Xing et al., 2021); besides, the applications
of ADSC exosomes have been conrmed to accelerate the
proliferation and inhibit apoptosis of target cells, and they
have anti-inammatory effects in the treatment of ruptured
tendon (Shi et al., 2018;Zhang et al., 2021c), which suggests
their potential ability in degenerative diseases like IDD. Studies
have obtained exosomes from ADSC immobilized on ECM
hydrogels and then successfully construct an injectable
thermosensitive hydrogel system through mutual crossing of
ADSC exosomes and ECM hydrogels to restore the
microenvironment as well as pyroptosis of IDD (Xing et al.,
2021). They have conrmed that ADSC exosomes could regulate
the expression of MMPs directly, inhibit the catabolism of ECM,
and thus promote the accumulation of aggrecan and COL2.
Besides, ADSC exosomes could also inactivate the
inammatory factor NLRP3, retard their release, and then
affect the pyroptosis and survival of NP cells. Compared to
traditional exosome release, dECM (decellularization ECM)
exosome combination possesses several merits, such as
preventing immune response, high load rate, slow release of
exosomes, and prolonging their activation time. The
researchers established dECM to load engineered exosomes to
delivery drugs into IVD and NP cells, and eventually enhance the
drug effects of small molecules.
Besides, ADSC exosomes have also been found to activate the
SMAD signaling pathway in tendon healing (Liu et al., 2021a),
which is consistent with the above ndings that SMAD
transduction is upregulated by BMSC and USC exosomes.
Thus, further analysis should be expanded about the SMAD
signaling pathway via ADSC exosomes in the treatment of
IDD, and the correlations between SMAD and other different
sources of exosomes should also be further observed.
Non-Stem Cell-Derived Exosomes
In addition to the advantages of stem cell-derived exosomes
mentioned above, they also possess disadvantages: they may
cause hypertrophic differentiation of newly formed tissues,
together with unexpected angiogenesis (Chen et al., 2018),
which are the limitations in such closed and avascular IVD.
However, human cartilage cell-derived exosomes have been
found to facilitate chondrogenesis of cartilage progenitor cells
without such deciency (Chen et al., 2018), and normal
chondrocytes are located in a similar avascular and closed
environment and share many common features like NP cells
(Rosenthal et al., 2015). Additionally, existing studies have
reported that exosomes from non-stem cells like NP cells
could promote the migration of MSC cells and induce MSC
into IVD differentiation (NP-like phenotype) (Chen et al., 2018);
AF-derived exosomes could also have the same functions. At
present, non-stem cell-derived exosomes that could ameliorate
IDD progression are from NP, AF, and notochordal cells; the
specic mechanisms of these exosomes are discussed as follows:
Exosomes Derived From Nucleus Pulposus
Recent studies of exosomes regarding IDD mainly focus on MSC-
derived exosomes, while the application of NP cells has not been
widely reported yet. Zhang et al. (2021a) have discovered that NP
cells could secrete exosomes and deliver miR-27A to prevent
ECM degradation by targeting MMP-13. Autophagy activation
could promote release of NP exosomes and thereby prevent the
NP cell matrix from degradation, and it also ameliorates
degeneration of IVD, at least partly via exosomal miR-27A,
which targets and inhibits MMP-13 expression. In an in vitro
mutual experiment, Lu et al. (2017) have elucidated the
interaction roles between NP and BMSC cells via exosomes,
that BMSCs could spontaneously migrate across the transwell
membrane to IVD in a dose-dependent manner induced by NP
exosomes, and NP exosomes could induce BMSC differentiation
toward NP-like lineage, suggesting that NP exosomes could
recruit BMSCs and induce BMSC multidirectional
differentiation, and then replenish IVD cells and appropriate
ECM. Besides, they have demonstrated that NP exosomes are
more effective in inducing BMSCs to differentiate toward NP-like
cells than an indirect coculture system of BMSCs and NP cells.
Strassburg et al. (2012) also reported that the formation of gap
junctions and cell fusion are not the predominant mechanisms of
interaction; intercellular transfer of membrane components is the
main interactive mechanism between BMSCs and NP cells. These
ndings indicate that direct injection of BMSCs into IVD is not
effective compared to injection of NP exosomes into IVD, which
is consistent with the nding of Sun et al. (2021) that indirect
cellcell communication and paracrine are predominant in
keeping the avascular condition of IVD. However, the detailed
interactive mechanisms between BMSC and NP cells remain
unclear, and more specic components like proteins, miRNAs,
transcription factors, and lipids are expected for further research.
Furthermore, interactions between NP cells or NP exosomes and
other cells in IVD (such as AF cells and cartilage EP cells) should
be further analyzed (Hampton et al., 1989) to reveal their roles in
the progression of IDD.
Continuing with autophagy mentioned above, autophagy is
considered as a protective procedure for cell survival under stress
situations; in such process, excessive proteins or aging organelles
in cells are degraded to provide extra energies and, in most cases,
restore homeostasis (Mizushima, 2009). Highly conservative
serine/threonine kinase, as a mechanistic target of rapamycin
(mTOR, also known as mammalian target of rapamycin), is a
crucial cell growth regulatory cytokine that connects cellular
metabolism and growth with multiple environment inputs
(Kim and Guan, 2019). mTOR as a core component
autophagic pathway could negatively regulate autophagy.
Several studies have implied that autophagy activation could
maintain the balance of NPC, ECM, and vitality under
inammation (Jiang et al., 2013;Li et al., 2019). Serving as an
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 8221499
Li et al. Different Exosomes for IDD Therapy
effective agonist of autophagy and immunosuppressor, the
application of rapamycin has been demonstrated to increase
the release of NP exosomes, upregulate expression of miR-
27A, and target MMP-13 to prevent ECM degradation.
Rapamycin may also overcome the limitation that non-stem
cell exosomes are difcult to collect and reproduce (Zhang
et al., 2021a). Moreover, NP exosomes have been found to be
secreted in an autophagy-dependent manner, and rapamycin has
been conrmed to stimulate NP cell release exosomes via the
RhoC/ROCK2 signaling pathway (Hu et al., 2020b).
Consequently, these ndings prove that NP exosomes also
have high therapeutic potentials and provide us with novel
insights into the usage of NP exosomes via rapamycin
activation in the treatment of IDD.
NP cells also secrete different numbers of exosomes in
different degenerative grades; exosomes secreted from high
degenerative levels of NP cells could promote the apoptosis
and inhibit the proliferation of NP cells (Song et al., 2020),
indicating a positive correlation between the functions of
degenerative exosomes and IDD progression, as well as the
duality of exosome application. TGF-βis abundant in NP
exosomes, which is well studied to transform BMSCs into NP-
like cells (Steck et al., 2005), suggesting the direct functions of NP
exosomes to BMSCs and the ability to carry specic genes
through NP exosomes. Circular RNAs (circRNAs) are a class
of covalently closed non-coding RNA molecules generated by
reverse splicing of the exon of precursor mRNA in eukaryotes (Li
et al., 2018b); studies have shown the tight connections between
circRNAs and IDD (Wang et al., 2018). A rat model in Song et al.
(2020) has suggested that circRNA_0000253 is highly expressed
in IDD rat with positive correlations; they could target and absorb
miRNA141-5p, in a negative manner with IDD, and thus
downregulate the expression of Sirt1. Since the continuous
release of exosomes, the degenerative exosomes from NP cells
may secrete and further affect neighboring normal tissues and
thus aggravate the progression of IDD. As a result, there is an
urgent need to discover new approaches targeting degenerative
NP cells, among which circRNA_0000253 and miRNA141-5p
have been demonstrated to be effective, through delivering
siRNA-circRNA_0000253 or mimic-miRNA141-5p into
degenerative NP cells to prevent ECM degradation. Related
signaling pathways are circRNA_0000253/miRNA141-5p/Sirt1.
Given the multiple functions and multiple advantages of
exosomes, exosomes alone or with specic genes or drugs
would be a suitable choice for cell-free strategies in the
treatment of IDD.
Sirt1 (Sirtuin 1) is a nicotinamide adenine dinucleotide
(NAD+)-dependent histone deacetylase, which could reduce
apoptosis in different cells and correlated with various diseases
including cancer, metabolic disorders, COPD, and aging-related
disorders like degenerative and cardiovascular diseases (Dai et al.,
2018). Sirt1 participates in many pivotal cellular biological
processes, such as inammatory response, oxidative stress, and
mitochondrial functional homeostasis, which are consistent with
the mechanisms in IDD progression in that Sirt1 plays a
protective role in senescence and apoptosis of NP cells (Guo
et al., 2017;Zhang et al., 2019c). Guo et al. (2017) have found that
Prrmann grade is negatively correlated with Sirt1 expression in
IDD. In vitro experiments have veried that resveratrol and
quercetin could promote cell proliferation and senescence-
related protein expression (Guo et al., 2017;Wang et al.,
2020b). Shen et al. (2016) and He et al. (2019) have shown
that Sirt1 prevents NP cells from apoptosis via TLR2/Sirt1/NF-kB
transduction, and Sirt1 could ameliorate oxidative stress-induced
senescence of NP cells regulated by the Akt/FoxO1 pathway; a
previous study has also reported that Sirt1 could modulate
autophagy, which may further mediate the status of IVD
(Wang et al., 2020b). These observations imply that Sirt1
could serve as a novel therapeutic target in the treatment of
IDD, and the applications of exosomes mediating Sirt1 have
already been demonstrated in some diseases like brain injury,
photodamage, and neuronal autophagy (Chen et al., 2020;Liu
et al., 2021b;Wu et al., 2021). To the best of our knowledge, the
applications of Sirt1 via exosomes in the treatment of IDD have
not been reported; further relevant studies could focus on
exosomes and Sirt1 to reveal detailed mechanisms.
In terms of osteoarthritic cartilage, osteoarthritic cartilage and
degenerative IVD possess the common feature of signicant loss
of ECM, including COL2 and aggrecan; MMPs are chief catabolic
factors responsible for these pathological changes. Recent studies
have discovered that the applications of autophagy agonists like
rapamycin, chloramphenicol, and ozone inhibit MMP-13
expression and thus attenuate OA processes (Zhao et al., 2018;
Ma et al., 2019;Wu et al., 2019). A previous study has reported the
association between autophagy and exosome release in
chondrocytes, that they depend on caspase-3 and Rho/ROCK
transduction (Rosenthal et al., 2015); these ndings are consistent
with the autophagy-activated signaling pathway of exosomes
secreted from NP cells mentioned above. From inverted phase
contrast microscopy, NP cells are presented as polygonal shape,
and toluidine blue staining suggests that there are a large number
of notochord cells in IVD. Further IHC analysis also shows the
abundance of COL2; these results all conrm that NP cells
possess cartilage-like characteristics (Zhang et al., 2021a).
These results elucidate the tight connections between
chondrocytes and NP cells, based on the similar features of
chondrocytes and NP cells; the wide research about
chondrocytes and subsequent research regarding NP cells
could focus on the interactions with chondrocytes, to nd
novel ideas for treating IDD.
Exosomes Derived From Annulus Fibrosis
AF, the cells enclosing NP, consists of concentric layers composed
of alternatively aligned oblique collagen bers (mainly type I
collagen) interspersed with AF cells (Kadow et al., 2015), which
serve as a physical barrier to separate the internal NP and external
blood vessels and immune system. Due to the special structure of
AF that consists of 99% ECM and 1% AF cells, the indirect
cellcell contact or paracrine of AF cells may play pivotal roles in
maintaining avascular conditions in IVD. Vascularization has
been widely observed in IDD and has been considered as a
pathological feature, which mainly occurs in AF tissue, and
blood vessels grow and inltrate inwards, could cause NP
tissue exposure to immune system, and thereby damage
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214910
Li et al. Different Exosomes for IDD Therapy
immune privilege of IVD (Kim et al., 1981;Di Martino et al.,
2013). Sun et al. (2021) have veried that the exosomes could be
released by both degenerated and non-degenerated AF cells; they
could also be internalized by HUVECs, and degenerative AF
exosomes could enforce HUVEC migration faster than non-
degenerative AF exosomes, elucidating that degenerative AF
cells could induce IVD vascularization via exosome-mediated
effects, while non-degenerative AF tissues could suppress blood
vessels ingrowth as a physical barrier, and its exosomes could also
be regarded as an angiogenesis inhibitor to maintain the healthy
avascular condition of IVD. These results imply that AF
exosomes could serve as one of the mediators in intercellular
communication of IVD and modulate the vascularization
of IVD.
Within the region of IVD, the position of AF and blood vessels
is very close to each other, which may hypothesize the interactive
potential effects between AF and vascular endothelial cells. Pohl
et al. (2016) have discovered that endothelial microparticles
secreted from vascular endothelial cells enhance MMP
expression in AF cells, suggesting that the vascular endothelial
cells could act on AF cells through microparticle delivery, and it
could help blood vessel ingrowth into IVD by promoting ECM
catabolism. Together with the ndings that degenerative AF cells
affect vascularization of vascular endothelial cells by delivering
AF exosomes (Sun et al., 2021). Both AF and vascular endothelial
cells secrete microparticles like exosomes, and they endocytose
microparticles secreted from each other, and these reports all
display the essential roles of microparticles/exosomes in AF and
blood vessel communication. Besides, degenerative AF exosomes
could lead to upregulation of IL-6, TNF-α, MMP-3, and MMP-13
as well as VEGF, which are consistent with previous ndings that
these factors could cause NP cell apoptosis and IDD progression,
and these indicators could induce the inammation of IVD.
VEGF is a major proangiogenic factor that could trigger the
growth, expansion, and relocation of endothelial cells, and play
essential roles in vascularization of IVD (Capossela et al., 2018).
These ndings suggest the roles of degenerative AF exosomes; if
situations are not promptly treated and intervened, IVD
progression would produce a vicious cycle by degenerative AF
exosomes, leading to further development of IDD.
Up to now, the existence and functions of AF exosomes have
been rarely reported; although the interactions between AF
exosomes and endothelial cell HUVECs have been veried, the
relevant bioactive substances in AF exosomes related to these
effects have not been discovered, and more research should be
conducted to explore the potential molecular mechanisms and
signaling pathways. Besides, only one kind of vascular-related
cellsHUVECshave been conducted to analyze the
interactions with AF exosomes, while other vascular cells, like
vascular smooth muscle cells, or arterial endothelial cells, have
not been fully understood. Furthermore, due to the special
position of AF and NP cells, whether AF have interactions
with NP via exosomes still need to be further analyzed.
Consequently, the application of AF exosomes is a promising
research orientation whether in IDD or other elds, and these
issues need to be followed up and resolved by researchers.
Exosomes Derived From Notochordal Cells
Notochord is an embryonic structure of chordates; during the
embryogenesis period, the rod-shaped notochord is enclosed
from the vertebral body and develops to the NP tissue soon in
the early stage of fetal life (Cornejo et al., 2015), and blood vessels
recede and vanish slowly from IVD. In human IVD of immature
individuals (embryonic, fetal, and juvenile), NP tissues are mainly
populated by large vacuolated notochordal cells, while IVDs in
adult are populated with small and non-vacuolated chondrocyte-
like cells (Bach et al., 2018), and with the aging of humans, early
IDD begins to happen with the disappearance of NC cells (Risbud
and Shapiro, 2011). Several in vivo experiments have veried the
important roles in maintaining homeostasis of IVD (Bergknut
et al., 2013) and that NC loss coincides with the onset of IDD.
Increasing lines of evidence have also concluded the roles of NC
in the development and functions of IVD: NC could stimulate
proliferation of degenerative NP cells as well as BMSCs (de Vries
et al., 2015;Bai et al., 2017), and it may stimulate chondrogenic
differentiation, reducing natural cell necrosis. It has also been
demonstrated to inhibit angiogenesis and maintain the avascular
state of human IVD (Gruber et al., 2006;Mehrkens et al., 2017).
In conclusion, the functions of NC indicate the essential effects
and the potential roles in maintaining the NP tissues and IVD
healthy, and therefore, NC could be regarded as a promising
source for regenerative and symptom amelioration for IVD
disease.
The existence of NC exosomes has rstly been conrmed by
Sun et al. (2020b); they have found the best appropriate
mechanical stress to stimulate the release of NC exosomes,
and that NC exosomes could be internalized by endothelial
cells. Mechanical stress is of great importance considering the
environment of IVD, which is implicated as the predominant
inductive cause of IDD; they are interconnected and amplify each
other, and cellular physiology is strongly affected by mechanical
loading (Vergroesen et al., 2015). In the early stage of spine
development, NC cells are squeezed into IVD through the thrust
of forming vertebrae, and then they enter the specic
physiological environment and are compressed by mechanical
force. Compressive load cultures have been found to induce the
CK8 phosphorylation and downregulation in NP cells (Sun et al.,
2013a), and NC cells are more resistant to mechanical forces
compared to NP cells (Saggese et al., 2020), while high mechanical
stress may lead to the exhaustion of NC resources (Hong et al.,
2018). These ndings hypothesize that the appropriate range of
mechanical force is necessary to maintain the normal survival and
function of NC cells, as well as the secretion of exosomes; 0.5 Mpa
is found to be the suitable mechanical condition for inducing
secretion of NC exosomes and modulating related functions.
NC exosomes have also been unveiled to deliver miR-140-5p
to endothelial cells and then inhibit angiogenesis to maintain the
avascular status of IVD, which are achieved by the Wnt/β-catenin
signaling pathway (Sun et al., 2020b). Among them, miR-140-5p
has been proven to participate in cell migration, proliferation, and
metastasis (Rothman et al., 2016;Fang et al., 2017), and the
exosomal miR-140-5p from NC could suppress the expression of
MMP-2 and MMP-7 and thus ameliorate NP cell apoptosis and
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214911
Li et al. Different Exosomes for IDD Therapy
IDD. Wnt11, one of Wnt family members, has been shown to
stimulate the proliferation, migration, and invasion of various
types of cells via inducing β-catenin (Stefater et al., 2011;Franco
et al., 2016), which is regarded as the pivotal downstream
transduction of Wnt; these are consistent with the interactions
via NC exosomes, that Wnt transduction is involved in
angiogenesis through the modulation of endothelial cell
proliferation and vascular sprouting (Zhang et al., 2017).
Consequently, NC may have anti-angiogenesis ability via the
NC-exosomal miR-140-5p/Wnt/β-catenin axis. According to
their mass spectrometry study, Matta et al. (2017) have
discovered from a non-chondrodystrophic dog notochordal
cell conditioned medium that transforming growth factor
beta1 (TGF-β1) and connective tissue growth (CTGF) are
major hubs in protein interaction networks, which are
essential for the homeostasis regulation of healthy NP tissues.
Interestingly, VEGFA is also predicted to be inhibited among
growth factors in NC cells (Rodrigues-Pinto et al., 2018). The
identication of key biological factors derived from NC cells that
delay the progression of IDD is still at an early stage; based on
these ndings, more studies about the functions mediated by NC
exosomes need further exploration.
THE RELATIONSHIPS BETWEEN IVD
VASCULARIZATION, INFLAMMATION AND
IVD CELLS, AND EXOSOMES
As the largest avascular organ, IVDs are composed of three parts,
namely, central NP cells, surrounding AF cells, and the adjacent
cartilage endplates. Generally, blood vessels are conned to the
outer surface of AF cells in normal IVD, while neovascularization
has been widely observed and considered as a common
pathological phenomenon in IDD. The inward growth of
blood vessels causes NP exposure to immune system, leading
to permeation of inammatory factors and immune cells into NP
tissues, and nally damage to the immune privilege and
homeostasis of IVD (Sun et al., 2021). Additionally, the
ingrowth of blood vessels in IVD enhances neuralization and
pain sensitization (Choi, 2009); it also affects the phenotype and
functions of NP cells by upregulating oxygen concentration in NP
tissue (Wang et al., 2019). Therefore, the maintenance of the
avascular condition of IVD is essential to sustain the homeostasis
and functions of normal IVD (Chu et al., 2018).
In terms of the mechanisms of vascularization in IDD, it is
generally considered to be the breakdown of physical barrier,
such as the ssure of AF tissue, together with the increased
expression of pro-angiogenesis factors, like VEGF and platelet-
derived growth factor (PDGF) (Tolonen et al., 1997;Fujita et al.,
2008), and pro-inammatory cytokines like IL-1βand TNF-α
(Lee et al., 2011;Risbud and Shapiro, 2014), that nally induce
invasion of blood vessels. Focal proteoglycan loss has been
noticed to cause alteration of ECM, which facilitates the
growth of nerves and blood vessels (Stefanakis et al., 2012);
increased blood vessel ingrowth is correlated with
proteoglycan depletion AF lesion (Moon et al., 2012) and IVD
aggrecan could inhibit migration and invasion of endothelial cells
(Johnson et al., 2005). Furthermore, mechanical loading has also
been proven to inuence the capacity of IVD to stimulate the
migration of endothelial cells (Neidlinger-Wilke et al., 2009).
These results demonstrate the pivotal roles of a passive physical
barrier in preventing IVD angiogenesis. As for molecular level,
Wiet et al. (2017) have described the interactions between mast
cells and IVD: that healthy AF culture medium could suppress the
activation of mast cells by downregulating expression of
inammatory cytokines and then inhibit mast cell-induced
angiogenesis. Cornejo et al. (2015) have provided evidence
that soluble factors derived from notochordal-rich IVD could
suppress angiogenesis via inhibiting VEGF signaling pathways,
and NC-derived ligands are of signicance in targeting
neurovascular ingrowth and pain in the degenerative IVD. A
previous study has also reported the importance of FasFasL (Fas
Ligand) interaction, showing that FasL could induce apoptosis of
endothelial cells (Sun et al., 2013b); besides, FasL generated by
IVD could mediate the apoptosis of Fas-bearing cancer cells (Park
et al., 2007), and the FasFasL network may provide a novel target
for the treatment strategies of IDD. These results all suggest that
these factors or cytokines might be the molecular monitor for
maintaining functions through inducing apoptosis of vascular
endothelial cells in addition to the traditional physical barrier.
In the aspect of cells and related exosomes, a previous study
has displayed that degenerative AF cells could promote
vascularization. Moon et al. (2014) have reported that AF cells
from degenerative IVD stimulate endothelial cells and produce
factors known to induce ECM degradation, angiogenesis, and
innervation (Moon et al., 2014); degenerative NP cells have been
proven to promote blood vessel growth by secreting pro-
inammatory factors (He et al., 2020). In the early stage of
disc development, notochordal cells existing in NP cells could
inhibit angiogenesis of IVD (Cornejo et al., 2015); additionally, a
previous study has also provided evidence that degenerative AF
exosomes could induce IVD vascularization and inammation
directly through upregulation of IL-6, TNF-α, and VEGF, and
indirectly through enhancing the invasion of blood vessels via
accumulation of MMPs (Sun et al., 2021). Furthermore, as for
exosomes derived from MSC cells, a report indicated that miR-
125a represses angiogenic inhibitor DLL4 in endothelial cells and
thereby promotes angiogenesis (Liang et al., 2016). In terms of
NP, although NP exosomes have been reported to exist and have
numerous biological effects, they are mainly focused on
stimulating MSC differentiation (Lan et al., 2019;Yuan et al.,
2020); detailed research about NP exosomes and blood vessels
remains limited. As a result, there is an urgent need to clarify the
effects of NP exosomes on vascular endothelial cells.
PMSCs possess several characteristics such as proliferation,
migration, cloning, and immune regulation, and have broad
applications in clinical practice (Mathew et al., 2020). The
numbers of cytokines and chemokines released from PMSC
are a key point to modulate angiogenesis, which facilitates the
possibility of considering PMSC exosomes as a target therapy to
prompt angiogenesis. Liang et al. (2017) have reported that the
co-culture system between PMSCs and EPCs enhances the
angiogenic potential of EPCs through PDGF and the Notch
signaling pathway, and conditioned media that may contain
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214912
Li et al. Different Exosomes for IDD Therapy
exosomes have signicant pro-angiogenesis effects on EPCs and
HUVECs (Komaki et al., 2017). PMSCs generate various kinds of
angiogenic factors like VEGF, bFGF, IL-6, IL-8, and HGF
(Komaki et al., 2017;Liang et al., 2017), and possible
mechanisms of angiogenesis are also reported to include
activation of PAKT and p38MAPK/pSTAT3 that induce
VEGF secretion and recruitment of smooth muscle cells and
pericytes (Chen et al., 2015;Makhoul et al., 2016). Several in vivo
studies have gained promising results, namely, that PMSCs could
enhance vessel density, blood ow, and perfusion in a dose- and
site-dependent manner, especially in ischemia mice models
(Francki et al., 2016;Xie et al., 2016;Zahavi-Goldstein et al.,
2017). Interestingly, a comparative study (König et al., 2015)
revealed that the angiogenic ability of MSCs derived from blood
vessels is stronger than that in avascular sources, indicating the
importance of determining the components of the conditioned
medium from different cell sources. The above comments have
fully discussed the mechanisms in terms of angiogenic potential,
while in the aspect of anti-angiogenic potential of PMSCs,
Alshareeda et al. (2018) have found that the co-culture of
PMSCs along with breast cancer cells could signicantly
inhibit the migration, invasion, and tube formation ability of
HUVECs. Zhang et al. (2014b) have shown that PMSCs could
inhibit peritoneal tumorigenesis via downregulating blood vessel
counts, and injection of PMSC into retinopathy mouse models
has been demonstrated to prevent neovascularization through
upregulation of TGF-β1(Kim et al., 2016); these results could
partly be explained by the upregulation of miRNAs like miR-136
under pathology situations that modulate the inhibition of
capillary formation (Ji et al., 2017). Related studies about
PMSC exosomes remain at an early stage; PMSC exosomes are
known to modulate osteogenic and adipogenic differentiation by
upregulating OCT4 and NANOG in dermal broblasts (Tooi
et al., 2016), and they could enhance the migration and tube
formation of endothelial cells (Komaki et al., 2017), while Yuan
et al. (2020) have found the applications of PMSC exosomes in
the treatment of IDD and that exosomes could upregulate
ZNF121 and thus ameliorate IDD by delivering miR-4450
inhibitor (AntagomiR-4450). Basically, the applications of
angiogenesis and anti-angiogenesis by PMSC exosomes are a
controversial issue, modulated in a very contextual manner. More
specic miRNAs from PMSC exosomes targeting IDD or
endothelial cell functions as well as their potential roles are to
be further identied.
PROSPECTS OF EXOSOME THERAPY
FOR IDD
Accumulating lines of evidence have already reported the
essential applications of different sources of exosomes in the
treatment of IDD, which have been fully described in this
review. Many studies have already demonstrated the role of
exosomal miRNAs in inhibiting apoptosis of NP cells and
suppressing MMP expression; however, the correlations
among different sources of exosomes and whether the high
expressed miRNAs in one kind of exosome are also detected in
other exosomes remain unknown. These issues all need to be
further addressed.
In addition to the inammatory environment, the central
portion of degenerative disc also has low cell density, low
glucose, low pH value, low oxygen, high osmotic pressure, and
high mechanical variations (Lu et al., 2017). Although MSC and
MSC-derived exosomes have shown an effective inuence on
degenerative IVD, and cell-injection strategy has suggested
promising results, there remain obstacles regarding MSC in
clinical practice, especially how transplanted cells are able to
survive and adapt in avascular IVD conditions and how to inhibit
angiogenesis rather than promote blood vessel ingrowth of IVD
when using exosomes resembling PMSC-derived (Krock et al.,
2015;Sakai and Andersson, 2015); these issues are worth
pondering and need to be resolved by scientic researchers.
Lastly, the most appropriate dose of exosome injection, along
with the most optimal route of administration, remains unclear and
requires further research (Loibl et al., 2019;Forsberg et al., 2020).
The current studies about the administration route mainly focus on
two points: direct intradiscal injection and systemic injection. Due to
the avascular characteristics of IVD, it is hypothesized that direct
injection of exosomes into the disc would be the most effective
approach (Noriega et al., 2017), while an in vivo study of MSC
exosomes administrated through tail vein by Zhang et al. (2020) also
displayed promising results in the treatment of IDD. In the aspect of
systemic injection, current studies only use single dose in vivo,andif
systemic injections are applied, multiple doses may be conducted to
maintain the therapeutic effects, and in this situation, it is essential to
determinesafetyandefciency, as well as the dose and frequency of
injections. In terms of direct intradiscal injection, the following
questions need to be addressed: Would the puncture needle for
injection cause extra damage to the vertebral body or IVD? What is
the denition of additional hurt to disc? Would the injection
approach be applied in the clinic? To the best of our knowledge,
there is only one clinical trial relating to the treatment of IDD by
exosomes in India, which would be conducted when all participants
are recruited. This clinical trial aims to observe the efcacy of PRP
exosomes by intradiscal injection [Intra-discal Injection of Platelet-
rich Plasma (PRP) Enriched With Exosomes in Chronic Low Back
Pain, https://clinicaltrials.gov/ct2/show/NCT04849429]. Once these
considerations are fully addressed, more clinical trials about IDD
therapy using an exosome approach are needed.
AUTHOR CONTRIBUTIONS
This study was completed with teamwork. Each author had made
corresponding contribution to the study. Conceived the idea: MY, BG,
and WL. Wrote the main manuscript: WL, SZ, and DW. Perpared
gures:WL,HZ,QS,YZ,MW,andZD.Redressedthemanuscript:SX,
MY, BG, and WL. Reviewed the manuscript: SX, MY, BG, and WL.
FUNDING
This study was supported by grants from the National Natural
Science Foundation of China (Nos. 82072475 and 82172475).
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214913
Li et al. Different Exosomes for IDD Therapy
REFERENCES
Adamopoulos, C., Mihailidou, C., Grivaki, C., Papavassiliou, K. A., Kiaris, H.,
Piperi, C., et al. (2016). Systemic Effects of AGEs in ER Stress Induction In Vivo.
Glycoconj J. 33 (4), 537544. doi:10.1007/s10719-016-9680-4
Adams, M. A., Freeman, B. J., Morrison, H. P., Nelson, I. W., and Dolan, P. (2000).
Mechanical Initiation of Intervertebral Disc Degeneration. Spine (Phila Pa
1976) 25 (13), 16251636. doi:10.1097/00007632-200007010-00005
Akers, J. C., Ramakrishnan, V., Kim, R., Skog, J., Nakano, I., Pingle, S., et al. (2013).
MiR-21 in the Extracellular Vesicles (EVs) of Cerebrospinal Fluid (CSF): a
Platform for Glioblastoma Biomarker Development. PLoS One 8 (10), e78115.
doi:10.1371/journal.pone.0078115
Akkoc, Y., and Gozuacik, D. (2020). MicroRNAs as Major Regulators of the
Autophagy Pathway. Biochim. Biophys. Acta (Bba) - Mol. Cel Res. 1867 (5),
118662. doi:10.1016/j.bbamcr.2020.118662
Al-Nedawi, K., Meehan, B., and Rak, J. (2009). Microvesicles: Messengers and
Mediators of Tumor Progression. Cel. Cycle 8 (13), 20142018. doi:10.4161/cc.
8.13.8988
Alshareeda, A. T., Rakha, E., Alghwainem, A., Alrfaei, B., Alsowayan, B., Albugami,
A., et al. (2018). The Effect of Human Placental Chorionic Villi Derived
Mesenchymal Stem Cell on Triple-Negative Breast Cancer Hallmarks. PLoS
One 13 (11), e0207593. doi:10.1371/journal.pone.0207593
Andersson, G. B. (1999). Epidemiological Features of Chronic Low-Back Pain.
Lancet 354 (9178), 581585. doi:10.1016/s0140-6736(99)01312-4
Antoniou, J., Steffen, T., Nelson, F., Winterbottom, N., Hollander, A. P., Poole, R.
A., et al. (1996). The Human Lumbar Intervertebral Disc: Evidence for Changes
in the Biosynthesis and Denaturation of the Extracellular Matrix with Growth,
Maturation, Ageing, and Degeneration. J. Clin. Invest. 98 (4), 9961003. doi:10.
1172/JCI118884
Bach, F. C., Tellegen, A. R., Beukers, M., Miranda-Bedate, A., Teunissen, M., de
Jong, W. A. M., et al. (2018). Biologic Canine and Human Intervertebral Disc
Repair by Notochordal Cell-Derived Matrix: from Bench towards Bedside.
Oncotarget 9 (41), 2650726526. doi:10.18632/oncotarget.25476
Bai, X.-D., Li, X.-C., Chen, J.-H., Guo, Z.-M., Hou, L.-S., Wang, D.-L., et al. (2017).
Coculture with Partial Digestion Notochordal Cell-Rich Nucleus Pulposus
Tissue Activates Degenerative Human Nucleus Pulposus Cells. Tissue Eng.
A23 (15-16), 837846. doi:10.1089/ten.TEA.2016.0428
Battié,M.C.,Videman,T.,Gibbons,L.E.,Fisher,L.D.,Manninen,H.,andGill,
K. (1995). 1995 Volvo Award in Clinical Sciences. Determinants of Lumbar
Disc Degeneration. A Study Relating Lifetime Exposures and Magnetic
Resonance Imaging Findings in Identical Twins. Spine (Phila Pa 1976) 20
(24), 26012612.
Bergknut, N., Smolders, L. A., Grinwis, G. C. M., Hagman, R., Lagerstedt, A.-S.,
Hazewinkel, H. A. W., et al. (2013). Intervertebral Disc Degeneration in the
Dog. Part 1: Anatomy and Physiology of the Intervertebral Disc and
Characteristics of Intervertebral Disc Degeneration. Vet. J. 195 (3), 282291.
doi:10.1016/j.tvjl.2012.10.024
Bharadwaj, S., Liu, G., Shi, Y., Markert, C., Andersson, K.-E., Atala, A., et al. (2011).
Characterization of Urine-Derived Stem Cells Obtained from Upper Urinary
Tract for Use in Cell-Based Urological Tissue Engineering. Tissue Eng. Part A
17 (15-16), 21232132. doi:10.1089/ten.TEA.2010.0637
Bibby, S. R. S., and Urban, J. P. G. (2004). Effect of Nutrient Deprivation on the
Viability of Intervertebral Disc Cells. Eur. Spine J. 13 (8), 695701. doi:10.1007/
s00586-003-0616-x
Bijkerk, C., Houwing-Duistermaat, J. J., Valkenburg, H. A., Meulenbelt, I., Hofman,
A., Breedveld, F. C., et al. (1999). Heritabilities of Radiologic Osteoarthritis in
Peripheral Joints and of Disc Degeneration of the Spine. Arthritis Rheum. 42
(8), 17291735. doi:10.1002/1529-0131(199908)42:8<1729:aid-anr23>3.0.co;
2-h
Binch, A. L. A., Fitzgerald, J. C., Growney, E. A., and Barry, F. (2021). Cell-based
Strategies for IVD Repair: Clinical Progress and Translational Obstacles. Nat.
Rev. Rheumatol. 17 (3), 158175. doi:10.1038/s41584-020-00568-w
Bodin, A., Bharadwaj, S., Wu, S., Gatenholm, P., Atala, A., and Zhang, Y. (2010).
Tissue-engineered Conduit Using Urine-Derived Stem Cells Seeded Bacterial
Cellulose Polymer in Urinary Reconstruction and Diversion. Biomaterials 31
(34), 88898901. doi:10.1016/j.biomaterials.2010.07.108
Boubriak, O. A., Watson, N., Sivan, S. S., Stubbens, N., and Urban, J. P. G. (2013).
Factors Regulating Viable Cell Density in the Intervertebral Disc: Blood Supply
in Relation to Disc Height. J. Anat. 222 (3), 341348. doi:10.1111/joa.12022
Bowles, R. D., and Setton, L. A. (2017). Biomaterials for Intervertebral Disc
Regeneration and Repair. Biomaterials 129, 5467. doi:10.1016/j.
biomaterials.2017.03.013
Brinckmann, P., and Grootenboer, H. (1991). Change of Disc Height, Radial Disc
Bulge, and Intradiscal Pressure from Discectomy. An In Vitro Investigation on
Human Lumbar Discs. Spine (Phila Pa 1976) 16 (6), 641646. doi:10.1097/
00007632-199106000-00008
Cabral, J., Ryan, A. E., Grifn, M. D., and Ritter, T. (2018). Extracellular Vesicles as
Modulators of Wound Healing. Adv. Drug Deliv. Rev. 129, 394406. doi:10.
1016/j.addr.2018.01.018
Caby, M.-P., Lankar, D., Vincendeau-Scherrer, C., Raposo, G., and Bonnerot, C.
(2005). Exosomal-like Vesicles Are Present in Human Blood Plasma. Int.
Immunol. 17 (7), 879887. doi:10.1093/intimm/dxh267
Camussi, G., Deregibus, M. C., Bruno, S., Grange, C., Fonsato, V., and Tetta, C.
(2011). Exosome/microvesicle-mediated Epigenetic Reprogramming of Cells.
Am. J. Cancer Res. 1 (1), 98110.
Capossela, S., Schläi, P., Bertolo, A., Janner, T., Stadler, B. M., Pötzel, T., et al.
(2014). Degenerated Human Intervertebral Discs Contain Autoantibodies
against Extracellular Matrix Proteins. Eur. Cel. Mater 27, 251263. doi:10.
22203/ecm.v027a18
Capossela, S., Bertolo, A., Gunasekera, K., Pötzel, T., Baur, M., and Stoyanov, J. V.
(2018). VEGF Vascularization Pathway in Human Intervertebral Disc Does Not
Change during the Disc Degeneration Process. BMC Res. Notes 11 (1), 333.
doi:10.1186/s13104-018-3441-3
Chan, C.-M., Huang, D.-Y., Huang, Y.-P., Hsu, S.-H., Kang, L.-Y., Shen, C.-M.,
et al. (2016). Methylglyoxal Induces Cell Death through Endoplasmic
Reticulum Stress-Associated ROS Production and Mitochondrial
Dysfunction. J. Cel. Mol. Med. 20 (9), 17491760. doi:10.1111/jcmm.12893
Chen, C.-Y., Liu, S.-H., Chen, C.-Y., Chen, P.-C., and Chen, C.-P. (2015). Human
Placenta-Derived Multipotent Mesenchymal Stromal Cells Involved in
Placental Angiogenesis via the PDGF-BB and STAT3 Pathways1. Biol.
Reprod. 93 (4), 103. doi:10.1095/biolreprod.115.131250
Chen, Y., Xue, K., Zhang, X., Zheng, Z., and Liu, K. (2018). Exosomes Derived from
Mature Chondrocytes Facilitate Subcutaneous Stable Ectopic Chondrogenesis
of Cartilage Progenitor Cells. Stem Cel. Res. Ther. 9 (1), 318. doi:10.1186/
s13287-018-1047-2
Chen, W., Wang, H., Zhu, Z., Feng, J., and Chen, L. (2020). Exosome-Shuttled
circSHOC2 from IPASs Regulates Neuronal Autophagy and Ameliorates
Ischemic Brain Injury via the miR-7670-3p/SIRT1 Axis. Mol. Ther. - Nucleic
Acids 22, 657672. doi:10.1016/j.omtn.2020.09.027
Cheng, X., Zhang, G., Zhang, L., Hu, Y., Zhang, K., Sun, X., et al. (2018).
Mesenchymal Stem Cells Deliver Exogenous miR-21viaexosomes to Inhibit
Nucleus Pulposus Cell Apoptosis and Reduce Intervertebral Disc Degeneration.
J. Cel. Mol. Med. 22 (1), 261276. doi:10.1111/jcmm.13316
Chiang, C.-K., Wang, C.-C., Lu, T.-F., Huang, K.-H., Sheu, M.-L., Liu, S.-H., et al.
(2016). Involvement of Endoplasmic Reticulum Stress, Autophagy and
Apoptosis in Advanced Glycation End Products-Induced Glomerular
Mesangial Cell Injury. Sci. Rep. 6, 34167. doi:10.1038/srep34167
Choi, Y.-S. (2009). Pathophysiology of Degenerative Disc Disease. Asian Spine J. 3
(1), 3944. doi:10.4184/asj.2009.3.1.39
Chu, G., Shi, C., Wang, H., Zhang, W., Yang, H., and Li, B. (2018). Strategies for
Annulus Fibrosus Regeneration: From Biological Therapies to Tissue
Engineering. Front. Bioeng. Biotechnol. 6, 90. doi:10.3389/fbioe.2018.00090
Cocucci, E., Racchetti, G., and Meldolesi, J. (2009). Shedding Microvesicles:
Artefacts No More. Trends Cel. Biol. 19 (2), 4351. doi:10.1016/j.tcb.2008.
11.003
Cornejo, M. C., Cho, S. K., Giannarelli, C., Iatridis, J. C., and Purmessur, D. (2015).
Soluble Factors from the Notochordal-Rich Intervertebral Disc Inhibit
Endothelial Cell Invasion and Vessel Formation in the Presence and
Absence of Pro-inammatory Cytokines. Osteoarthritis Cartilage 23 (3),
487496. doi:10.1016/j.joca.2014.12.010
Dai, H., Sinclair, D. A., Ellis, J. L., and Steegborn, C. (2018). Sirtuin Activators and
Inhibitors: Promises, Achievements, and Challenges. Pharmacol. Ther. 188,
140154. doi:10.1016/j.pharmthera.2018.03.004
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214914
Li et al. Different Exosomes for IDD Therapy
de Vries, S. A. H., Potier, E., van Doeselaar, M., Meij, B. P., Tryfonidou, M. A., and
Ito, K. (2015). Conditioned Medium Derived from Notochordal Cell-Rich
Nucleus Pulposus Tissue Stimulates Matrix Production by Canine Nucleus
Pulposus Cells and Bone Marrow-Derived Stromal Cells. Tissue Eng. Part A 21
(5-6), 10771084. doi:10.1089/ten.TEA.2014.0309
Di Martino, A., Merlini, L., and Faldini, C. (2013). Autoimmunity in Intervertebral
Disc Herniation: from Bench to Bedside. Expert Opin. Ther. Targets 17 (12),
14611470. doi:10.1517/14728222.2013.834330
Dieleman, J. L., Cao, J., Chapin, A., Chen, C., Li, Z., Liu, A., et al. (2020). US Health
Care Spending by Payer and Health Condition, 1996-2016. JAMA 323 (9),
863884. doi:10.1001/jama.2020.0734
Dixon, C. L., Sheller-Miller, S., Saade, G. R., Fortunato, S. J., Lai, A., Palma, C., et al.
(2018). Amniotic Fluid Exosome Proteomic Prole Exhibits Unique Pathways
of Term and Preterm Labor. Endocrinology 159 (5), 22292240. doi:10.1210/en.
2018-00073
Dou, Y., Sun, X., Ma, X., Zhao, X., and Yang, Q. (2021). Intervertebral Disk
Degeneration: The Microenvironment and Tissue Engineering Strategies.
Front. Bioeng. Biotechnol. 9, 592118. doi:10.3389/fbioe.2021.592118
Dragovic, R. A., Gardiner, C., Brooks, A. S., Tannetta, D. S., Ferguson, D. J. P., Hole,
P., et al. (2011). Sizing and Phenotyping of Cellular Vesicles Using Nanoparticle
Tracking Analysis. Nanomedicine: Nanotechnol. Biol. Med. 7 (6), 780788.
doi:10.1016/j.nano.2011.04.003
Du, L., Yang, Q., Zhang, J., Zhu, M., Ma, X., Zhang, Y., et al. (2019). Engineering a
Biomimetic Integrated Scaffold for Intervertebral Disc Replacement. Mater. Sci.
Eng. C 96, 522529. doi:10.1016/j.msec.2018.11.087
El Andaloussi, S., Lakhal, S., Mäger, I., and Wood, M. J. A. (2013). Exosomes for
Targeted siRNA Delivery across Biological Barriers. Adv. Drug Deliv. Rev. 65
(3), 391397. doi:10.1016/j.addr.2012.08.008
Fang, Z., Yin, S., Sun, R., Zhang, S., Fu, M., Wu, Y., et al. (2017). miR-140-5p
Suppresses the Proliferation, Migration and Invasion of Gastric Cancer by
Regulating YES1. Mol. Cancer 16 (1), 139. doi:10.1186/s12943-017-0708-6
Feng, F.-B., and Qiu, H.-Y. (2018). Effects of Artesunate on Chondrocyte
Proliferation, Apoptosis and Autophagy through the PI3K/AKT/mTOR
Signaling Pathway in Rat Models with Rheumatoid Arthritis. Biomed.
Pharmacother. 102, 12091220. doi:10.1016/j.biopha.2018.03.142
Fields, A. J., Berg-Johansen, B., Metz, L. N., Miller, S., La, B., Liebenberg, E. C., et al.
(2015). Alterations in Intervertebral Disc Composition, Matrix Homeostasis
and Biomechanical Behavior in the UCD-T2dm Rat Model of Type 2 Diabetes.
J. Orthop. Res. 33 (5), 738746. doi:10.1002/jor.22807
Forsberg, M. H., Kink, J. A., Hematti, P., and Capitini, C. M. (2020). Mesenchymal
Stromal Cells and Exosomes: Progress and Challenges. Front. Cel Dev. Biol. 8,
665. doi:10.3389/fcell.2020.00665
Francki, A., Labazzo, K., He, S., Baum, E. Z., Abbot, S. E., Herzberg, U., et al. (2016).
Angiogenic Properties of Human Placenta-Derived Adherent Cells and Efcacy
in Hindlimb Ischemia. J. Vasc. Surg. 64 (3), 746756. doi:10.1016/j.jvs.2015.
04.387
Franco, C. A., Jones, M. L., Bernabeu, M. O., Vion, A.-C., Barbacena, P., Fan, J.,
et al. (2016). Non-canonical Wnt Signalling Modulates the Endothelial Shear
Stress Flow Sensor in Vascular Remodelling. Elife 5, e07727. doi:10.7554/eLife.
07727
Freemont, T. J., LeMaitre, C., Watkins, A., and Hoyland, J. A. (2001). Degeneration
of Intervertebral Discs: Current Understanding of Cellular and Molecular
Events, and Implications for Novel Therapies. Expert Rev. Mol. Med. 2001,
110. doi:10.1017/S1462399401002885
Fujita, N., Imai, J.-i., Suzuki, T., Yamada, M., Ninomiya, K., Miyamoto, K., et al.
(2008). Vascular Endothelial Growth Factor-A Is a Survival Factor for Nucleus
Pulposus Cells in the Intervertebral Disc. Biochem. Biophys. Res. Commun. 372
(2), 367372. doi:10.1016/j.bbrc.2008.05.044
Gawri, R., Rosenzweig, D. H., Krock, E., Ouellet, J. A., Stone, L. S., Quinn, T. M.,
et al. (2014). High Mechanical Strain of Primary Intervertebral Disc Cells
Promotes Secretion of Inammatory Factors Associated with Disc
Degeneration and Pain. Arthritis Res. Ther. 16 (1), R21. doi:10.1186/ar4449
Geiss, A., Larsson, K., Rydevik, B., Takahashi, I., and Olmarker, K. (2007).
Autoimmune Properties of Nucleus Pulposus: an Experimental Study in
Pigs. Spine (Phila Pa 1976) 32 (2), 168173. doi:10.1097/01.brs.0000251651.
61844.2d
Geiss, A., Sobottke, R., Delank, K. S., and Eysel, P. (2014). Macrophages Do Not
Represent the Main Cell Type in Sequestrated and Extruded Intervertebral
Discs: Evidence for Their Involvement in Disc Resorption, rather Than
Initiation of an Immune Response. Glob. Spine J. 04 (1_Suppl. l), s-0034-
1376618. doi:10.1055/s-0034-1376618
Go, B. S., Kim, J., Yang, J. H., and Choe, E. S. (2017). Psychostimulant-Induced
Endoplasmic Reticulum Stress and Neurodegeneration. Mol. Neurobiol. 54 (6),
40414048. doi:10.1007/s12035-016-9969-0
Goc, A., Choudhary, M., Byzova, T. V., and Somanath, P. R. (2011). TGFβ- and
Bleomycin-Induced Extracellular Matrix Synthesis Is Mediated through Akt
and Mammalian Target of Rapamycin (mTOR). J. Cel. Physiol. 226 (11),
30043013. doi:10.1002/jcp.22648
Gorodetsky, R., and Aicher, W. K. (2021). Allogenic Use of Human Placenta-
Derived Stromal Cells as a Highly Active Subtype of Mesenchymal Stromal
Cells for Cell-Based Therapies. Ijms 22 (10), 5302. doi:10.3390/ijms22105302
Goto, T., Fujiya, M., Konishi, H., Sasajima, J., Fujibayashi, S., Hayashi, A., et al.
(2018). An Elevated Expression of Serum Exosomal microRNA-191, 21,
451a of Pancreatic Neoplasm Is Considered to Be Efcient Diagnostic Marker.
BMC Cancer 18 (1), 116. doi:10.1186/s12885-018-4006-5
Gruber, H. E., Ingram, J. A., and Hanley, E. N. (2006). Immunolocalization of
Thrombospondin in the Human and Sand Rat Intervertebral Disc. Spine (Phila
Pa 1976) 31 (22), 25562561. doi:10.1097/01.brs.0000241117.31510.e3
Gruenberg, J., and van der Goot, F. G. (2006). Mechanisms of Pathogen Entry
through the Endosomal Compartments. Nat. Rev. Mol. Cel. Biol. 7 (7), 495504.
doi:10.1038/nrm1959
Guo, J., Shao, M., Lu, F., Jiang, J., and Xia, X. (2017). Role of Sirt1 Plays in Nucleus
Pulposus Cells and Intervertebral Disc Degeneration. Spine (Phila Pa 1976) 42
(13), E757E766. doi:10.1097/BRS.0000000000001954
Guo, Z., Su, W., Zhou, R., Zhang, G., Yang, S., Wu, X., et al. (2021). Exosomal
MATN3 of Urine-Derived Stem Cells Ameliorates Intervertebral Disc
Degeneration by Antisenescence Effects and Promotes NPC Proliferation
and ECM Synthesis by Activating TGF-β.Oxid. Med. Cel. Longev. 2021,
118. doi:10.1155/2021/5542241
Hamidi, A., Song, J., Thakur, N., Itoh, S., Marcusson, A., Bergh, A., et al. (2017).
TGF-βPromotes PI3K-AKT Signaling and Prostate Cancer Cell Migration
through the TRAF6-Mediated Ubiquitylation of P85α.Sci. Signal. 10, eaal4186.
doi:10.1126/scisignal.aal4186
Hampton, D., Laros, G., McCarron, R., and Franks, D. (1989). Healing Potential of
the Anulus Fibrosus. Spine (1976) 14 (4), 398401. Epub 1989/04/01. doi:10.
1097/00007632-198904000-00009
Hashimoto, K., Aizawa, T., Kanno, H., and Itoi, E. (2019). Adjacent Segment
Degeneration after Fusion Spinal Surgery-A Systematic Review. Int.
Orthopaedics (Sicot) 43 (4), 987993. doi:10.1007/s00264-018-4241-z
He, J., Zhang, A., Song, Z., Guo, S., Chen, Y., Liu, Z., et al. (2019). The Resistant
Effect of SIRT1 in Oxidative Stress-Induced Senescence of Rat Nucleus
Pulposus Cell Is Regulated by Akt-FoxO1 Pathway. Biosci. Rep. 39 (5),
BSR20190112. doi:10.1042/BSR20190112
He, M., Pang, J., Sun, H., Zheng, G., Lin, Y., and Ge, W. (2020). P14ARF Inhibits
Regional Inammation and Vascularization in Intervertebral Disc
Degeneration by Upregulating TIMP3. Am. J. Physiology-Cell Physiol. 318
(4), C751C761. doi:10.1152/ajpcell.00271.2019
Heijnen, H. F. G., Schiel, A. E., Fijnheer, R., Geuze, H. J., and Sixma, J. J. (1999).
Activated Platelets Release Two Types of Membrane Vesicles: Microvesicles by
Surface Shedding and Exosomes Derived from Exocytosis of Multivesicular
Bodies and alpha-Granules. Blood 94 (11), 37913799. doi:10.1182/blood.v94.
11.3791.423a22_3791_3799
Hejazi, F., Ebrahimi, V., Asgary, M., Piryaei, A., Fridoni, M. J., Kermani, A. A., et al.
(2021). Improved Healing of Critical-Size Femoral Defect in Osteoporosis Rat
Models Using 3D elastin/polycaprolactone/nHA Scaffold in Combination with
Mesenchymal Stem Cells. J. Mater. Sci. Mater. Med. 32 (3), 27. doi:10.1007/
s10856-021-06495-w
Hermantin, F. U., Peters, T., Quartararo, L., and Kambin, P. (1999). A Prospective,
Randomized Study Comparing the Results of Open Discectomy with Those of
Video-Assisted Arthroscopic Microdiscectomy*.J. Bone Jt. Surg. 81 (7),
958965. doi:10.2106/00004623-199907000-00008
Hetz, C. (2012). The Unfolded Protein Response: Controlling Cell Fate Decisions
under ER Stress and beyond. Nat. Rev. Mol. Cel. Biol. 13 (2), 89102. doi:10.
1038/nrm3270
Hong, X., Zhang, C., Wang, F., and Wu, X.-T. (2018). Large Cytoplasmic Vacuoles
within Notochordal Nucleus Pulposus Cells: A Possible Regulator of
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214915
Li et al. Different Exosomes for IDD Therapy
Intracellular Pressure that Shapes the Cytoskeleton and Controls Proliferation.
Cel. Tissues Organs 206 (1-2), 915. doi:10.1159/000493258
Höög, J. L., and Lötvall, J. (2015). Diversity of Extracellular Vesicles in Human
Ejaculates Revealed by Cryo-Electron Microscopy. J. Extracell. Vesicles 4, 28680.
doi:10.3402/jev.v4.28680
Horner, H. A., and Urban, J. P. G. (2001). 2001 Volvo Award Winner in Basic
Science Studies: Effect of Nutrient Supply on the Viability of Cells from the
Nucleus Pulposus of the Intervertebral Disc. Spine 26 (23), 25432549. doi:10.
1097/00007632-200112010-00006
Hu, Z.-L., Li, H.-Y., Chang, X., Li, Y.-Y., Liu, C.-H., Gao, X.-X., et al. (2020).
Exosomes Derived from Stem Cells as an Emerging Therapeutic Strategy for
Intervertebral Disc Degeneration. Wjsc 12 (8), 803813. doi:10.4252/wjsc.v12.
i8.803
Hu, S.-Q., Zhang, Q.-C., Meng, Q.-B., Hu, A.-N., Zou, J.-P., and Li, X.-L. (2020).
Autophagy Regulates Exosome Secretion in Rat Nucleus Pulposus Cells via the
RhoC/ROCK2 Pathway. Exp. Cel Res. 395 (2), 112239. doi:10.1016/j.yexcr.2020.
112239
Huan, C., Xiaoxu, C., and Xifang, R. (2019). Zinc Finger Protein 521, Negatively
Regulated by MicroRNA-204-5p, Promotes Proliferation, Motility and
Invasion of Gastric Cancer Cells. Technol. Cancer Res. Treat. 18,
153303381987478. doi:10.1177/1533033819874783
Hunt, M. A., Charlton, J. M., and Esculier, J.-F. (2020). Osteoarthritis Year in
Review 2019: Mechanics. Osteoarthritis Cartilage 28 (3), 267274. doi:10.1016/
j.joca.2019.12.003
Hunter, M. P., Ismail, N., Zhang, X., Aguda, B. D., Lee, E. J., Yu, L., et al. (2008).
Detection of microRNA Expression in Human Peripheral Blood Microvesicles.
PLoS One 3 (11), e3694. doi:10.1371/journal.pone.0003694
In t Anker, P. S., Scherjon, S. A., Kleijburg-van der Keur, C., de Groot-Swings, G.
M. J. S., Claas, F. H. J., Fibbe, W. E., et al. (2004). Isolation of Mesenchymal Stem
Cells of Fetal or Maternal Origin from Human Placenta. Stem Cel. 22,
13381345. doi:10.1634/stemcells.2004-0058
Ji,L.,Zhang,L.,Li,Y.,Guo,L.,Cao,N.,Bai,Z.,etal.(2017).MiR-136ContributestoPre-
eclampsia through its Effects on Apoptosis and Angiogenesis of Mesenchymal Stem
Cells. Placenta 50, 102109. doi:10.1016/j.placenta.2017.01.102
Ji, M.-l., Jiang, H., Zhang, X.-j., Shi, P.-l., Li, C., Wu, H., et al. (2018). Preclinical
Development of a microRNA-Based Therapy for Intervertebral Disc
Degeneration. Nat. Commun. 9 (1), 5051. doi:10.1038/s41467-018-07360-1
Jiang, L., Zhang, X., Zheng, X., Ru, A., Ni, X., Wu, Y., et al. (2013). Apoptosis,
Senescence, and Autophagy in Rat Nucleus Pulposus Cells: Implications for
Diabetic Intervertebral Disc Degeneration. J. Orthop. Res. 31 (5), 692702.
doi:10.1002/jor.22289
Jiang, Y., Zhang, C., Long, L., Ge, L., Guo, J., Fan, Z., et al. (2021). A Comprehensive
Analysis of SE-lncRNA/mRNA Differential Expression Proles during
Chondrogenic Differentiation of Human Bone Marrow Mesenchymal Stem
Cells. Front. Cel. Dev. Biol. 9, 721205. doi:10.3389/fcell.2021.721205
Jin, L., Shimmer, A. L., and Li, X. (2013). The challenge and Advancement of
Annulus Fibrosus Tissue Engineering. Eur. Spine J. 22 (5), 10901100. doi:10.
1007/s00586-013-2663-2
Jing, W., and Jiang, W. (2015). MicroRNA-93 Regulates Collagen Loss by
Targeting MMP3 in Human Nucleus Pulposus Cells. Cell Prolif. 48 (3),
284292. doi:10.1111/cpr.12176
Johnson, W. E., Caterson, B., Eisenstein, S. M., and Roberts, S. (2005). Human
Intervertebral Disc Aggrecan Inhibits Endothelial Cell Adhesion and Cell
Migration In Vitro.Spine (Phila Pa 1976) 30 (10), 11391147. doi:10.1097/
01.brs.0000162624.95262.73
Johnstone, R., Bianchini, A., and Teng, K. (1989). Reticulocyte Maturation and
Exosome Release: Transferrin Receptor Containing Exosomes Shows Multiple
Plasma Membrane Functions. Blood 74 (5), 18441851. doi:10.1182/blood.v74.
5.1844.1844
Jung, M. K., and Mun, J. Y. (2018). Sample Preparation and Imaging of Exosomes
by Transmission Electron Microscopy. JoVE 131, 56482. doi:10.3791/56482
Kadow, T., Sowa, G., Vo, N., and Kang, J. D. (2015). Molecular Basis of
Intervertebral Disc Degeneration and Herniations: what Are the Important
Translational Questions? Clin. Orthop. Relat. Res. 473 (6), 19031912. doi:10.
1007/s11999-014-3774-8
Kepler, C. K., Ponnappan, R. K., Tannoury, C. A., Risbud, M. V., and Anderson, D.
G. (2013). The Molecular Basis of Intervertebral Disc Degeneration. Spine J. 13
(3), 318330. doi:10.1016/j.spinee.2012.12.003
Kim, J., and Guan, K.-L. (2019). mTOR as a central Hub of Nutrient Signalling and
Cell Growth. Nat. Cel. Biol. 21 (1), 6371. doi:10.1038/s41556-018-0205-1
Kim, N. H., Kang, E. S., Han, C. D., Kim, J. D., and Kim, C. H. (1981). Auto-
immune Response in Degenerated Lumbar Disk. Yonsei Med. J. 22 (1), 2632.
doi:10.3349/ymj.1981.22.1.26
Kim, I., Xu, W., and Reed, J. C. (2008). Cell Death and Endoplasmic Reticulum
Stress: Disease Relevance and Therapeutic Opportunities. Nat. Rev. Drug
Discov. 7 (12), 10131030. doi:10.1038/nrd2755
Kim, K.-S., Park, J.-M., Kong, T., Kim, C., Bae, S.-H., Kim, H. W., et al. (2016).
Retinal Angiogenesis Effects of TGF-β1 and Paracrine Factors Secreted from
Human Placental Stem Cells in Response to a Pathological Environment. Cel.
Transpl. 25 (6), 11451157. doi:10.3727/096368915X688263
Komaki, M., Numata, Y., Morioka, C., Honda, I., Tooi, M., Yokoyama, N., et al.
(2017). Exosomes of Human Placenta-Derived Mesenchymal Stem Cells
Stimulate Angiogenesis. Stem Cel. Res. Ther. 8 (1), 219. doi:10.1186/s13287-
017-0660-9
König, J., Weiss, G., Rossi, D., Wankhammer, K., Reinisch, A., Kinzer, M., et al.
(2015). Placental Mesenchymal Stromal Cells Derived from Blood Vessels or
Avascular Tissues: what Is the Better Choice to Support Endothelial Cell
Function? Stem Cell Dev. 24 (1), 115131. doi:10.1089/scd.2014.0115
Kowal, E. J. K., Ter-Ovanesyan, D., Regev, A., and Church, G. M. (2017).
Extracellular Vesicle Isolation and Analysis by Western Blotting. Methods
Mol. Biol. 1660, 143152. doi:10.1007/978-1-4939-7253-1_12
Krock, E., Rosenzweig, D., and Haglund, L. (2015). The Inammatory Milieu of the
Degenerate Disc: Is Mesenchymal Stem Cell-Based Therapy for Intervertebral
Disc Repair a Feasible Approach? Cscr 10 (4), 317328. doi:10.2174/
1574888x10666150211161956
Krut, Z., Pelled, G., Gazit, D., and Gazit, Z. (2021). Stem Cells and Exosomes: New
Therapies for Intervertebral Disc Degeneration. Cells 10 (9), 2241. doi:10.3390/
cells10092241
Kwon, W.-K., Moon, H. J., Kwon, T.-H., Park, Y.-K., and Kim, J. H. (2017).
Inuence of Rabbit Notochordal Cells on Symptomatic Intervertebral Disc
Degeneration: Anti-angiogenic Capacity on Human Endothelial Cell
Proliferation under Hypoxia. Osteoarthritis Cartilage 25 (10), 17381746.
doi:10.1016/j.joca.2017.06.003
Ladomery, M., and Dellaire, G. (2002). Multifunctional Zinc nger Proteins in
Development and Disease. Ann. Hum. Genet. 66 (Pt 5-6), 331342. doi:10.1017/
S0003480002001215
Lai, H.-C., Chang, Q.-Y., and Hsieh, C.-L. (2019). Signal Transduction Pathways of
Acupuncture for Treating Some Nervous System Diseases. Evid.-Based
Complement. Altern. Med. 2019, 137. doi:10.1155/2019/2909632
Lam, S., Chan, S. C. W., Chan, S., Leung, V., Lu, W., Cheung, K., et al. (2011). The
Role of Cryopreservation in the Biomechanical Properties of the Intervertebral
Disc. eCM 22, 393402. doi:10.22203/ecm.v022a29
Lan, P.-H., Liu, Z.-H., Pei, Y.-J., Wu, Z.-G., Yu, Y., Yang, Y.-F., et al. (2016).
Landscape of RNAs in Human Lumbar Disc Degeneration. Oncotarget 7 (39),
6316663176. doi:10.18632/oncotarget.11334
Lan, T., Ma, W., Hong, Z., Wu, L., Chen, X., and Yuan, Y. (2017). Long Non-coding
RNA Small Nucleolar RNA Host Gene 12 (SNHG12) Promotes Tumorigenesis
and Metastasis by Targeting miR-199a/b-5p in Hepatocellular Carcinoma.
J. Exp. Clin. Cancer Res. 36 (1), 11. doi:10.1186/s13046-016-0486-9
Lan, W.-r., Pan, S., Li, H.-y., Sun, C., Chang, X., Lu, K., et al. (2019). Inhibition of
the Notch1 Pathway Promotes the Effects of Nucleus Pulposus Cell-Derived
Exosomes on the Differentiation of Mesenchymal Stem Cells into Nucleus
Pulposus-like Cells in Rats. Stem Cel. Int. 2019, 112. doi:10.1155/2019/
8404168
Lan, T., Shiyu-Hu, H., Shen, Z., Yan, B., and Chen, J. (2021). New Insights into the
Interplay between miRNAs and Autophagy in the Aging of Intervertebral Discs.
Ageing Res. Rev. 65, 101227. doi:10.1016/j.arr.2020.101227
Le Maitre, C. L., Pockert, A., Buttle, D. J., Freemont, A. J., and Hoyland, J. A. (2007).
Matrix Synthesis and Degradation in Human Intervertebral Disc Degeneration.
Biochem. Soc. Trans. 35 (Pt 4), 652655. doi:10.1042/BST0350652
Lee, J. M., Song, J. Y., Baek, M., Jung, H.-Y., Kang, H., Han, I. B., et al. (2011).
Interleukin-1βInduces Angiogenesis and Innervation in Human Intervertebral
Disc Degeneration. J. Orthop. Res. 29 (2), 265269. doi:10.1002/jor.21210
Li, Z., Yu, X., Shen, J., Chan, M. T. V., and Wu, W. K. K. (2015). MicroRNA in
Intervertebral Disc Degeneration. Cel Prolif. 48 (3), 278283. doi:10.1111/cpr.
12180
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214916
Li et al. Different Exosomes for IDD Therapy
Li, Z., Wang, Y., Xiao, K., Xiang, S., Li, Z., and Weng, X. (2018). Emerging Role of
Exosomes in the Joint Diseases. Cel. Physiol. Biochem. 47 (5), 20082017.
doi:10.1159/000491469
Li, X., Yang, L., and Chen, L.-L. (2018). The Biogenesis, Functions, and Challenges
of Circular RNAs. Mol. Cel. 71 (3), 428442. doi:10.1016/j.molcel.2018.06.034
Li, Z., Chen, S., Chen, S., Huang, D., Ma, K., and Shao, Z. (2019). Moderate
Activation of Wnt/β-catenin Signaling Promotes the Survival of Rat Nucleus
Pulposus Cells via Regulating Apoptosis, Autophagy, and Senescence. J. Cel.
Biochem. 120 (8), 1251912533. doi:10.1002/jcb.28518
Li, Z.-q., Kong, L., Liu, C., and Xu, H.-G. (2020). Human Bone Marrow
Mesenchymal Stem Cell-Derived Exosomes Attenuate IL-1β-induced
Annulus Fibrosus Cell Damage. Am. J. Med. Sci. 360 (6), 693700. doi:10.
1016/j.amjms.2020.07.025
Liang, X., Zhang, L., Wang, S., Han, Q., and Zhao, R. C. (2016). Exosomes
Secreted by Mesenchymal Stem Cells Promote Endothelial Cell Angiogenesis
by Transferring miR-125a. J. Cel. Sci. 129 (11), 21822189. doi:10.1242/jcs.
170373
Liang, T., Zhu, L., Gao, W., Gong, M., Ren, J., Yao, H., et al. (2017). Coculture of
Endothelial Progenitor Cells and Mesenchymal Stem Cells Enhanced Their
Proliferation and Angiogenesis through PDGF and Notch Signaling. FEBS
Open Bio. 7 (11), 17221736. doi:10.1002/2211-5463.12317
Liao, Z., Luo, R., Li, G., Song, Y., Zhan, S., Zhao, K., et al. (2019). Exosomes from
Mesenchymal Stem Cells Modulate Endoplasmic Reticulum Stress to Protect
against Nucleus Pulposus Cell Death and Ameliorate Intervertebral Disc
Degeneration In Vivo.Theranostics 9 (14), 40844100. doi:10.7150/thno.33638
Liu, H., Sun, X., Gong, X., and Wang, G. (2019). Human Umbilical Cord
Mesenchymal Stem Cells Derived Exosomes Exert Antiapoptosis Effect via
Activating PI3K/Akt/mTOR Pathway on H9C2 Cells. J. Cel. Biochem. 120 (9),
1445514464. doi:10.1002/jcb.28705
Liu, H., Zhang, M., Shi, M., Zhang, T., Lu, W., Yang, S., et al. (2021). Adipose-
derived Mesenchymal Stromal Cell-Derived Exosomes Promote Tendon
Healing by Activating Both SMAD1/5/9 and SMAD2/3. Stem Cel. Res. Ther
12 (1), 338. doi:10.1186/s13287-021-02410-w
Liu, M., Yang, Y., Zhao, B., Yang, Y., Wang, J., Shen, K., et al. (2021). Exosomes
Derived from Adipose-Derived Mesenchymal Stem Cells Ameliorate
Radiation-Induced Brain Injury by Activating the SIRT1 Pathway. Front.
Cel Dev. Biol. 9, 693782. doi:10.3389/fcell.2021.693782
Livshits, M. A., Khomyakova, E., Evtushenko, E. G., Lazarev, V. N., Kulemin, N. A.,
Semina, S. E., et al. (2015). Isolation of Exosomes by Differential Centrifugation:
Theoretical Analysis of a Commonly Used Protocol. Sci. Rep. 5, 17319. doi:10.
1038/srep17319
Loibl, M., Wuertz-Kozak, K., Vadala, G., Lang, S., Fairbank, J., and Urban, J. P.
(2019). Controversies in Regenerative Medicine: Should Intervertebral Disc
Degeneration Be Treated with Mesenchymal Stem Cells? JOR Spine 2 (1), e1043.
doi:10.1002/jsp2.1043
Longo, U. G., Petrillo, S., Franceschetti, E., Maffulli, N., and Denaro, V. (2012).
Growth Factors and Anticatabolic Substances for Prevention and Management
of Intervertebral Disc Degeneration. Stem Cell Int. 2012, 19. doi:10.1155/2012/
897183
Lu, K., Li, H.-y., Yang, K., Wu, J.-l., Cai, X.-w., Zhou, Y., et al. (2017). Exosomes as
Potential Alternatives to Stem Cell Therapy for Intervertebral Disc
Degeneration: In-Vitro Study on Exosomes in Interaction of Nucleus
Pulposus Cells and Bone Marrow Mesenchymal Stem Cells. Stem Cel. Res.
Ther. 8 (1), 108. doi:10.1186/s13287-017-0563-9
Luo, A., Zhang, X., Fu, L., Zhu, Z., and Dong, J.-T. (2016). Zinc nger Factor
ZNF121 Is a MYC-Interacting Protein Functionally Affecting MYC and Cell
Proliferation in Epithelial Cells. J. Genet. Genomics 43 (12), 677685. doi:10.
1016/j.jgg.2016.05.006
Ma, C.-J., Liu, X., Che, L., Liu, Z.-H., Samartzis, D., and Wang, H.-Q. (2015). Stem
Cell Therapies for Intervertebral Disc Degeneration: Immune Privilege
Reinforcement by Fas/FasL Regulating Machinery. Cscr 10 (4), 285295.
doi:10.2174/1574888x10666150416114027
Ma, L., Liu, Y., Zhao, X., Li, P., and Jin, Q. (2019). Rapamycin Attenuates Articular
Cartilage Degeneration by Inhibiting β-catenin in a Murine Model of
Osteoarthritis. Connect. Tissue Res. 60 (5), 452462. doi:10.1080/03008207.
2019.1583223
Makhoul, G., Jurakhan, R., Jaiswal, P. K., Ridwan, K., Li, L., Selvasandran, K., et al.
(2016). Conditioned Medium of H9c2 Triggers VEGF Dependent Angiogenesis
by Activation of p38/pSTAT3 Pathways in Placenta Derived Stem Cells for
Cardiac Repair. Life Sci. 153, 213221. doi:10.1016/j.lfs.2016.04.009
Maldonado-Lasunción, I., Haggerty, A. E., Okuda, A., Mihara, T., de la Oliva, N.,
Verhaagen, J., et al. (2021). The Effect of Inammatory Priming on the
Therapeutic Potential of Mesenchymal Stromal Cells for Spinal Cord
Repair. Cells 10 (6), 1316. doi:10.3390/cells10061316
Mannion, A. F., Adams, M. A., and Dolan, P. (2000). Sudden and Unexpected
Loading Generates High Forces on the Lumbar Spine. Spine (Phila Pa 1976) 25
(7), 842852. doi:10.1097/00007632-200004010-00013
Mathew, S. A., Naik, C., Cahill, P. A., and Bhonde, R. R. (2020). Placental
Mesenchymal Stromal Cells as an Alternative Tool for Therapeutic
Angiogenesis. Cel. Mol. Life Sci. 77 (2), 253265. doi:10.1007/s00018-019-
03268-1
Mathivanan, S., Ji, H., and Simpson, R. J. (2010). Exosomes: Extracellular
Organelles Important in Intercellular Communication. J. Proteomics 73 (10),
19071920. doi:10.1016/j.jprot.2010.06.006
Matta, A., Karim, M. Z., Isenman, D. E., and Erwin, W. M. (2017). Molecular
Therapy for Degenerative Disc Disease: Clues from Secretome Analysis of the
Notochordal Cell-Rich Nucleus Pulposus. Sci. Rep. 7, 45623. doi:10.1038/
srep45623
Mehrkens, A., Matta, A., Karim, M. Z., Kim, S., Fehlings, M. G., Schaeren, S., et al.
(2017). Notochordal Cell-Derived Conditioned Medium Protects Human
Nucleus Pulposus Cells from Stress-Induced Apoptosis. Spine J. 17 (4),
579588. doi:10.1016/j.spinee.2017.01.003
Milasan, A., Tessandier, N., Tan, S., Brisson, A., Boilard, E., and Martel, C. (2016).
Extracellular Vesicles Are Present in Mouse Lymph and Their Level Differs in
Atherosclerosis. J. Extracell. Vesicles 5, 31427. doi:10.3402/jev.v5.31427
Millecamps, M., Czerminski, J. T., Mathieu, A. P., and Stone, L. S. (2015).
Behavioral Signs of Axial Low Back Pain and Motor Impairment Correlate
with the Severity of Intervertebral Disc Degeneration in a Mouse Model. Spine J.
15 (12), 25242537. doi:10.1016/j.spinee.2015.08.055
Misek, S. A., Chen, J., Schroeder, L., Rattanasinchai, C., Sample, A., Sarkaria, J. N.,
et al. (2017). EGFR Signals through a DOCK180-MLK3 Axis to Drive
Glioblastoma Cell Invasion. Mol. Cancer Res. 15 (8), 10851095. doi:10.
1158/1541-7786.MCR-16-0318
Mizushima, N. (2009). Physiological Functions of Autophagy. Curr. Top.
Microbiol. Immunol. 335, 7184. doi:10.1007/978-3-642-00302-8_3
Mohanakrishnan, V., Balasubramanian, A., Mahalingam, G., Partridge, N. C.,
Ramachandran, I., and Selvamurugan, N. (2018). Parathyroid Hormone-
induced Down-regulation of miR-532-5p for Matrix Metalloproteinase-13
Expression in Rat Osteoblasts. J. Cel. Biochem. 119 (7), 61816193. doi:10.
1002/jcb.26827
Moon, H. J., Kim, J. H., Lee, H. S., Chotai, S., Kang, J. D., Suh, J. K., et al. (2012).
Annulus Fibrosus Cells Interact with Neuron-like Cells to Modulate Production
of Growth Factors and Cytokines in Symptomatic Disc Degeneration. Spine
(1976) 37 (1), 29. doi:10.1097/BRS.0b013e31820cd2d8
Moon, H. J., Yurube, T., Lozito, T. P., Pohl, P., Hartman, R. A., Sowa, G. A., et al.
(2014). Effects of Secreted Factors in Culture Medium of Annulus Fibrosus
Cells on Microvascular Endothelial Cells: Elucidating the Possible
Pathomechanisms of Matrix Degradation and Nerve In-Growth in Disc
Degeneration. Osteoarthritis Cartilage 22 (2), 344354. doi:10.1016/j.joca.
2013.12.008
Mulcahy, L. A., Pink, R. C., and Carter, D. R. F. (2014). Routes and Mechanisms of
Extracellular Vesicle Uptake. J. Extracell. Vesicles 3, 24641. doi:10.3402/jev.v3.
24641
Munich, S., Sobo-Vujanovic, A., Buchser, W. J., Beer-Stolz, D., and Vujanovic, N. L.
(2012). Dendritic Cell Exosomes Directly Kill Tumor Cells and Activate Natural
Killer Cells via TNF Superfamily Ligands. Oncoimmunology 1 (7), 10741083.
doi:10.4161/onci.20897
Murai, K., Sakai, D., Sakai, D., Nakamura, Y., Nakai, T., Igarashi, T., et al. (2010).
Primary Immune System Responders to Nucleus Pulposus Cells: Evidence for
Immune Response in Disc Herniation. eCM 19, 1321. doi:10.22203/ecm.
v019a02
Neidlinger-Wilke, C., Liedert, A., Wuertz, K., Buser, Z., Rinkler, C., Käfer, W., et al.
(2009). Mechanical Stimulation Alters Pleiotrophin and Aggrecan Expression
by Human Intervertebral Disc Cells and Inuences Their Capacity to Stimulate
Endothelial Cell Migration. Spine (1976) 34 (7), 663669. doi:10.1097/BRS.
0b013e318194e20c
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214917
Li et al. Different Exosomes for IDD Therapy
Noriega, D. C., Ardura, F., Hernández-Ramajo, R., Martín-Ferrero, M. Á., Sánchez-
Lite, I., Toribio, B., et al. (2017). Intervertebral Disc Repair by Allogeneic
Mesenchymal Bone Marrow Cells. Transplantation 101 (8), 19451951. doi:10.
1097/TP.0000000000001484
Pan, B.-T., and Johnstone, R. M. (1983). Fate of the Transferrin Receptor during
Maturation of Sheep Reticulocytes In Vitro: Selective Externalization of the
Receptor. Cell 33 (3), 967978. doi:10.1016/0092-8674(83)90040-5
Pan, B. T., and Johnstone, R. (1984). Selective Externalization of the Transferrin
Receptor by Sheep Reticulocytes In Vitro. Response to Ligands and Inhibitors of
Endocytosis. J. Biol. Chem. 259 (15), 97769782. doi:10.1016/s0021-9258(17)
42767-0
Park, J.-B., Lee, J.-K., Cho, S.-T., Park, E.-Y., and Daniel Riew, K. (2007). A
Biochemical Mechanism for Resistance of Intervertebral Discs to Metastatic
Cancer: Fas Ligand Produced by Disc Cells Induces Apoptotic Cell Death of
Cancer Cells. Eur. Spine J. 16 (9), 13191324. doi:10.1007/s00586-007-0463-2
Parolini, O., Alviano, F., Bagnara, G. P., Bilic, G., Bühring, H. J., Evangelista, M.,
et al. (2008). Concise Review: Isolation and Characterization of Cells from
Human Term Placenta: Outcome of the First International Workshop on
Placenta Derived Stem Cells. Stem Cel. 26 (2), 300311. doi:10.1634/stemcells.
2007-0594
Pavathuparambil Abdul Manaph, N., Al-Hawwas, M., Bobrovskaya, L., Coates, P.
T., and Zhou, X.-F. (2018). Urine-derived Cells for Human Cell Therapy. Stem
Cel. Res. Ther 9 (1), 189. doi:10.1186/s13287-018-0932-z
Pohl, P. H. I., Lozito, T. P., Cuperman, T., Yurube, T., Moon, H. J., Ngo, K., et al.
(2016). Catabolic Effects of Endothelial Cell-Derived Microparticles on Disc
Cells: Implications in Intervertebral Disc Neovascularization and Degeneration.
J. Orthop. Res. 34 (8), 14661474. doi:10.1002/jor.23298
Pullig, O., Weseloh, G., Klatt, A. R., Wagener, R., and Swoboda, B. (2002). Matrilin-
3 in Human Articular Cartilage: Increased Expression in Osteoarthritis.
Osteoarthritis Cartilage 10 (4), 253263. doi:10.1053/joca.2001.0508
Qin, D., Long, T., Deng, J., and Zhang, Y. (2014). Urine-Derived Stem Cells for
PotentialUse in Bladder Repair. Stem Cel. Res. Ther. 5 (3), 69. doi:10.1186/scrt458
Raposo, G., and Stoorvogel, W. (2013). Extracellular Vesicles: Exosomes,
Microvesicles, and Friends. J. Cel. Biol. 200 (4), 373383. doi:10.1083/jcb.
201211138
Richardson, S. M., Kalamegam, G., Pushparaj, P. N., Matta, C., Memic, A.,
Khademhosseini, A., et al. (2016). Mesenchymal Stem Cells in Regenerative
Medicine: Focus on Articular Cartilage and Intervertebral Disc Regeneration.
Methods 99, 6980. doi:10.1016/j.ymeth.2015.09.015
Risbud, M. V., and Shapiro, I. M. (2011). Notochordal Cells in the Adult
Intervertebral Disc: New Perspective on an Old Question. Crit. Rev. Eukar
Gene Expr. 21 (1), 2941. doi:10.1615/critreveukargeneexpr.v21.i1.30
Risbud, M. V., and Shapiro, I. M. (2014). Role of Cytokines in Intervertebral Disc
Degeneration: Pain and Disc Content. Nat. Rev. Rheumatol. 10 (1), 4456.
doi:10.1038/nrrheum.2013.160
Rodrigues-Pinto, R., Ward, L., Humphreys, M., Zeef, L. A. H., Berry, A., Hanley, K.
P., et al. (2018). Human Notochordal Cell Transcriptome Unveils Potential
Regulators of Cell Function in the Developing Intervertebral Disc. Sci. Rep. 8
(1), 12866. doi:10.1038/s41598-018-31172-4
Rosenthal, A. K., Gohr, C. M., Mitton-Fitzgerald, E., Grewal, R., Ninomiya, J.,
Coyne, C. B., et al. (2015). Autophagy Modulates Articular Cartilage Vesicle
Formation in Primary Articular Chondrocytes. J. Biol. Chem. 290 (21),
1302813038. doi:10.1074/jbc.M114.630558
Rothman, A. M. K., Arnold, N. D., Pickworth, J. A., Iremonger, J., Ciuclan, L.,
Allen, R. M. H., et al. (2016). MicroRNA-140-5p and SMURF1 Regulate
Pulmonary Arterial Hypertension. J. Clin. Invest. 126 (7), 24952508. doi:10.
1172/JCI83361
Saggese, T., Thambyah, A., Wade, K., and McGlashan, S. R. (2020). Differential
Response of Bovine Mature Nucleus Pulposus and Notochordal Cells to
Hydrostatic Pressure and Glucose Restriction. Cartilage 11 (2), 221233.
doi:10.1177/1947603518775795
Sahoo, S., Klychko, E., Thorne, T., Misener, S., Schultz, K. M., Millay, M., et al.
(2011). Exosomes from Human CD34 + Stem Cells Mediate Their
Proangiogenic Paracrine Activity. Circ. Res. 109 (7), 724728. doi:10.1161/
CIRCRESAHA.111.253286
Sakai, D., and Andersson, G. B. J. (2015). Stem Cell Therapy for Intervertebral Disc
Regeneration: Obstacles and Solutions. Nat. Rev. Rheumatol. 11 (4), 243256.
doi:10.1038/nrrheum.2015.13
Sambrook, P. N., MacGregor, A. J., and Spector, T. D. (1999). Genetic Inuences on
Cervical and Lumbar Disc Degeneration: a Magnetic Resonance Imaging Study
in Twins. Arthritis Rheum. 42 (2), 366372. doi:10.1002/1529-0131(199902)42:
2<366:aid-anr20>3.0.co;2-6
Sampey, G. C., Meyering, S. S., Asad Zadeh, M., Saifuddin, M., Hakami, R. M., and
Kashanchi, F. (2014). Exosomes and Their Role in CNS Viral Infections.
J. Neurovirol. 20 (3), 199208. doi:10.1007/s13365-014-0238-6
Saravanan, P. B., Vasu, S., Yoshimatsu, G., Darden, C. M., Wang, X., Gu, J., et al.
(2019). Differential Expression and Release of Exosomal miRNAs by Human
Islets under Inammatory and Hypoxic Stress. Diabetologia 62 (10),
19011914. doi:10.1007/s00125-019-4950-x
Satoh, K., Konno, S., Nishiyama, K., Olmarker, K., and Kikuchi, S. (1999). Presence
and Distribution of Antigen-Antibody Complexes in the Herniated Nucleus
Pulposus. Spine (Phila Pa 1976) 24 (19), 19801984. doi:10.1097/00007632-
199910010-00003
Sawe, N., Steinberg, G., and Zhao, H. (2008). Dual Roles of the MAPK/ERK1/2 Cell
Signaling Pathway after Stroke. J. Neurosci. Res. 86 (8), 16591669. doi:10.1002/
jnr.21604
Scoville, W. B., and Corkill, G. (1973). Lumbar Disc Surgery: Technique of Radical
Removal and Early Mobilization. J. Neurosurg. 39 (2), 265269. doi:10.3171/jns.
1973.39.2.0265
Shen, J., Fang, J., Hao, J., Zhong, X., Wang, D., Ren, H., et al. (2016). SIRT1 Inhibits
the Catabolic Effect of IL-1βthrough TLR2/SIRT1/NF-κB Pathway in Human
Degenerative Nucleus Pulposus Cells. Pain Physician 19 (1), E215E226.
Shi, R., Wang, P.-Y., Li, X.-Y., Chen, J.-X., Li, Y., Zhang, X.-Z., et al. (2015).
Exosomal Levels of miRNA-21 from Cerebrospinal Fluids Associated with Poor
Prognosis and Tumor Recurrence of Glioma Patients. Oncotarget 6 (29),
2697126981. doi:10.18632/oncotarget.4699
Shi, Y., Wang, Y., Li, Q., Liu, K., Hou, J., Shao, C., et al. (2018).
Immunoregulatory Mechanisms of Mesenchymal Stem and Stromal Cells
in Inammatory Diseases. Nat. Rev. Nephrol. 14 (8), 493507. doi:10.1038/
s41581-018-0023-5
Silva, J., Garcia, V., Zaballos, A., Provencio, M., Lombardia, L., Almonacid, L., et al.
(2011). Vesicle-related microRNAs in Plasma of Nonsmall Cell Lung Cancer
Patients and Correlation with Survival. Eur. Respir. J. 37 (3), 617623. doi:10.
1183/09031936.00029610
Skog, J., Würdinger, T., van Rijn, S., Meijer, D. H., Gainche, L., Curry, W. T., et al.
(2008). Glioblastoma Microvesicles Transport RNA and Proteins that Promote
Tumour Growth and Provide Diagnostic Biomarkers. Nat. Cel. Biol. 10 (12),
14701476. doi:10.1038/ncb1800
Song, Y., Li, S., Geng, W., Luo, R., Liu, W., Tu, J., et al. (2018). Sirtuin 3-dependent
Mitochondrial Redox Homeostasis Protects against AGEs-Induced
Intervertebral Disc Degeneration. Redox Biol. 19, 339353. doi:10.1016/j.
redox.2018.09.006
Song, J., Chen, Z.-H., Zheng, C.-J., Song, K.-H., Xu, G.-Y., Xu, S., et al. (2020).
Exosome-Transported circRNA_0000253 Competitively Adsorbs MicroRNA-
141-5p and Increases IDD. Mol. Ther. - Nucleic Acids 21, 10871099. doi:10.
1016/j.omtn.2020.07.039
Steck, E., Bertram, H., Abel, R., Chen, B., Winter, A., and Richter, W. (2005).
Induction of Intervertebral Disc-like Cells from Adult Mesenchymal Stem
Cells. Stem Cells 23 (3), 403411. doi:10.1634/stemcells.2004-0107
Stefanakis, M., Al-Abbasi, M., Harding, I., Pollintine, P., Dolan, P., Tarlton, J., et al.
(2012). Annulus Fissures Are Mechanically and Chemically Conducive to the
Ingrowth of Nerves and Blood Vessels. Spine (1976) 37 (22), 18831891. doi:10.
1097/BRS.0b013e318263ba59
Stefater, J. A., III, Lewkowich, I., Rao, S., Mariggi, G., Carpenter, A. C., Burr, A. R.,
et al. (2011). Regulation of Angiogenesis by a Non-canonical Wnt-Flt1 Pathway
in Myeloid Cells. Nature 474 (7352), 511515. doi:10.1038/nature10085
Strassburg, S., Hodson, N. W., Hill, P. I., Richardson, S. M., and Hoyland, J. A.
(2012). Bi-directional Exchange of Membrane Components Occurs during Co-
Culture of Mesenchymal Stem Cells and Nucleus Pulposus Cells. PLoS One 7
(3), e33739. Epub 2012/03/23. doi:10.1371/journal.pone.0033739
Su, T., Xiao, Y., Xiao, Y., Guo, Q., Li, C., Huang, Y., et al. (2019). Bone Marrow
Mesenchymal Stem Cells-Derived Exosomal MiR-29b-3p Regulates Aging-
Associated Insulin Resistance. ACS Nano 13 (2), 24502462. doi:10.1021/
acsnano.8b09375
Sun, Z., Guo, Y.-S., Yan, S.-J., Wan, Z.-Y., Gao, B., Wang, L., et al. (2013). CK8
Phosphorylation Induced by Compressive Loads Underlies the Downregulation
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214918
Li et al. Different Exosomes for IDD Therapy
of CK8 in Human Disc Degeneration by Activating Protein Kinase C. Lab.
Invest. 93 (12), 13231330. doi:10.1038/labinvest.2013.122
Sun, Z., Wan, Z. Y., Guo, Y. S., Wang, H. Q., and Luo, Z. J. (2013). FasL on Human
Nucleus Pulposus Cells Prevents Angiogenesis in the Disc by Inducing Fas-
Mediated Apoptosis of Vascular Endothelial Cells. Int. J. Clin. Exp. Pathol. 6
(11), 23762385.
Sun, Z., Liu, B., and Luo, Z.-J. (2020). The Immune Privilege of the Intervertebral
Disc: Implications for Intervertebral Disc Degeneration Treatment. Int. J. Med.
Sci. 17 (5), 685692. doi:10.7150/ijms.42238
Sun, Z., Liu, B., Liu, Z.-H., Song, W., Wang, D., Chen, B.-Y., et al. (2020).
Notochordal-Cell-Derived Exosomes Induced by Compressive Load Inhibit
Angiogenesis via the miR-140-5p/Wnt/β-Catenin Axis. Mol. Ther. - Nucleic
Acids 22, 10921106. doi:10.1016/j.omtn.2020.10.021
Sun, Z., Zhao, H., Liu, B., Gao, Y., Tang, W.-H., Liu, Z.-H., et al. (2021). AF Cell
Derived Exosomes Regulate Endothelial Cell Migration and Inammation:
Implications for Vascularization in Intervertebral Disc Degeneration. Life Sci.
265, 118778. doi:10.1016/j.lfs.2020.118778
Suryaraja, R., Anitha, M., Anbarasu, K., Kumari, G., and Mahalingam, S. (2013).
The E3 Ubiquitin Ligase Itch Regulates Tumor Suppressor Protein RASSF5/
NORE1 Stability in an Acetylation-dependent Manner. Cel. Death Dis. 4, e565.
doi:10.1038/cddis.2013.91
Takada, T., Nishida, K., Maeno, K., Kakutani, K., Yurube, T., Doita, M., et al.
(2012). Intervertebral Disc and Macrophage Interaction Induces Mechanical
Hyperalgesia and Cytokine Production in a Herniated Disc Model in Rats.
Arthritis Rheum. 64 (8), 26012610. doi:10.1002/art.34456
Tao, S.-C., Guo, S.-C., and Zhang, C.-Q. (2018). Modularized Extracellular
Vesicles: The Dawn of Prospective Personalized and Precision Medicine.
Adv. Sci. 5 (2), 1700449. doi:10.1002/advs.201700449
Taylor, V. M., Deyo, R. A., Cherkin, D. C., and Kreuter, W. (1994). Low Back
Pain Hospitalization. Recent United States Trends and Regional Variations.
Spine (Phila Pa 1976) 19 (11), 12071213. doi:10.1097/00007632-199405310-
00002
Tian, T., Zhu, Y.-L., Hu, F.-H., Wang, Y.-Y., Huang, N.-P., and Xiao, Z.-D. (2013).
Dynamics of Exosome Internalization and Trafcking. J. Cel. Physiol. 228 (7),
14871495. doi:10.1002/jcp.24304
Tibrewal, S. B., and Pearcy, M. J. (1985). Lumbar Intervertebral Disc Heights in
normal Subjects and Patients with Disc Herniation. Spine (Phila Pa 1976) 10
(5), 452454. doi:10.1097/00007632-198506000-00009
Tolonen, J., Grönblad, M., Virri, J., Seitsalo, S., Rytömaa, T., and Karaharju, E. O.
(1997). Platelet-derived Growth Factor and Vascular Endothelial Growth
Factor Expression in Disc Herniation Tissue: an Immunohistochemical
Study. Eur. Spine J. 6 (1), 6369. doi:10.1007/bf01676576
Tooi, M., Komaki, M., Morioka, C., Honda, I., Iwasaki, K., Yokoyama, N., et al.
(2016). Placenta Mesenchymal Stem Cell Derived Exosomes Confer Plasticity
on Fibroblasts. J. Cel. Biochem. 117 (7), 16581670. doi:10.1002/jcb.25459
van den Boorn, J. G., Schlee, M., Coch, C., and Hartmann, G. (2011). SiRNA
Delivery with Exosome Nanoparticles. Nat. Biotechnol. 29 (4), 325326. doi:10.
1038/nbt.1830
van Dommelen, S. M., Vader, P., Lakhal, S., Kooijmans, S. A. A., van Solinge, W.
W., Wood, M. J. A., et al. (2012). Microvesicles and Exosomes: Opportunities
for Cell-Derived Membrane Vesicles in Drug Delivery. J. Control. Release 161
(2), 635644. doi:10.1016/j.jconrel.2011.11.021
Vergroesen, P.-P. A., Kingma, I., Emanuel, K. S., Hoogendoorn, R. J. W., Welting,
T. J., van Royen, B. J., et al. (2015). Mechanics and Biology in Intervertebral Disc
Degeneration: a Vicious circle. Osteoarthritis Cartilage 23 (7), 10571070.
doi:10.1016/j.joca.2015.03.028
Vo, N. V., Hartman, R. A., Yurube, T., Jacobs, L. J., Sowa, G. A., and Kang, J. D.
(2013). Expression and Regulation of Metalloproteinases and Their Inhibitors
in Intervertebral Disc Aging and Degeneration. Spine J. 13 (3), 331341. doi:10.
1016/j.spinee.2012.02.027
Vogt, M. T., Hanscom, B., Lauerman, W. C., and Kang, J. D. (2002). Inuence of
Smoking on the Health Status of Spinal Patients: the National Spine Network
Database. Spine (Phila Pa 1976) 27 (3), 313319. doi:10.1097/00007632-
200202010-00022
Vojtech, L., Woo, S., Hughes, S., Levy, C., Ballweber, L., Sauteraud, R. P., et al.
(2014). Exosomes in Human Semen Carry a Distinctive Repertoire of Small
Non-coding RNAs with Potential Regulatory Functions. Nucleic Acids Res. 42
(11), 72907304. doi:10.1093/nar/gku347
Wang, M., and Kaufman, R. J. (2016). Protein Misfolding in the Endoplasmic
Reticulum as a Conduit to Human Disease. Nature 529 (7586), 326335. doi:10.
1038/nature17041
Wang, H. Q., and Samartzis, D. (2014). Clarifying the Nomenclature of
Intervertebral Disc Degeneration and Displacement: from Bench to Bedside.
Int. J. Clin. Exp. Pathol. 7 (4), 12931298.
Wang, D., Nasto, L. A., Roughley, P., Leme, A. S., Houghton, A. M., Usas, A., et al.
(2012). Spine Degeneration in a Murine Model of Chronic Human Tobacco
Smokers. Osteoarthritis Cartilage 20 (8), 896905. doi:10.1016/j.joca.2012.
04.010
Wang, C., Gonzales, S., Levene, H., Gu, W., and Huang, C.-Y. C. (2013). Energy
Metabolism of Intervertebral Disc under Mechanical Loading. J. Orthop. Res. 31
(11), 17331738. doi:10.1002/jor.22436
Wang, T., Li, P., Ma, X., Tian, P., Han, C., Zang, J., et al. (2015). MicroRNA-494
Inhibition Protects Nucleus Pulposus Cells from TNF-α-Induced Apoptosis by
Targeting JunD. Biochimie 115, 17. doi:10.1016/j.biochi.2015.04.011
Wang, H., He, P., Pan, H., Long, J., Wang, J., Li, Z., et al. (2018 ). Circular RNA Circ-
4099 Is Induced by TNF-αand Regulates ECM Synthesis by Blocking miR-616-
5p Inhibition of Sox9 in Intervertebral Disc Degeneration. Exp. Mol. Med. 50
(4), 114. doi:10.1038/s12276-018-0056-7
Wang, D., Zhu, H., Cheng, W., Lin, S., Shao, R., and Pan, H. (2019). Effects of
Hypoxia and ASIC3 on Nucleus Pulposus Cells: From Cell Behavior to
Molecular Mechanism. Biomed. Pharmacother. 117, 109061. doi:10.1016/j.
biopha.2019.109061
Wang, Y., Che, M., Xin, J., Zheng, Z., Li, J., and Zhang, S. (2020). The Role of IL-1β
and TNF-αin Intervertebral Disc Degeneration. Biomed. Pharmacother. 131,
110660. doi:10.1016/j.biopha.2020.110660
Wang, D., He, X., Wang, D., Peng, P., Xu, X., Gao, B., et al. (2020). Quercetin
Suppresses Apoptosis and Attenuates Intervertebral Disc Degeneration via the
SIRT1-Autophagy Pathway. Front. Cel Dev. Biol. 8, 613006. doi:10.3389/fcell.
2020.613006
Wang, Z., Wu, Y., Zhao, Z., Liu, C., and Zhang, L. (2021). Study on Transorgan
Regulation of Intervertebral Disc and Extra-Skeletal Organs through Exosomes
Derived from Bone Marrow Mesenchymal Stem Cells. Front. Cel Dev. Biol. 9,
741183. doi:10.3389/fcell.2021.741183
Wiet, M. G., Piscioneri, A., Khan, S. N., Ballinger, M. N., Hoyland, J. A., and
Purmessur, D. (2017). Mast Cell-Intervertebral Disc Cell Interactions Regulate
Inammation, Catabolism and Angiogenesis in Discogenic Back Pain. Sci. Rep.
7 (1), 12492. doi:10.1038/s41598-017-12666-z
Wu, S., Wang, Z., Bharadwaj, S., Hodges, S. J., Atala, A., and Zhang, Y. (2011).
Implantation of Autologous Urine Derived Stem Cells Expressing Vascular
Endothelial Growth Factor for Potential Use in Genitourinary Reconstruction.
J. Urol. 186 (2), 640647. doi:10.1016/j.juro.2011.03.152
Wu, S., Liu, Y., Bharadwaj, S., Atala, A., and Zhang, Y. (2011). Human Urine-
Derived Stem Cells Seeded in a Modied 3D Porous Small Intestinal
Submucosa Scaffold for Urethral Tissue Engineering. Biomaterials 32 (5),
13171326. doi:10.1016/j.biomaterials.2010.10.006
Wu, T., Lin, Y., and Xie, Z. (2018). MicroRNA-1247 Inhibits Cell Proliferation by
Directly Targeting ZNF346 in Childhood Neuroblastoma. Biol. Res. 51 (1), 13.
doi:10.1186/s40659-018-0162-y
Wu, X., Cai, Y., Lu, S., Xu, K., Shi, X., Yang, L., et al. (2019). Intra-articular Injection
of Chloramphenicol Reduces Articular Cartilage Degeneration in a Rabbit
Model of Osteoarthritis. Clin. Orthop. Relat. Res. 477 (12), 27852797. doi:10.
1097/CORR.0000000000001016
Wu, P., Zhang, B., Han, X., Sun, Y., Sun, Z., Li, L., et al. (2021). HucMSC Exosome-
Delivered 14-3-3ζAlleviates Ultraviolet Radiation-Induced Photodamage via
SIRT1 Pathway Modulation. Aging 13 (8), 1154211563. doi:10.18632/aging.
202851
Xia, C., Zeng, Z., Fang, B., Tao, M., Gu, C., Zheng, L., et al. (2019). Mesenchymal
Stem Cell-Derived Exosomes Ameliorate Intervertebral Disc Degeneration via
Anti-oxidant and Anti-inammatory Effects. Free Radic. Biol. Med. 143, 115.
doi:10.1016/j.freeradbiomed.2019.07.026
Xiang, H., Su, W., Wu, X., Chen, W., Cong, W., Yang, S., et al. (2020). Exosomes
Derived from Human Urine-Derived Stem Cells Inhibit Intervertebral Disc
Degeneration by Ameliorating Endoplasmic Reticulum Stress. Oxid. Med. Cel.
Longev. 2020, 121. doi:10.1155/2020/6697577
Xie, N., Li, Z., Adesanya, T. M., Guo, W., Liu, Y., Fu, M., et al. (2016).
Transplantation of Placenta-derived Mesenchymal Stem Cells Enhances
Frontiers in Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214919
Li et al. Different Exosomes for IDD Therapy
Angiogenesis after Ischemic Limb Injury in Mice. J. Cel. Mol. Med. 20 (1),
2937. doi:10.1111/jcmm.12489
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 (1), 264. doi:10.1186/s12951-021-00991-5
Yang, S., Yuan, Y., Jiao, S., Luo, Q., and Yu, J. (2016). Calcitonin Gene-Related
Peptide Protects Rats from Cerebral Ischemia/reperfusion Injury via a
Mechanism of Action in the MAPK Pathway. Biomed. Rep. 4 (6), 699703.
doi:10.3892/br.2016.658
Yang, H., Wen, Y., Zhang, M., Liu, Q., Zhang, H., Zhang, J., et al. (2020). MTORC1
Coordinates the Autophagy and Apoptosis Signaling in Articular Chondrocytes
in Osteoarthritic Temporomandibular Joint. Autophagy 16 (2), 271288.
doi:10.1080/15548627.2019.1606647
Yeo, R. W. Y., Lai, R. C., Zhang, B., Tan, S. S., Yin, Y., Teh, B. J., et al. (2013).
Mesenchymal Stem Cell: an Efcient Mass Producer of Exosomes for Drug
Delivery. Adv. Drug Deliv. Rev. 65 (3), 336341. doi:10.1016/j.addr.2012.07.001
Yoon, S. B., and Chang, J. H. (2017). Extracellular Vesicles in Bile: a Game Changer
in the Diagnosis of Indeterminate Biliary Stenoses? Hepatobiliary Surg. Nutr. 6
(6), 408410. doi:10.21037/hbsn.2017.10.01
Yorimitsu, E., Chiba, K., Toyama, Y., and Hirabayashi, K. (2001). Long-term
Outcomes of Standard Discectomy for Lumbar Disc Herniation: a Follow-Up
Study of More Than 10 Years. Spine (Phila Pa 1976) 26 (6), 652657. doi:10.
1097/00007632-200103150-00019
Yoshida, Y., Yamamoto, H., Morita, R., Oikawa, R., Matsuo, Y., Maehata, T., et al.
(2014). Detection of DNA Methylation of Gastric Juice-Derived Exosomes in
Gastric Cancer. Integr. Mol. Med. 7 (7), e184. doi:10.15761/IMM.1000105
Yuan, Z., Bedi, B., and Sadikot, R. T. (2018). Bronchoalveolar Lavage Exosomes in
Lipopolysaccharide-Induced Septic Lung Injury. JoVE 135, 57737. doi:10.3791/57737
Yuan, Q., Wang, X., Liu, L.,Cai, Y., Zhao, X., Ma, H.,et al. (2020). ExosomesDerived
from Human Placental Mesenchymal Stromal Cells Carrying AntagomiR-4450
Alleviate Intervertebral Disc Degeneration through Upregulation of ZNF121.
Stem Cel. Dev. 29 (16), 10381058. doi:10.1089/scd.2020.0083
Zahavi-Goldstein, E., Blumenfeld, M., Fuchs-Telem, D., Pinzur, L., Rubin, S.,
Aberman, Z., et al. (2017). Placenta-derived PLX-PAD Mesenchymal-like
Stromal Cells Are Efcacious in Rescuing Blood Flow in Hind Limb
Ischemia Mouse Model by a Dose- and Site-dependent Mechanism of
Action. Cytotherapy 19 (12), 14381446. doi:10.1016/j.jcyt.2017.09.010
Zhang, J., Lu, L., Xiong, Y., Qin, W., Zhang, Y., Qian, Y., et al. (2014). MLK3
Promotes Melanoma Proliferation and Invasion and Is a Target of microRNA-
125b. Clin. Exp. Dermatol. 39 (3), 376384. doi:10.1111/ced.12286
Zhang, D., Zheng, L., Shi, H., Chen, X., Wan, Y., Zhang, H., et al. (2014).
Suppression of Peritoneal Tumorigenesis by Placenta-Derived Mesenchymal
Stem Cells Expressing Endostatin on Colorectal Cancer. Int. J. Med. Sci. 11 (9),
870879. doi:10.7150/ijms.8758
Zhang, Q., Lou, Y., Zhang, J., Fu, Q., Wei, T., Sun, X., et al. (2017). Hypoxia-
inducible Factor-2αPromotes Tumor Progression and Has Crosstalk with
Wnt/β-Catenin Signaling in Pancreatic Cancer. Mol. Cancer 16 (1), 119. doi:10.
1186/s12943-017-0689-5
Zhang, H., Wang, L., Li, C., Yu, Y., Yi, Y., Wang, J., et al. (2019). Exosome-Induced
Regulation in Inammatory Bowel Disease. Front. Immunol. 10, 1464. doi:10.
3389/mmu.2019.01464
Zhang, Z. G., Buller, B., and Chopp, M. (2019). Exosomes - beyond Stem Cells for
Restorative Therapy in Stroke and Neurological Injury. Nat. Rev. Neurol. 15 (4),
193203. doi:10.1038/s41582-018-0126-4
Zhang, Z., Lin, J., Tian, N., Wu, Y., Zhou, Y., Wang, C., et al. (2019). Melatonin
Protects Vertebral Endplate Chondrocytes against Apoptosis and Calcication
via the Sirt1-Autophagy Pathway. J. Cel. Mol. Med. 23 (1), 177193. doi:10.
1111/jcmm.13903
Zhang, J., Zhang, J., Zhang, Y., Liu, W., Ni, W., Huang, X., et al. (2020).
Mesenchymal Stem Cells-derived Exosomes Ameliorate Intervertebral Disc
Degeneration through Inhibiting Pyroptosis. J. Cel. Mol. Med. 24 (20),
1174211754. doi:10.1111/jcmm.15784
Zhang, Q. C., Hu, S. Q., Hu, A. N., Zhang, T. W., Jiang, L. B., and Li, X. L. (2021).
Autophagy-activated Nucleus Pulposus Cells Deliver Exosomal miR-27a to
Prevent Extracellular Matrix Degradation by Targeting MMP-13. J. Orthop.
Res. 39 (9), 19211932. doi:10.1002/jor.24880
Zhang, H.-J., Liao, H.-Y., Bai, D.-Y., Wang, Z.-Q., and Xie, X.-W. (2021). MAPK/
ERK Signaling Pathway: A Potential Target for the Treatment of Intervertebral
Disc Degeneration. Biomed. Pharmacother. 143, 112170. doi:10.1016/j.biopha.
2021.112170
Zhang, X., Cai, Z., Wu, M., Huangfu, X., Li, J., and Liu, X. (2021). Adipose Stem
Cell-Derived Exosomes Recover Impaired Matrix Metabolism of Torn Human
Rotator Cuff Tendons by Maintaining Tissue Homeostasis. Am. J. Sports Med.
49 (4), 899908. doi:10.1177/0363546521992469
Zhao, B., Yu, Q., Li, H., Guo, X., and He, X. (2014). Characterization of microRNA
Expression Proles in Patients with Intervertebral Disc Degeneration. Int.
J. Mol. Med. 33 (1), 4350. doi:10.3892/ijmm.2013.1543
Zhao, X., Li, Y., Lin, X., Wang, J., Zhao, X., Xie, J., et al. (2018). Ozone Induces
Autophagy in Rat Chondrocytes Stimulated with IL-1βthrough the AMPK/
mTOR Signaling Pathway. Jpr 11, 30033017. doi:10.2147/JPR.S183594
Zhou, Y., Zhou, G., Tian, C., Jiang, W., Jin, L., Zhang, C., et al. (2016). Exosome-
mediated Small RNA Delivery for Gene Therapy. WIREs RNA 7(6),758771.
doi:10.1002/wrna.1363
Zhu, G., Yang, X., Peng, C., Yu, L., and Hao, Y. (2020). Exosomal miR-532-5p from
Bone Marrow Mesenchymal Stem Cells Reduce Intervertebral Disc
Degeneration by Targeting RASSF5. Exp. Cel. Res. 393 (2), 112109. doi:10.
1016/j.yexcr.2020.112109
Zhu, L., Shi, Y., Liu, L., Wang, H., Shen, P., and Yang, H. (2020). Mesenchymal
Stem Cells-Derived Exosomes Ameliorate Nucleus Pulposus Cells Apoptosis
via Delivering miR-142-3p: Therapeutic Potential for Intervertebral Disc
Degenerative Diseases. Cel. Cycle 19 (14), 17271739. doi:10.1080/15384101.
2020.1769301
Zlotogorski-Hurvitz, A., Dayan, D., Chaushu, G., Korvala, J., Salo, T., Sormunen,
R., et al. (2015). Human Saliva-Derived Exosomes. J. Histochem. Cytochem. 63
(3), 181189. doi:10.1369/0022155414564219
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.
Copyright © 2022 Li, Zhang, Wang, Zhang, Shi, Zhang, Wang, Ding, Xu, Gao and
Yan. 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 copyrigh t 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 Cell and Developmental Biology | www.frontiersin.org February 2022 | Volume 9 | Article 82214920
Li et al. Different Exosomes for IDD Therapy
... The survival and adaption of exogenous transplanted cells in intervertebral disc were questioned (72), which raised interest in endogenous intervertebral disc regeneration and cell-free therapy. With the identification of the natural existence of stem/progenitor cells in degenerate intervertebral disc (114), possible approaches were conceived to overcome the endogenous repair failure in natural IDD process (77,101,116,115). In the most recent papers, therapies based on MSC-derived exosomes (116,117,118) or notochordal cells (119,120) were regarded as novel strategies for ameliorating IDD, with a relative preference for exosomes according to the number of articles. ...
... With the identification of the natural existence of stem/progenitor cells in degenerate intervertebral disc (114), possible approaches were conceived to overcome the endogenous repair failure in natural IDD process (77,101,116,115). In the most recent papers, therapies based on MSC-derived exosomes (116,117,118) or notochordal cells (119,120) were regarded as novel strategies for ameliorating IDD, with a relative preference for exosomes according to the number of articles. ...
Article
Full-text available
• Mesenchymal stromal cells (MSCs) are important potential candidates for regenerative therapy for intervertebral disc degeneration (IDD). This scientometric study aimed to summarize the main research trends, identify current research hotspots, and measure the networks of the contributors and their scientific productivity. • A total of 1102 publications regarding MSC in IDD were recognized from January 2000 to April 2022. The number of records every year followed an overall uptrend with fluctuations. • The main trend of research demonstrated the practice of gradually applying MSC-based therapy to IDD with the assistance of advances in biomaterials and IDD pathology. A recent focus on MSC-derived exosomes and notochordal cells was detected. • The basic studies in this field were mainly contributed to by Japan, the USA, and European countries, while China dominated in the number of recent publications. Tokai University with Daisuke Sakai was the most productive contributor. • Cell biology, tissue engineering, and biomaterials were the categories with deep engagement in research of this field.
... This study further proposed that personalized therapeutic strategies are needed to be formulated based on specific monocyte/ macrophage subtypes and degenerative stages of IDD. KEYWORDS intervertebral disk degeneration, monocytes/macrophage subtypes evolution, oxidative stress, trajectory lineage analysis, single-cell transcriptome landscape, longitudinal approach Introduction Low back pain (LBP) has continuously been the major cause of disability in human adults, affecting almost 80% of the population worldwide, leading to heavy socioeconomic burden (1,2). There are lots of triggers of LBP, and intervertebral disk degeneration (IDD) is considered as one of the common reasons (3). ...
... Increasing evidence has reported that despite multifactorial etiology like loading changes, smoking, senescence, and poor nutrient supply, genetic factor is the essential risk of IDD, which accounts for more than 70% of the risks (2). Meanwhile, as the largest avascular organ within body, IVD consists of the middle nucleus pulposus (NP), surrounding annulus fibrosis (AF), and cartilage endplate (CEP) on the inferior and superior sides. ...
Article
Full-text available
Intervertebral disk degeneration (IDD) is a chronic inflammatory disease with intricate connections between immune infiltration and oxidative stress (OS). Complex cell niches exist in degenerative intervertebral disk (IVD) and interact with each other and regulate the disk homeostasis together. However, few studies have used longitudinal approach to describe the immune response of IDD progression. Here, we conducted conjoint analysis of bulk-RNA sequencing and single-cell sequencing, together with a series of techniques like weighted gene co-expression network analysis (WGCNA), immune infiltration analysis, and differential analysis, to systematically decipher the difference in OS-related functions of different cell populations within degenerative IVD tissues, and further depicted the longitudinal alterations of immune cells, especially monocytes/macrophages in the progression of IDD. The OS-related genes CYP1A1, MMP1, CCND1, and NQO1 are highly expressed and might be diagnostic biomarkers for the progression of IDD. Further landscape of IVD microenvironment showed distinct changes in cell proportions and characteristics at late degeneration compared to early degeneration of IDD. Monocytes/macrophages were classified into five distinct subpopulations with different roles. The trajectory lineage analysis revealed transcriptome alterations from effector monocytes/macrophages and regulatory macrophages to other subtypes during the evolution process and identified monocytes/macrophage subpopulations that had rapidly experienced the activation of inflammatory or anti-inflammatory responses. This study further proposed that personalized therapeutic strategies are needed to be formulated based on specific monocyte/macrophage subtypes and degenerative stages of IDD.
... The immune homeostasis plays an important role in the process of IDD and is maintained by a variety of immune cells, such as B cells, T Am J Transl Res 2024; 16(5): [1891][1892][1893][1894][1895][1896][1897][1898][1899][1900][1901][1902][1903][1904][1905][1906] cells, and macrophages [4]. Recently, macrophages have received significant attention in the IDD process. ...
... In recent years, lower back pain has become an important social and medical problem worldwide and one of the causes of total disability in elderly individuals; this condition not only causes great pain and psychological burdens to patients and their families but also consumes a large amount of medical resources (1). Globally, it has been reported that approximately 60 to 80% of people will experience lower back pain symptoms (2). ...
Article
Full-text available
Intervertebral disk (IVD) degeneration (IVDD) is a main factor in lower back pain, and immunomodulation plays a vital role in disease progression. The IVD is an immune privileged organ, and immunosuppressive molecules in tissues reduce immune cell (mainly monocytes/macrophages and mast cells) infiltration, and these cells can release proinflammatory cytokines and chemokines, disrupting the IVD microenvironment and leading to disease progression. Improving the inflammatory microenvironment in the IVD through immunomodulation during IVDD may be a promising therapeutic strategy. This article reviews the normal physiology of the IVD and its degenerative mechanisms, focusing on IVDD-related immunomodulation, including innate immune responses involving Toll-like receptors, NOD-like receptors and the complement system and adaptive immune responses that regulate cellular and humoral immunity, as well as IVDD-associated immunomodulatory therapies, which mainly include mesenchymal stem cell therapies, small molecule therapies, growth factor therapies, scaffolds, and gene therapy, to provide new strategies for the treatment of IVDD.
Article
Full-text available
Background Intervertebral disc degeneration (IVDD) is a common chronic disease in orthopedics, and its molecular mechanisms are still not well explained. Aim This study's objective was to bioinformatics‐based discovery of IVDD biomarkers and immune‐inflammatory infiltrates. Materials and Methods The IVDD illness gene collection was gathered from GeneCards, DisGeNet, and gene expression profiles were chosen from the extensive Gene Expression Omnibus database (GSE124272, GSE150408, and GSE153761). The STRING database was used to create a network of protein–protein interactions, while the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) databases were used for functional enrichment analysis. Using hub genes, the immune cell infiltration between IVDD patient samples and control tissues was examined. Finally, quantitative polymerase chain reaction and Western blot experiments were used to verify the expression of hub genes. Results A total of 27 differentially expressed hub genes were identified by bioinformatics. According to GO and KEGG analyses, hub genes were prominent in immunological responses, chemokine‐mediated signaling pathways, and inflammatory responses, with the key signaling pathways engaged in cellular senescence, apoptosis, Th1 and Th2 cell differentiation, and Th17 cell differentiation. Immune cell infiltration research revealed that T cells, lymphocytes, B cells, and NK cells were decreased in IVDD patients while monocytes, neutrophils, and CD8 T cells were increased. The expression levels of the senescence hub genes SP1, VEGFA, IL‐6, and the apoptosis key gene CASP3 were considerably greater in the IVDD model group than in the control group, according to in vitro validation. Conclusion In conclusion, the cellular senescence signaling pathway, the apoptosis signaling pathway, and associated hub genes play significant roles in the development and progression of IVDD, this finding may help direct future research on the senescence signaling route in IVDD.
Article
Full-text available
A major pathological basis for low back pain is intervertebral disk degeneration, which is primarily caused by the degeneration of nucleus pulposus cells due to imbalances in extracellular matrix (ECM) anabolism and catabolism. The phenotype of macrophages in the local immune microenvironment greatly influences the balance of ECM metabolism. Therefore, the control over the macrophage phenotype of the ECM is promising to repair intervertebral disk degeneration. Herein, the preparation of an injectable nanocomposite hydrogel is reported by embedding epigallocatechin‐3‐gallate‐coated hydroxyapatite nanorods in O‐carboxymethyl chitosan cross‐linked with aldehyde hyaluronic acid that is capable of modulating the phenotype of macrophages. The bioactive components play a primary role in repairing the nucleus pulposus, where the hydroxyapatite nanorods can promote anabolism in the ECM through the nucleopulpogenic differentiation of mesenchymal stem cells. In addition, epigallocatechin‐3‐gallate can decrease catabolism in the ECM in nucleus pulposus by inducing M2 macrophage polarization, which exists in normal intervertebral disks and can alleviate degeneration. The nanocomposite hydrogel system shows promise for the minimally invasive and effective treatment of intervertebral disk degeneration by controlling anabolism and catabolism in the ECM and inhibiting the IL17 signaling pathway (M1‐related pathway) in vitro and in vivo.
Article
Full-text available
Millions of people worldwide suffer from intervertebral disc degeneration (IVDD), which imposes a significant socioeconomic burden on society. There is an urgent clinical demand for more effective treatments for IVDD because conventional treatments can only alleviate the symptoms rather than preventing the progression of IVDD. Hydrogels, a class of elastic biomaterials with good biocompatibility, are promising candidates for intervertebral disc repair and regeneration. In recent years, various hydrogels have been investigated in vitro and in vivo for the repair of intervertebral discs, some of which are ready for clinical testing. This review summarizes the latest findings and developments in using bioactive factors-released bioactive injectable hydrogels for the repair and regeneration of intervertebral discs. It focuses on the analysis and summary of the use of multifunctional injectable hydrogels to delivery bioactive factors (cells, exosomes, growth factors, genes, drugs) for disc regeneration, providing guidance for future study. Finally, we discussed and analyzed the optimal timing for the application of controlled-release hydrogels in the treatment of IVDD to meet the high standards required for intervertebral disc regeneration and precision medicine.
Preprint
Full-text available
Intervertebral disc degeneration (IVDD) is a common chronic disease in orthopedics, and its molecular mechanisms are still not well explained. This study's objective was to bioinformatics-based discovery of IVDD biomarkers and immune- inflammatory infiltrates. The IVDD illness gene collection was gathered from GeneCards, DisGeNet, and gene expression profiles were chosen from the extensive gene expression omnibus database (GSE124272, GSE150408, and GSE153761). The STRING database was used to create network of protein-protein interactions, while the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) databases were used for functional enrichment analysis. Using hub genes, the immune cell infiltration between IVDD patient samples and control tissues was examined. Finally, qPCR and western blot experiments were used to verify the expression of hub genes. A total of 27 differentially expressed hub genes were identified by bioinformatics. According to GO and KEGG analyses, hub genes were prominent in immunological responses, chemokine-mediated signaling pathways, and inflammatory responses, with the key signaling pathways engaged in Cellular Senescence, Apoptosis, Th1 and Th2 cell differentiation. Immune cell infiltration research revealed that T cells, lymphocytes, B cells, and NK cells were decreased in IVDD patients while monocytes, neutrophils, and CD8 T cells were increased. The expression levels of the senescence hub genes SP1, VEGFA, IL-6, and the apoptosis key gene CASP3 were considerably greater in the IVDD model group than in the control group, according to in vitro validation. In conclusion, the cellular senescence signaling pathway, the apoptosis signaling pathway, and associated hub genes all play significant roles in the development and progression of IVDD, this finding may help direct future research on the senescence signaling route in IVDD.
Article
Full-text available
Intervertebral disc degeneration (IDD) is the basic pathological process of many degenerative diseases of the spine, characterized by series of symptoms, among which low back pain (LBP) is the most common symptom that patients suffer a lot, which not only makes patients and individual families bear a huge pain and psychological burden, but also consumes a lot of medical resources. IDD is usually thought to be relevant with various factors such as genetic predisposition, trauma and aging, and IDD progression is tightly relevant with structural and functional alterations. IDD processes are caused by series of pathological processes, including oxidative stress, matrix decomposition, inflammatory reaction, apoptosis, abnormal proliferation, cell senescence, autophagy as well as sepsis process, among which the oxidative stress and inflammatory response are considered as key link in IDD. The production and clearance of ROS are tightly connected with oxidative stress, which would further simulate various signaling pathways. The phenotype of disc cells could change from matrix anabolism-to matrix catabolism- and proinflammatory-phenotype during IDD. Recent decades, with the relevant reports about oxidative stress and inflammatory response in IDD increasing gradually, the mechanisms researches have attracted much more attention. Consequently, this study focused on the indispensable roles of the oxidative stress and inflammatory response (especially macrophages and cytokines) to illustrate the origin, development, and deterioration of IDD, aiming to provide novel insights in the molecular mechanisms as well as significant clinical values for IDD.
Article
Full-text available
Exosomes are membranous lipid vesicles fused with intracellular multicellular bodies and then released into the extracellular environment. They contain various bioactive substances, including proteins, mRNA, miRNAs, lncRNAs, circRNAs, lipids, transcription factors, and cytokine receptors. Under certain conditions, bone marrow mesenchymal stem cells (BMSCs) can differentiate into osteoblasts, chondrocytes, adipocytes, and biological functions. This study provides a theoretical basis for the application of exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos) in osteology, exploring different sources of exosomes to improve bone microenvironment and resist bone metastasis. We also provided new ideas for the prevention and rehabilitation of human diseases by exosomes.
Article
Full-text available
Objective: Articular cartilage injury is common and difficult to treat clinically because of the characteristics of the cartilage. Bone marrow-derived mesenchymal stem cell (BMSC)-mediated cartilage regeneration is a promising therapy for treating articular cartilage injury. BMSC differentiation is controlled by numerous molecules and signaling pathways in the microenvironment at both the transcriptional and post-transcriptional levels. However, the possible function of super enhancer long non-coding RNAs (SE-lncRNAs) in the chondrogenic differentiation of BMSCs is still unclear. Our intention was to explore the expression profile of SE-lncRNAs and potential target genes regulated by SE-lncRNAs during chondrogenic differentiation in BMSCs. Materials and Methods: In this study, we conducted a human Super-Enhancer LncRNA Microarray to investigate the differential expression profile of SE-lncRNAs and mRNAs during chondrogenic differentiation of BMSCs. Subsequent bioinformatic analysis was performed to clarify the important signaling pathways, SE-lncRNAs, and mRNAs associated with SE-lncRNAs regulating the chondrogenic differentiation of BMSCs. Results: A total of 77 SE-lncRNAs were identified, of which 47 were upregulated and 30 were downregulated during chondrogenic differentiation. A total of 308 mRNAs were identified, of which 245 were upregulated and 63 were downregulated. Some pathways, such as focal adhesion, extracellular matrix (ECM)–receptor interaction, transforming growth factor-β (TGF-β) signaling pathway, and PI3K–Akt signaling pathway, were identified as the key pathways that may be implicated in the chondrogenic differentiation of BMSCs. Moreover, five potentially core regulatory mRNAs (PMEPA1, ENC1, TES, CDK6, and ADIRF) and 37 SE-lncRNAs in chondrogenic differentiation were identified by bioinformatic analysis. Conclusion: We assessed the differential expression levels of SE-lncRNAs and mRNAs, along with the chondrogenic differentiation of BMSCs. By analyzing the interactions and co-expression, we identified the core SE-lncRNAs and mRNAs acting as regulators of the chondrogenic differentiation potential of BMSCs. Our study also provided novel insights into the mechanism of BMSC chondrogenic and cartilage regeneration.
Article
Full-text available
Exosome therapy is a promising therapeutic approach for intervertebral disc degeneration (IVDD) and achieves its therapeutic effects by regulating metabolic disorders, the microenvironment and cell homeostasis with the sustained release of microRNAs, proteins, and transcription factors. However, the rapid clearance and disruption of exosomes are the two major challenges for the application of exosome therapy in IVDD. Herein, a thermosensitive acellular extracellular matrix (ECM) hydrogel coupled with adipose-derived mesenchymal stem cell (ADSC) exosomes (dECM@exo) that inherits the superior properties of nucleus pulposus tissue and ADSCs was fabricated to ameliorate IVDD. This thermosensitive dECM@exo hydrogel system can provide not only in situ gelation to replenish ECM leakage in nucleus pulposus cells (NPCs) but also an environment for the growth of NPCs. In addition, sustained release of ADSC-derived exosomes from this system regulates matrix synthesis and degradation by regulating matrix metalloproteinases (MMPs) and inhibits pyroptosis by mitigating the inflammatory response in vitro. Animal results demonstrated that the dECM@exo hydrogel system maintained early IVD microenvironment homeostasis and ameliorated IVDD. This functional system can serve as a powerful platform for IVD drug delivery and biotherapy and an alternative therapy for IVDD.
Article
Full-text available
Intervertebral disc degeneration (IVDD) occurs as a result of an imbalance of the anabolic and catabolic processes in the intervertebral disc, leading to an alteration in the composition of the extracellular matrix (ECM), loss of nucleus pulposus (NP) cells, excessive oxidative stress and inflammation. Degeneration of the IVD occurs naturally with age, but mechanical trauma, lifestyle factors and certain genetic abnormalities can increase the likelihood of symptomatic disease progression. IVDD, often referred to as degenerative disc disease (DDD), poses an increasingly substantial financial burden due to the aging population and increasing incidence of obesity in the United States. Current treatments for IVDD include pharmacological and surgical interventions, but these lack the ability to stop the progression of disease and restore the functionality of the IVD. Biological therapies have been evaluated but show varying degrees of efficacy in reversing disc degeneration long-term. Stem cell-based therapies have shown promising results in the regeneration of the IVD, but face both biological and ethical limitations. Exosomes play an important role in intercellular communication, and stem cell-derived exosomes have been shown to maintain the therapeutic benefit of their origin cells without the associated risks. This review highlights the current state of research on the use of stem-cell derived exosomes in the treatment of IVDD.
Article
Full-text available
Objective Studies have shown that the therapeutic effects of mesenchymal stem cells (MSCs) are mediated in a paracrine manner, mainly through extracellular vesicles such as exosomes. Here, we designed a study to investigate whether exosomes derived from adipose-derived mesenchymal stem cells (ADMSC-Exos) had protective effects in a rat model of radiation-induced brain injury and in microglia. Methods Male adult Sprague-Dawley (SD) rats were randomly divided into three groups: the control group, the radiation group (30 Gy), and the radiation + exosomes group (30 Gy + 100 ug exosomes). Meanwhile, microglia were divided into four groups: the control group, the radiation group (10 Gy), the radiation + exosomes group (10 Gy + 4 ug exosomes), and radiation + exosomes + EX527 group (10 Gy + 4 ug exosomes + 100 nM EX527). Tissue samples and the levels of oxidative stress and inflammatory factors in each group were compared. Results Statistical analysis showed that after irradiation, ADMSC-Exos intervention in vivo significantly reduced the levels of caspase-3, malondialdehyde (MDA), 8-hydroxydeoxyguanosine (8-OHdG), tumor necrosis factor-α (TNF-α), interleukin-4 (IL-4), and promoted the recovery of superoxide dismutase (SOD), catalase (CAT), IL-4, and IL-10. Moreover, ADMSC-Exos intervention inhibited microglial infiltration and promoted the expression of SIRT1. Furthermore, the results in vitro showed that the above effects of ADMSC-Exos could be reversed by SIRT-1 inhibitor EX527. Conclusion This study demonstrated that ADMSC-Exos exerted protective effects against radiation-induced brain injury by reducing oxidative stress, inflammation and microglial infiltration via activating the SIRT1 pathway. ADMSC-Exos may serve as a promising therapeutic tool for radiation-induced brain injury.
Article
Full-text available
Intervertebral disk degeneration (IVDD) is a leading cause of disability. The degeneration is inevitable, and the mechanisms are complex. Current therapeutic strategies mainly focus on the relief of symptoms, not the intrinsic regeneration of the intervertebral disk (IVD). Tissue engineering is a promising strategy for IVDD due to its ability to restore a healthy microenvironment and promote IVD regeneration. This review briefly summarizes the IVD anatomy and composition and then sets out elements of the microenvironment and the interactions. We rationalized different scaffolds based on tissue engineering strategies used recently. To fulfill the complete restoration of a healthy IVD microenvironment, we propose that various tissue engineering strategies should be combined and customized to create personalized therapeutic strategies for each individual.
Article
Full-text available
Background The use of adipose-derived mesenchymal stromal cell-derived exosomes (ADSC-Exos) may become a new therapeutic method in biomedicine owing to their important role in regenerative medicine. However, the role of ADSC-Exos in tendon repair has not yet been evaluated. Therefore, we aimed to clarify the healing effects of ADSC-Exos on tendon injury. Methods The adipose-derived mesenchymal stromal cells (ADSCs) and tendon stem cells (TSCs) were isolated from the subcutaneous fat and tendon tissues of Sprague-Dawley rats, respectively, and exosomes were isolated from ADSCs. The proliferation and migration of TSCs induced by ADSC-Exos were analyzed by EdU, cell scratch, and transwell assays. We used western blot to analyze the tenogenic differentiation of TSCs and the role of the SMAD signaling pathways. Then, we explored a new treatment method for tendon injury, combining exosome therapy with local targeting using a biohydrogel. Immunofluorescence and immunohistochemistry were used to detect the expression of inflammatory and tenogenic differentiation after tendon injury, respectively. The quality of tendon healing was evaluated by hematoxylin-eosin (H&E) staining and biomechanical testing. Results ADSC-Exos could be absorbed by TSCs and promoted the proliferation, migration, and tenogenic differentiation of these cells. This effect may have depended on the activation of the SMAD2/3 and SMAD1/5/9 pathways. Furthermore, ADSC-Exos inhibited the early inflammatory reaction and promoted tendon healing in vivo. Conclusions Overall, we demonstrated that ADSC-Exos contributed to tendon regeneration and provided proof of concept of a new approach for treating tendon injuries.
Article
Full-text available
Objective. Low back pain (LBP) is one of the top three causes of disability in developed countries, and intervertebral disc degeneration (IDD) is a major contributor to LBP. In the process of IDD, there is a gradual decrease in nucleus pulposus cells (NPCs) and extracellular matrix (ECM). Exosomes are important exocrine mediators of stem cells that can act directly on cells for tissue repair and regeneration. In this study, we determined the antisenescence, cell proliferation promotion, and ECM modulation effects of human urine-derived stem cell (USC) exosomes (USC-exos) on degenerated intervertebral discs and explored the underlying mechanism. Methods and Materials. USCs were identified by multipotent differentiation and flow cytometry for mesenchymal stem cell- (MSC-) specific surface protein markers. USC-exos were isolated from the conditioned medium of USCs by ultracentrifugation and then analyzed by transmission electron microscopy (TEM), particle size analysis, and western blotting (WB) for exosome marker proteins. The effects of USC-exos on NPC proliferation and ECM synthesis were assessed by Cell Counting Kit-8 (CCK-8), WB, and immunofluorescence (IF) analyses. The protein differences between normal and degenerative intervertebral discs were mined, and the temporal and spatial variations in matrilin-3 (MATN3) content were determined by WB and IF in the intervertebral disc tissues. The candidate molecules that mediated the function of USC-exos were screened out and confirmed by multiple assays. Meanwhile, the mechanism underlying the candidate protein in USC-exos-induced cell proliferation and regulation of ECM synthesis promoting the activities of NPCs was explored. In addition, the effects of USC-exos on ameliorating intervertebral disc degeneration (IVD) in mice were examined by assessing computed tomography (CT), magnetic resonance imaging (MRI), and histological analyses. Results. The flow cytometry results showed that USCs were positive for CD29, CD44, and CD73, which are USC surface-specific markers, but negative for CD34 and CD45. In addition, USCs showed osteogenic, adipogenic, and chondrogenic differentiation potential. USC-exos exhibited a cup-shaped morphology, with a mean diameter of , and were positive for CD63 and TSG101 and negative for calnexin. USC-exos could promote NPC proliferation and ECM synthesis. The protein content of the matrilin family was significantly reduced in degenerative intervertebral discs, and the decrease in MATN3 was the most significant. USC-exos were found to be rich in MATN3 protein, and exosomal MATN3 was required for USC-exos-induced promotion of NPC proliferation and ECM synthesis, as well as alleviation of intervertebral disc degeneration in IVD rats. In addition, the effects of MATN3 in USC-exos were demonstrated to be achieved by activating TGF-β, which elevated the phosphorylation level of SMAD and AKT. Conclusions. Our study suggests that reduced MATN3 can be considered a characteristic of intervertebral disc degeneration. USC-exos may represent a potentially effective agent for alleviating intervertebral disc degeneration by promoting NPC proliferation and ECM synthesis by transferring the MATN3 protein. 1. Introduction Low back pain (LBP) is a very common problem experienced by most people at a certain time in their life, and it is among the top three causes of disability in developed countries [1–3]. The definite causes of low back pain remain unclear; however, intervertebral disc degeneration (IDD) has been documented to be a major contributor to LBP and is the pathological basis for spinal instability, disc herniation, and other spinal degenerative diseases, which cause a considerable burden to society and families and thus are the major global public health issues [4, 5]. In disc degeneration, the main pathological change is a gradual reduction in the total NPCs and extracellular matrix (ECM). NPCs are the main functional cells responsible for ECM synthesis. The homeostatic imbalance between anabolism and catabolism leads to the loss of collagen and proteoglycan [6, 7]. Collagen type II (COL2) and proteoglycan (predominantly aggrecan (ACAN)) are crucial ECMs for discs to maintain proper function, particularly for the nucleus pulposus [8, 9]. ACAN is a biological macromolecule formed by one or more glycosaminoglycan (GAG) chains covalently connected to a core protein. It is the main noncollagen component of the intervertebral disc. The glycosaminoglycans contained in ACAN are mainly chondroitin sulfate and keratan sulfate. The rich and unique molecular characteristics of intervertebral discs allow these structures to penetrate and withstand pressure. One of the reasons for the damage and degeneration of the intervertebral disc is the degradation and loss of ACAN. COL2 is one of the most important collagen components in intervertebral discs. COL2 is the main collagen in cartilage, accounting for more than 50% of the extracellular matrix of cartilage. COL2 is mainly expressed by chondrocytes and is abundantly present in the nucleus pulposus [10, 11]. In ECM, COL2 and ACAN are the two most representative components; therefore, this experiment measured the expression of COL2 and ACAN to illustrate ECM conditions. Determining the methods of rebalancing disordered COL2 and ACAN expression and increasing their synthesis is considered a key factor for slowing down or even reversing IVD damage. Recently, increasing evidence has revealed that mesenchymal stem cells (MSCs) can release exosomes, which are specialized extracellular vesicles that could provide therapeutic benefits [12, 13]. Exosomes are membranous vesicles with a diameter of 50-200 nm, and they contain multiple cellular components, such as proteins, nucleic acids, and lipids. Exosomes act as a cell-free mediator and transfer particular cytokines into recipient cells to achieve their therapeutic paracrine effects in inhibiting senescence, modulating metabolism, and promoting regeneration [14]. Thus, stem cell exosomes may have potential applications as effective cell-free therapeutic agents [15]. However, MSCs have a limited source and cause certain trauma to the body, which limits their application. Human urine-derived stem cells are stem cells with multidifferentiation potential obtained from human urine. These cells have a wide range of sources, are convenient to obtain, present safe and noninvasive characteristics, do not violate ethics, and represent a better choice for obtaining exosomes [16–18]. MATN3 is a member of the matrilin protein family, and it is mainly distributed in cartilage cells and plays an important role in the synthesis of cartilage ECM. Variations or decreases in its content can lead to cartilage and intervertebral disc degeneration. In previous studies, we found that USC-exos are rich in this protein. A number of studies have shown that USC transplantation is beneficial to degenerated intervertebral discs [12, 13]. Exosomes are exocrine vesicles of stem cells that play a paracrine role and deliver a variety of biological effectors of the parent cells. Exosomes that can be absorbed by recipient cells are involved in cellular communication, signaling pathway activation, and metabolism modulation and play an important role in MSC-based therapy. Exosomes are widely distributed and readily available and have no immunogenicity; therefore, they are ideal agents for the treatment of tissue repair and regeneration [19]. In the previous study [20], the team studied the effect of USC-exos on intervertebral disc degeneration by inhibiting the apoptosis of NPCs. We now further explored the promotion of cell proliferation and ECM modulatory effects of human urine-derived stem cell (USC) exosomes (USC-exos) on degenerated intervertebral discs in cell and rat models and explored the underlying mechanism. 2. Materials and Methods 2.1. Isolation and Culture of NPCs The experimental scheme was approved by the Ethics Committee of the Affiliated Hospital of Qingdao University (approval number: QDFY-19-012-03). Nucleus pulposus cells were obtained from 6 patients with lumbar disc herniation, and they had an average age of . The clinical symptoms and physical examinations were consistent with the surgical indications. Before surgery, three experienced chief physicians of spine surgery and one chief physician of the imaging department evaluated their MRI, and the modified Pfirrmann scale of the lumbar intervertebral disc was grade 5. This finding indicated that the nucleus pulposus and the medial annular fiber showed low signal and the intervertebral disc was highly normal. After obtaining informed consent from the patients and their relatives and signing a donation agreement for the study, disc tissue was obtained from patients undergoing posterior lumbar foraminal surgery. The fibrous annulus (AF) and cartilaginous endplate (CEP) were carefully removed from the specimens under a microscope. After rinsing with phosphate buffered saline (PBS) 3 times, the nucleus pulposus tissue was cut to approximately 1 mm³ in size and then digested in 0.2% type II collagenase (Gibco, Grand Island, NY, USA) for 3 h. After digestion, a 75 μm filter was used to remove tissue residue, and then, the cell suspension was centrifuged at 800 r/min for 5-10 min. Dulbecco’s modified Eagle medium/F12 (DMEM/F-12) containing 10% fetal bovine serum (FBS; Gibco, Grand Island, NY, USA) and 1% penicillin-streptomycin (Gibco, Grand Island, NY, USA) was used to resuspend the NPCs; subsequently, the cells were placed in a cell incubator at 37°C with 90% N2, 5% CO2, and 5% O2 [21]. 2.2. USCs Extraction The fresh sterile urine of 6 healthy male adults (mean age, ) was collected under aseptic conditions. The obtained sample was centrifuged at 400 g for 10 min, the supernatant was discarded, and the cell precipitate was resuspended in phosphate buffer (PBS). The supernatant was carefully removed after centrifugation at 200 g for 10 min. The cell precipitates were resuspended in 4 ml DMEM/F-12 medium (HyClone, Utah, USA) composed of 10% FBS (Gibco, Australia), 1% penicillin-streptomycin, and RegM Singlequot growth factor additive (Lonza, Basel, Switzerland). The cells were then inoculated in 12-well plates precoated with gelatin and placed in an incubator at 37°C with 5% CO2. The medium was changed every two days until the cell clone was formed and then replaced with RE/MC medium for further culture. The RE cell proliferation medium was 500 ml RE cell base medium supplemented with the RegM Single kit. The MC propagation medium includes 10% FBS and 1% Glutamax added to DMEM/F-12 medium (Gibco, Japan), 1% NEAA (Gibco, Grand Island, USA), 1% Pen/Strep (Gibco, Grand Island, USA), 5 ng/ml BFGF (Peprotech, Rocky Hill, USA), 5 ng/ml PDGF-Ab (Peprotech, Rocky Hill, USA), and 5 ng/ml EGF (Peprotech, Rocky Hill, USA). Re/MC medium is a 1 : 1 mixture of Re and MC medium. When the cell density reached approximately 70%-80%, passaging was carried out, and then, P2-4 generation cells were selected for subsequent experiments. 2.3. Flow Cytometric Analysis of Surface Markers of USCs After trypsin digestion, P3 USCs in a good growth state were collected and then washed with PBS 3 times after centrifugation. A cell suspension with a final concentration of was prepared. Then, 100 μl of cell suspension was added to 10 μl of monoclonal antibody working solution for CD29, CD34, CD44, CD45, and CD73 (Santa Cruz Biotechnology, USA) and incubated for 1 hour at room temperature in the dark. The cells were washed for another 3 times and analyzed by flow cytometry. 2.4. Determination of the Multidirectional Differentiation Potential of USCs To evaluate the differentiation potential of human urine-derived stem cells, osteogenic, adipogenic, and chondrogenic differentiation was performed according to the associated kit instructions. USCs were inoculated into 6-well plates, and differentiation was induced when the cell fusion rate reached approximately 80%. Osteogenic induction differentiation medium (CYAGEN, China) was added and replaced every 3 days. After 21 days of induction, the USCs were fixed with 4% paraformaldehyde and stained with Alizarin Red for observation. The kit contained 175 ml basal medium, 20 ml serum, 2 ml penicillin-streptomycin, 2 ml glutamine, 2 ml β-sodium glycerophosphate, 400 μl ascorbic acid, and 20 μl dexamethasone. For adipogenic induction differentiation, adipogenic induction differentiation medium A (Cyagen, China) was first added and then replaced with adipogenic induction differentiation medium B (Cyagen, China) 3 days later, and after 24 h, it was replaced with liquid A again, with this process alternated 3 times. Finally, 4% paraformaldehyde was used for fixation, and oil red O staining was used for observation. Adipogenesis induction differentiation medium A contained 175 ml basal medium, 20 ml fetal bovine serum, 2 ml penicillin-streptomycin, 2 ml glutamine, 400 μl insulin, 200 μl 3-isobutyl-1-methyl xanthine (IBMX), 200 μl dexamethasone, and 200 μl rosiglitazone. The adipogenic induction differentiation medium B contained 175 ml basal medium, 20 ml fetal bovine serum, 2 ml penicillin-streptomycin, 2 ml glutamine, and 400 μl insulin. During chondroblast induction, the cells were first counted, and then USCs were collected. The supernatant was discarded after centrifugation in a 15 ml centrifuge tube at 150 g for 5 min, and then, 0.5 ml chondroblast induction medium was added and changed every 3 days. Approximately 21 days later, 4% paraformaldehyde was used for fixation, paraffin embedding was followed by sectioning, and allicin blue staining was used for observation. The chondroblast induction differentiation medium kit contained 194 ml basal medium, 600 μl ascorbic acid, 20 μl dexamethasone, 2 ml ITS+Supplement, 200 μl sodium pyruvate, 200 μl proline, and 2 ml factor-β3 (TGF-β3). 2.5. Exosome Extraction and Identification When the cells grew to 70-75% confluence, the culture medium was removed and washed 3 times with PBS. Serum-free medium was added, and the culture was continued for 48 h. The culture medium was then collected, centrifuged at 4°C and 500 g for 10 min to remove residual cells, and centrifuged at 4°C and 2000 g for 20 min to remove cell debris. The impurities were further removed at 4°C and 10,000 g for 30 min, and the supernatant was retained and then filtered with a 0.22 μm filter membrane to remove excess particles. The supernatant was centrifuged at 4°C and 100,000 g for 2 h, and the resulting precipitate was resuspended in PBS. The exosome morphology was observed by transmission electron microscopy (TEM) (JEM-1200EX, Japan). The number and size distribution of exosomes were analyzed using a NanoSight detector (Malvern, England) and NTA detection and analysis software. USCs were cleaved to obtain their cleavage products, which were used as a negative control to conduct WB with USC-exos. Western blotting was used to detect the exosome markers CD63, TSG101, and calnexin. 2.6. Exosome Uptake of NPCs P3 NPCs with good growth status were inoculated into 24-well plates for subsequent experiments after the cells adhered to the wall. First, GFP-lentivirus was transfected to observe the cell profile of NPCs. GFP virus and Lipofectamine 2000 (Thermo Fisher, Massachusetts, USA) were diluted with equal amounts of serum-free culture medium. The diluted GFP was mixed with Lipofectamine 2000 and kept at room temperature for 20 min. The mixture was added to the cell culture medium and transfected for approximately 3 h. Then, the exosomes were labeled with PKH26 (Sigma-Aldrich) fluorescent dye according to the operation instructions of the PKH26 fluorescent dye kit. The excess dye was neutralized with an equal volume of PBS containing 5% BSA. Finally, the supernatant was removed by centrifugation at 4°C for 70 min at 100,000 g and resuspended in 50 μl PBS. The prepared USC-exos labeled with PKH26 were added to GFP-transfected NPCs and incubated in the dark for 12 h. After fixation with 4% paraformaldehyde for 20 min, the nuclei were stained with DAPI. The glycerin was sealed, and uptake was observed by laser confocal microscopy. The Leica Application Suite Advanced Fluorescence software was used to analyze the images in the later stage. 2.7. CCK-8 (Cell Counting Kit-8) Detects NPC Proliferation NPCs were prepared into cell suspensions and counted and then seeded into 96-well plates, with cells in each well and each group set up with 3 duplicate wells. PBS was added to the control group, and USC-exos, USCshMATN3-exos, and USCconshRNA-exos were added to the other groups. The cell-free wells were used as blank controls. After intervention, 10 μl CCK-8 solution (Meilunbio, Dalian, China) was added on days 1, 3, 5, and 7 and then cultured in a cell incubator for approximately 3 h. A microplate reader (Molecular Devices, USA) was used to detect the absorbance at 450 nm, and then, the proliferation of NPCs was calculated based on the change in absorbance. 2.8. β-Galactosidase Staining to Detect Cell Senescence NPCs with good P3 growth were inoculated into a 6-well plate and treated according to the experimental groups after the cells adhered to the wall. After the intervention, the instructions of the β-galactosidase staining kit (Beyotime, China) were followed for cell senescence detection. The specific steps were as follows. Staining fixative solution was added to fix the cells at room temperature for 15 minutes. After washing 3 times with PBS, the following was added to each well: 1 ml of 930 μl β-galactosidase staining solution C, 10 μl β-galactosidase staining solution A, 10 μl β-galactosidase staining β-galactosidase staining working solution prepared from solution B, and 50 μl X-Gal solution. The cells were incubated overnight at 37°C, and the senescence of NPCs was observed under an inverted phase contrast microscope. 2.9. Western Blot (WB) Analysis After the intervention, the samples were collected and then lysed in RIPA lysis buffer (Solarbio, Beijing, China) containing 1 mM phenyl methylsulfonyl fluoride (PMSF) and protease inhibitors to extract proteins. The extracted protein was tested to determine its concentration using a BCA kit (Solarbio, Beijing, China). Then, the protein and loading buffer were mixed at a ratio of 4 : 1 () and boiled for 10 minutes. The proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to a polyvinylidene fluoride (PVDF) membrane. The PVDF membrane was blocked with 5% skimmed milk powder at room temperature and then incubated overnight at 4°C with primary antibodies against CD63, TSG101, calnexin, TGF-β3, p-SMAD3, SMAD3, AKT, p-AKT, β-actin (all the above antibodies were purchased from Santa Cruz Biotechnology, USA), MATN3 (Bioss, Beijing, China), COL2 (Bioss, Beijing, China), and ACAN (Millipore, Massachusetts, USA). The membrane was incubated with a horseradish peroxidase- (HRP-) labeled secondary antibody (ABclonal, Wuhan, China) for 1 hour, and then, an ECL kit (Thermo Fisher Scientific, Rockville, MD, USA) was used for luminescence observation. The Image Lab software (Bio-Rad, Hercules, CA, USA) was used to take images and analyze them. 2.10. Transfection of MATN3 Lentivirus-shRNA In the functional mechanism investigation, lentiviral shRNAs targeting MATN3 (shMATN3, sc106205V) (Santa Cruz Biotechnology, USA) and control shRNAs (Con shRNAs) were transfected before USC-exos treatment. In the feedback mechanism investigation, the overexpressed lentivirus of MATN3 (LV-MATN3) and the control lentiviral vectors (con-LV) were transfected. The lentivirus vectors were packaged by GeneChem (Shanghai, China). Transfection was conducted according to the manufacturer’s instructions. Briefly, NPCs were plated into dishes 1 day before transfection. The next day, the NPCs were transfected with the lentivirus vectors at an MOI of 100 supplemented with 10 μg/ml polybrene (Cyagen) for 24 h. The culture medium was replaced with fresh complete medium, and the cells were selected with 2.5 μg/ml puromycin (Sigma) 72 h after transfection. Total RNA was harvested and subjected to qPCR analysis for efficiency. Forty-eight hours after transfection, the total RNA was harvested and subjected to qPCR analysis. Seventy-two hours after transfection, the total proteins were harvested and subjected to western blot analysis. 2.11. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) Total RNA was extracted from the samples using TRIzol reagent (Invitrogen, Carlsbad, CA, USA). Then, the instructions of the reverse transcription kit for qRT-PCR were followed to reverse transcription and amplify related genes. GAPDH was used as an internal reference, and each sample was set with three auxiliary holes. The primer sequences are shown in Table 1. The data obtained were analyzed using the 2-ΔΔCt algorithm. Gene name Primer sequences (5-3) MATN3 Forward (F) 5-GGTGCAGGTGTTTGCAAGAG-3 Reverse (R) 5-TCCACTGTGAAGGCTTCGTC-3 GAPDH Forward (F) 5-GGTATCGTGGAAGGACTC-3 Reverse (R) 5-GTAGAGGCAGGGATGATG-3
Article
Full-text available
Mesenchymal stromal cells (MSC) are used for cell therapy for spinal cord injury (SCI) because of their ability to support tissue repair by paracrine signaling. Preclinical and clinical research testing MSC transplants for SCI have revealed limited success, which warrants the exploration of strategies to improve their therapeutic efficacy. MSC are sensitive to the microenvironment and their secretome can be altered in vitro by exposure to different culture media. Priming MSC with inflammatory stimuli increases the expression and secretion of reparative molecules. We studied the effect of macrophage-derived inflammation priming on MSC transplants and of primed MSC (pMSC) acute transplants (3 days) on spinal cord repair using an adult rat model of moderate–severe contusive SCI. We found a decrease in long-term survival of pMSC transplants compared with unprimed MSC transplants. With a pMSC transplant, we found significantly more anti-inflammatory macrophages in the contusion at 4 weeks post transplantation (wpt). Blood vessel presence and maturation in the contusion at 1 wpt was similar in rats that received pMSC or untreated MSC. Nervous tissue sparing and functional recovery were similar across groups. Our results indicate that macrophage-derived inflammation priming does not increase the overall therapeutic potential of an MSC transplant in the adult rat contused spinal cord.
Article
Intervertebral disc degeneration (IDD) is a chronic skeletal muscle degenerative disease, which is considered the main cause of low back pain. It seriously affects the quality of life of patients and consequently brings a heavy economic burden to their families and the society. Although IDD is considered a natural process in degenerative lesions, it is mainly caused by aging, trauma, genetic susceptibility and other factors. It is closely related to changes in the tissue structure and function, including the progressive destruction of extracellular matrix, cell aging, cell death of the intervertebral disc (IVD), inflammation, and impairment of tissue biomechanical function. Currently, the treatment of IDD is aimed at alleviating symptoms rather than at targeting pathological changes in the IVD. Furthermore, the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway is closely related to various pathological processes in IDD, and the activation of the MAPK/ERK pathway promotes the degradation of the IVD extracellular matrix, cell aging, apoptosis, and inflammatory responses. It also induces autophagy and oxidative stress that accelerate the IVD process. In our current review, we summarize the latest developments in the negative regulation of IDD after activation of the MAPK/ERK signaling pathway and emphasize on its influence on IDD. Targeting this pathway may become an attractive treatment strategy for IDD in the near future.