ArticlePDF Available

Injectable dECM-enhanced hyaluronic microgels with spatiotemporal release of cartilage-specific molecules to improve osteoarthritic chondrocyte's function

Authors:

Abstract and Figures

The interior environment of articular cartilage in osteoarthritis (OA) presents substantial hurdles, leading to the malfunction of chondrocytes and the breakdown of collagen II-enriched hyaline cartilage matrix. Despite this, most clinical treatments primarily provide temporary relief from OA discomfort without arresting OA progression. This study aimed to alleviate OA by developing intra-articular injectable dECM-enhanced hyaluronic (HE) microgels. The HE hydrogel was engineered and shaped into uniformly sized microgels using microfluidics and photopolymerization techniques. These microgels provided a spatiotemporal cascade effect, facilitating the rapid release of growth factors and a slower release of ECM macromolecules and proteins. This process assisted in the recovery of OA chondrocytes' function, promoting cell proliferation, matrix synthesis, and cartilage-specific gene expression in vitro. It also effectively aided repair of the collagen II-enriched hyaline cartilage and significantly reduced the severity of OA, as demonstrated by radiological observation, gross appearance, histological/immunohistochemical staining, and analysis in an OA rat model in vivo. Collectively, the HE injectable microgels with spatiotemporal release of cartilage-specific molecules have shown promise as a potential candidate for a cell-free OA therapy approach.
Content may be subject to copyright.
Dengetal. Collagen and Leather (2024) 6:14
https://doi.org/10.1186/s42825-024-00158-6
RESEARCH Open Access
© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http://creativecommons.org/licenses/by/4.0/.
Collagen and Leather
Injectable dECM-enhanced hyaluronic
microgels withspatiotemporal release
ofcartilage-specic molecules toimprove
osteoarthritic chondrocyte’s function
Siyan Deng1,2, Hongfu Cao1,2, Yan Lu1,2, Wenqing Shi1,2, Manyu Chen1,2, Xiaolin Cui3,4, Jie Liang1,2,5,
Yujiang Fan1,2*, Qiguang Wang1,2* and Xingdong Zhang1,2
Abstract
The interior environment of articular cartilage in osteoarthritis (OA) presents substantial hurdles, leading to the mal-
function of chondrocytes and the breakdown of collagen II-enriched hyaline cartilage matrix. Despite this, most
clinical treatments primarily provide temporary relief from OA discomfort without arresting OA progression. This study
aimed to alleviate OA by developing intra-articular injectable dECM-enhanced hyaluronic (HE) microgels. The HE
hydrogel was engineered and shaped into uniformly sized microgels using microfluidics and photopolymerization
techniques. These microgels provided a spatiotemporal cascade effect, facilitating the rapid release of growth factors
and a slower release of ECM macromolecules and proteins. This process assisted in the recovery of OA chondrocytes’
function, promoting cell proliferation, matrix synthesis, and cartilage-specific gene expression in vitro. It also effec-
tively aided repair of the collagen II-enriched hyaline cartilage and significantly reduced the severity of OA, as dem-
onstrated by radiological observation, gross appearance, histological/immunohistochemical staining, and analysis
in an OA rat model in vivo. Collectively, the HE injectable microgels with spatiotemporal release of cartilage-specific
molecules have shown promise as a potential candidate for a cell-free OA therapy approach.
Keywords Osteoarthritis, Decellularized cartilage matrix, Hyaluronic acid, Spatiotemporal release, Intra-articular
injection
*Correspondence:
Yujiang Fan
fan_yujiang@scu.edu.cn
Qiguang Wang
wqgwang@126.com
Full list of author information is available at the end of the article
Page 2 of 17
Dengetal. Collagen and Leather (2024) 6:14
Graphical Abstract
1 Introduction
Osteoarthritis (OA) is a prevalent chronic disease that
afflicts hundreds of millions of people worldwide each
year [1]. It is characterized by pain, an inflammatory
response, joint destabilization, and the loss of colla-
gen II-enriched hyaline cartilage, ultimately affecting
the entire joint. Early treatment of OA often involves
the use of pharmacological interventions such as non-
steroidal anti-inflammatory drugs (NSAIDs), corticos-
teroids, and analgesics to alleviate pain [2, 3]. However,
these treatments frequently fail to halt further deterio-
ration of the joint structure and microenvironment and
are associated with adverse effects [4, 5]. Once the dis-
ease advances to a severe stage, surgical joint replace-
ment becomes the only viable treatment option. us,
it is crucial to develop effective intervention programs
that target the early stages of OA.
Intra-articular hyaluronic acid (HA) injections have
been extensively used in OA treatment, providing
effective pain relief and symptomatic therapy [6]. HA, a
glycosaminoglycan found abundantly in synovial fluid,
plays a crucial role in protecting articular cartilage and
promoting cartilage regeneration due to its inherent
properties, particularly viscoelasticity [7]. High molec-
ular weight HA, which is often reduced in OA joints,
maintains the rheological homeostasis of the synovial
fluid and helps dampen shock, lubricate, and nourish
joint tissues [8, 9]. erefore, intra-articular HA injec-
tions have been shown to function as viscosupplemen-
tation, leading to significant functional improvement
in OA joints. However, most HA preparations have a
short half-life in the joints due to rapid clearance and
swift degradation in vivo by enzymatic or hydrolytic
reactions, remaining in the joints for only a few hours
to a couple of days [6, 10, 11]. Consequently, achiev-
ing effective treatment results often requires multiple
injections, which can lead to difficulties in ensuring
patient compliance and an increased risk of side effects.
Page 3 of 17
Dengetal. Collagen and Leather (2024) 6:14
erefore, it is essential to optimize biomaterials for
intra-articular injections to aid in OA treatment.
Injectable hydrogels have been widely investigated for
use in OA arthrocentesis due to their slow degradation,
enhanced stability, and strong adhesion, which allows
for prolonged retention [12, 13]. However, the irregular
shape and large size of hydrogels can lead to high and
uneven injection forces during injection [14], potentially
damaging healthy tissues in the affected area. Compared
to block hydrogels, hydrogel microspheres, also known
as microgels, with their uniform size and spherical shape,
provide improved injectability [15, 16]. Among the vari-
ous production methods, microfluidics offers significant
advantages such as the rapid generation of microgels
with precise and consistent sizes [17]. Consequently,
HA-based microgels have been extensively studied and
have shown to effectively extend the degradation of HA,
provide lubrication, and offer biocompatibility. How-
ever, within the complex intra-articular joint environ-
ment, a single HA component has limited capacity to
replace the cartilaginous extracellular matrix and offer
multifunctionality.
e extracellular matrix (ECM) serves as a natural
structure for cell adhesion, proliferation, migration, and
differentiation, in addition to providing biomechanical
capabilities. e decellularized ECM (dECM), derived
by eliminating cellular components and genetic material
from the ECM of human or animal tissues using decel-
lularization techniques, reduces immunogenicity and is
a promising biomaterial highly regarded in regenerative
tissue engineering applications, such as skin, cornea,
and nerve [18, 19]. Cartilage tissue-derived dECM has
been demonstrated to provide structural and functional
proteins that support cell attachment, growth, and dif-
ferentiation specific to cartilage tissue [20]. e dECM
retains most cartilage-specific matrix macromolecules,
including primarily collagen II and proteoglycans, along
with less abundant components such as fibulin and elas-
tin. ese contribute to the production and maintenance
of ECM, enhance chondrocyte morphology and pheno-
type, ensure cartilage homeostasis, and regulate newborn
cartilage tissues through various cytokines, enzymes,
and signaling pathways [2124]. Moreover, it contains
numerous growth factors involved in chondrocyte func-
tion, including transforming growth factor-β (TGF-β),
insulin-like growth factor-1 (IGF-1), bone morphoge-
netic protein (BMP), fibroblast growth factor (FGF), and
epidermal growth factor (EGF) [25]. Members of the
TGF-β superfamily play a pivotal role in cartilage devel-
opment and repair [26, 27]; IGF-1 stimulates the anabolic
activity of chondrocytes and inhibits chondrocyte apop-
tosis [28, 29]; BMPs, particularly BMP-2 and BMP-7, pro-
mote chondrogenesis in MSCs and enhance chondrocyte
matrix production [30, 31]. In addition to growth factors,
erefore, cartilage-derived dECM, due to its similarity
to the natural microenvironment and the bioactivity of
cartilage formation, exhibits beneficial effects on tissue
morphogenesis, differentiation, homeostasis, and repair.
is study aimed to enhance the HA-based hydrogel
formulation, referred to HE, by incorporating a solution
derived from cartilage dECM to augment the limited
bio-functionality of HA and improve the injectability of
dECM. e HE microgels were fabricated using micro-
fluidics and photopolymerization, ensuring their uniform
size and mechanical strength for effective lubrication
and durability under extensive joint movements. It was
hypothesized that the HE microgels facilitate a spati-
otemporal cascade effect, with cartilage-specific growth
factors being released in the short term to prompt the
swift restoration of OA chondrocyte function. Concur-
rently, the extended release of structural ECM macromol-
ecules and collagens offers a sustained supply of essential
components, which are vital for cartilage regeneration
and the maintenance of homeostasis as the HE microgels
degrade. is combined process was anticipated to pro-
duce synergistic and enduring effects across the various
stages of OA chondrocyte recovery and cartilage regener-
ation, including improved viability of OA chondrocytes,
enhanced matrix secretion, collagen II synthesis, and
the preservation of cellular function and cartilage matrix
integrity, potentially offering a therapeutic approach for
OA. To validate this hypothesis, the effects of HE micro-
gels on OA chondrocyte proliferation, morphology, and
matrix secretion were investigated invitro. e findings
were further corroborated through intra-articular injec-
tion in a rat OA model invivo (Scheme1).
2 Materials andmethods
2.1 Materials
Hyaluronic acid (HA, Mw = 0.94 MDa) was purchased
from Florida Biochemistry Limited (Shandong, China).
Methacrylic anhydride (MA) was obtained from Chengdu
Chemical Xia Reagent Co. Liquid paraffin and Span
80 were purchased from Cologne Chemical (Chengdu,
China). 3-[(3-cholamidopropyl) dimethylamino] pro-
panesulfonic acid internal salt (CHAPS) were bought
from Sigma-Aldrich (USA). Hyaluronidase, type I col-
lagenase and DNase I were obtained from Solarbio
(Beijing, China). α-MEM medium was bought from Ser-
vicebio (Wuhan, China). Penicillin–streptomycin and
parenzyme were purchased from Hyclone (USA). Fetal
bovine serum (FBS, Gibco, KEEL) was obtained from
Life Technologies Corporation (USA). Fluorescein diac-
etate (FDA) and propidium iodide (PI) were bought from
Sigma-Aldrich (USA).
Page 4 of 17
Dengetal. Collagen and Leather (2024) 6:14
2.2 Hyaluronic acid methacryloyl (HAMA) synthesis
HA powder (2.5g) was dissolved in ultrapure water and
stirred to dissolve. Subsequently, the pH value of the
solution was adjusted to 8.5 with 1M NaOH under ice
bath conditions. en, MA (5.6mL) was added dropwise
controlling the pH value at 7.5–8.5 for a duration of 4h
in an ice bath. e reaction system was stirred at room
temperature overnight. After that, the solution was trans-
ferred to dialysis bags (Mw = 8000–14000kDa) and dia-
lyzed in ultrapure water for 3days at room temperature
to remove unreacted methacrylate groups. e dialysis
solution was frozen and lyophilized to obtain HAMA.
e chemical structure of HAMA was confirmed by H1
NMR (400MHz, Bruker AMX-400, USA) with D2O as
the solvent.
2.3 Preparation andcharacterization ofdECM
Fresh porcine knee joints were collected, and the joint
cavity was exposed to extract transparent cartilage pieces
while avoiding calcified tissue. After ultrasonic clean-
ing in phosphate-buffered saline (PBS, Sigma Aldrich,
USA), the cartilage pieces were lyophilized. Subse-
quently, the cartilage pieces were ground into particles
using a cryogenic grinder at -20 , 60 Hz and 3000 r/
min. e obtained pellets were mixed with PBS, filtered
through a 175μm sieve, and then decellularized as the
following steps reported previously [32, 33]. Firstly, the
cartilage particles were alternately placed in hypotonic
solution (10 mM Tris-HCI, pH = 8) and hypertonic
solution (50mM Tris-HCI, 1M NaCl, pH = 8), repe ated
three times and performing five freeze–thaw cycles
(liquid nitrogen and 37 °C water bath). After that, 1%
CHAPS solution treatment for 5 h was applied to the
cartilage ECM. en, the obtained ECM was treated
with nuclease solution (200 U/mL DNase I dissolved in
10mM Tris-HCI, pH = 7.5) for 9h at 37°C with agita-
tion, with a new solution replaced every 3h. e ECM
was next treated with 0.1% Triton-100 solution with agi-
tation and washed several times with distilled water to
remove residual detergent. Finally, the ECM powder was
obtained by centrifugation and lyophilized. e powder
was ground again through a 50 μm sieve and digested
with 0.1M acetic acid solution containing pepsin (pep-
sin: decellularized matrix = 1:10) for 2days. e digestion
was terminated by adjusting the pH to neutral to obtain
the dECM solution stored at 4 .
e typical components retained in the dECM were
characterized as follows: 10mg decellularized tissues was
placed in 1mL papain working solution and treated with
a water bath at 65 overnight. After that, the superna-
tant was collected by centrifugation for the quantifica-
tion of DNA and GAGs content by a Quant-iT PicoGreen
dsDNA (B1302, Sigma) and the Blyscan glycosaminogly-
can (GAG) assay kit (B100, Biocolor) in accordance with
the outlined protocol. e dECM solution was applied to
slides, air-dried for 30min and then stained with hema-
toxylin–eosin (H&E) and toluidine blue (TB) according
to the Hematoxylin–Eosin Staining Kit (G1120, Solarbio)
Scheme1. Development of injectable dECM-enhanced hyaluronic microgels
Page 5 of 17
Dengetal. Collagen and Leather (2024) 6:14
and the Toluidine Blue Staining Kit (G2543, Solarbio),
respectively. e samples were then observed under
a light microscope and photographed for subsequent
analysis. e microstructure of decellularized tissues was
observed by imaging using a scanning electron micros-
copy (SEM, Japan).
2.4 Preparation andcharacterization ofHE hydrogel
HAMA (10 mg/mL) and dECM solution were mixed at
a mass ratio of 1:2 to obtain the experimental group HE.
en the solution supplemented with photo-initiator was
immediately injected into a homemade ring mold under
light protection. Afterwards, the liquid crosslinked to
form a gel under UV light (450nm), and a series of mate-
rial characterization was carried out in the subsequent
process. e pure HAMA solution was formed into a gel
in the same way as a control.
e lyophilized hydrogel was soaked in liquid nitrogen
for a few seconds, followed by fracturing in a brittle man-
ner to expose the cross-section, and the internal struc-
ture was observed by a scanning electron microscopy
after gold coating treatment. e SEM images were ana-
lyzed using ImageJ software to obtain the porosity and
pore size of the hydrogels.
e disintegration rate of the hydrogels was measured
using hyaluronidase and type I collagenase solutions to
simulate physiological conditions. e weight of fresh
hydrogel was recorded as W0. en the samples were
placed in PBS solutions containing type I collagenase (5
U/mL) and hyaluronidase (5 U/mL), respectively. Simul-
taneously they were placed in a thermostatic shaker at
90rpm and 37 . e fresh enzyme solution was replaced
daily. e hydrogel was removed at certain time intervals,
and the water on the surface was carefully wiped with
filter paper to weigh(W1). e weight measurement was
repeated three times for each sample. e disintegration
rate was calculated using the following formula:
Hydrogel swelling properties were measured as follows.
Briefly, the initial weight of the lyophilized hydrogel was
noted as W0. e samples were immersed in PBS buffer
and placed in a constant temperature shaker at 90rpm
and 37°C. Finally, the hydrogels were periodically taken
out and weighed noted as Ws. e weight measurement
was repeated three times for each sample. Until the
hydrogel swelling reached equilibrium, the swelling ratio
was calculated using the following equation:
In addition, fresh hydrogels were tested at room tem-
perature using a Dynamic Mechanical Analyzer (DMA,
WeightLoss
=
(W0
W1)/W0
100%
SwellingRatio
=
(Ws
W0)/W0
100%
TA-Q800, USA) under multi-frequency mode (a fixed
frequency of 1–10Hz, an amplitude of 40μm, a preload
force of 0.002 N and a force track of 105%) to test the
energy storage modulus (G’) and loss modulus (G’’). Each
sample was tested three times in parallel.
2.5 Preparation andcharacterization ofmicrogels
e microfluidics device was utilized to prepare micro-
gels with uniform size. Briefly, liquid paraffin oil was used
as the continuous phase and Span 80 (5 wt% concentra-
tion) served as the surfactant to stabilize the droplets.
e dispersed phase was composed of either a mixture
of HAMA (1 wt% final concentration) and dECM (2 wt%
final concentration), or a pure HAMA solution with a
photo-initiator. Afterwards, the two phases were individ-
ually injected into different miro-channels of the device
and the flow rates were adjusted by microfluidic pumps.
e change in flow rates resulted in the size of micro-
gels. An ultraviolet (UV) lamp placed above the channels
ensured that microdroplets generated due to the shear
stress of the continuous phase liquid formed solid hydro-
gels by photo-cross-linking at a constant rate within
channels. e microgels were collected in a centrifuge
tube at the outlet of the channel and purified by washing
with acetone once and with PBS repeatedly to obtain HE
and HA microgels.
e purified microgels were photographed under
an optical microscope and the microgel particle size
was analyzed using ImageJ software. SEM was used to
observe the surface morphology of microgels. A certain
amount of microgels was incubated in a PBS solution
placed in a constant temperature shaker at 37°C. At spe-
cific time points, an appropriate volume of the superna-
tant was collected to determine the release profile of each
component from the microgels. Specifically, the collagen
content was quantified using a hydroxyproline assay kit
(Solarbio, BC0250), while IGF-1 (Bioswamp, Wuhan,
PR40070), TGF-β1 (Bioswamp, Wuhan, PR40065), and
BMP-2 (Bioswamp, Wuhan, PR40118) were measured
using enzyme-linked immunosorbent assay (ELISA) kits
to determine the levels of growth factors. Additionally,
the GAG content was assessed using the Blyscan glycosa-
minoglycan (GAG) assay kit (B100, Biocolor).
2.6 Extraction andculture ofOA chondrocytes
e articular cartilage was cleaned with PBS several
times, cut into as thin and small pieces as possible and
digested with EDTA-free trypsase for 0.5 h. e pre-
vious solution was then replaced and incubated with
2mg/mL type II collagenase for 5h. e P0-generation
OA chondrocytes were obtained after passed through a
100μm strainer and cultured in α-MEM medium with
Page 6 of 17
Dengetal. Collagen and Leather (2024) 6:14
the addition of 1% double antibody, 20% KEL serum, and
50μg/mL VC. Once 90% confluence was reached, pas-
saging was accomplished in 24-well plates (P1) for subse-
quent cell experiments.
2.7 In vitroeects ofdierent microgels
2.7.1 Cell proliferation andmorphology
e in vitro effects of microgels on OA chondrocytes
were evaluated through a co-culture system within the
Transwell apparatus. Microgels, prepared at a concentra-
tion of 1mg/mL, were placed in the upper compartments
of a 24-well Transwell plate, with OA chondrocytes
seeded in the lower wells. e proliferation of OA chon-
drocytes, co-cultured with microgels for 3 and 7days,
was assessed using Live/dead cell staining and CCK-8. A
PBS solution containing FDA/PI was added to the well
plates and incubated in darkness for 5min. Samples were
then observed under a fluorescence microscope. Cell
proliferation was detected using the CCK-8 kit, with the
absorbance value of the incubation solution at 450nm
serving as the quantification metric. e cytoskeleton
was stained with a Rhodamine-phalloidin solution to
facilitate observation of cell morphology. Cells were
washed thrice with PBS and soaked in 4% paraformalde-
hyde for 30min at 4 . Subsequently, cells were permea-
bilized with 0.1% Triton X-100 for 10min and cleaned
with PBS three times. ese samples were then stained
with a Rhodamine-phalloidin working solution (5 μg/
mL) overnight at 4 . Finally, cells were washed with PBS
again and stained with a DAPI solution (10μg/mL) for
1 min before observation under a fluorescence micro-
scope. Photographs taken were analyzed by ImageJ soft-
ware to obtain semi-quantitative data on cell spreading
area and roundness.
2.7.2 Cellular matrix secretion
After culturing for 3 and 7days, cartilage-specific poly-
saccharides secreted by OA chondrocytes were assessed
by staining the cells with TB according to the Toluidine
Blue Staining Kit (G2543, Solarbio). Semi-quantitative
analysis of the staining images was done by ImageJ soft-
ware. For quantitative GAG determination, OA chondro-
cytes in the well plates were collected after 3 and 7days.
e DNA and GAG quantitation were performed as
mentioned in 2.4.1.
2.7.3 Chondrogenic phenotypes
To analyze RNA expression, OA chondrocytes after
5days of co-culture with microgels were collected in RNA
Later water and stored at -20 °C. e cellular RNA was
extracted with RNeasy Mini Kit (74,104, Qiagen) and the
concentration was detected by the microspectrophotom-
eter (ND1000, Nanodrop Technologies). Next, RNA was
reversed transcription (RT) extracted to cDNA using the
iScript cDNA Synthesis Kit (BIO-RAD). e polymer-
ase chain reaction (PCR) reaction was conducted on the
CFX96 Touch Real-Time Fluorescent PCR Detection Sys-
tem (BIO-RAD). e expression levels of cartilage matrix-
related genes (GAPDH, COL I, COL II, COL X, SOX9 and
Aggrecan) in OA cells were determined using GAPDH as
an internal reference. e primers are shown in Table S1.
2.8 In vivo eects ofdierent microgels
2.8.1 Establishment ofanOA rat model
e procedures adhered to the current guidelines for
the care of experimental animals. Male SD rats, aged
6–8 weeks and weighing approximately 250 g, were
selected for the establishment of the osteoarthritis
model. e rats were anesthetized with an intraperito-
neal injection of 2% sodium pentobarbital (2mL/kg), and
the anterior cruciate ligament in the knee joint cavity was
disrupted using previously reported methods, namely
anterior cruciate ligament transverse (ACLT) [34]. Four
weeks later, intra-articular injections of 100 μL of PBS, or
PBS mixed with a 10mg/mL concentration of HA or HE
microspheres ranging from 80 to 85μm, were randomly
administered to osteoarthritic rats. e meanings of the
group names are as follows: NA: as well as Native, healthy
rats; PBS: rats treated with PBS solution; HA: rats treated
with HA microgels; HE: rats treated with HE microgels.
2.8.2 Radiographic assessment
Four weeks post-injection, Micro-CT scans were per-
formed on the knee joints of the osteoarthritic rats, and
images were reconstructed based on the scanned data.
e knee width and joint space width were also semi-
quantitatively analyzed in each group.
2.8.3 Histologic evaluation
e rats were executed by over-anesthetized after four
weeks of sample injection, and the collected articular
joints were photographed to observe the width of the
articular synovial cartilage. e gross appearance was
scored according to the scoring criteria (Table S2) [35].
e knee joints were immersed in a 4% paraformalde-
hyde solution for 1week, decalcified with EDTA solu-
tion at 37 , embedded in paraffin, and sectioned. e
sections underwent H&E staining, TB staining, Masson
staining, and Sirius Red staining to analyze the histologi-
cal characteristics of the tissues. e severity of osteoar-
thritis in each group of specimens was assessed based on
the staining results, with reference to the Mankin histo-
logical score (Table S3). Subsequently, the sections were
immunologically stained using antibodies against type I
collagen and type II collagen, following the protocols of
the antibody kits.
Page 7 of 17
Dengetal. Collagen and Leather (2024) 6:14
2.9 Statistical analysis
Statistical analysis was performed using GraphPad
Prism 8 software, and multiple t-tests were used to ana-
lyze inter- and intra-group differences. Data results
were expressed as mean ± standard error (SE) and
mean ± standard deviation (SD) for at least three samples
per test, with p values indicating statistically significant
results (*p < 0.05, **p < 0.01 and ***p < 0.001).
3 Results anddiscussion
3.1 Preparation andcharacterization ofdECM
e decellularization of fresh porcine cartilage slices
was prepared by optimizing the previously established
decellularized cartilage ECM preparation protocol. is
involved performing multiple grinding and sieving to
retain small-sized particles, thereby eliminating cells
and preserving tissue-specific components as much as
possible. As shown in Fig. 1A, for further preparation
of the composite hydrogel, the dECM particles were
digested into the solution by pepsin to be co-mingled
with HAMA to form a gel. DAPI, H&E, and TB stain-
ing, as well as the results of microstructural images and
semiquantitative analyses (Fig.1B, C) demonstrated that
there were virtually no cells in the dECM as compared
to fresh natural cartilage. e success of decellulariza-
tion was further confirmed through DNA quantitative
analysis, revealing the removal of approximately 98% of
immunogenic cellular components. e remaining DNA
concentration in dECM was 44.30 ± 0.51 ng/mg tissue
weight, which is below the critical decellularization crit-
erium of 50ng/mg (Fig.1D) [36]. Critical components of
the ECM were characterized before and after decellulari-
zation. Despite the decellularization process inevitably
results in the loss of ECM components, collagen, with its
Fig. 1 (A) Scheme of decellularized cartilage ECM preparation. (B) Representative DAPI, histologic staining of H&E staining, TB staining and SEM
images of native ECM and dECM. (C) Semi-quantitative determination of DAPI. (D) DNA, (E) Collagen, (F) GAG contents of native ECM and dECM.
Error bars represent SD (n ≥ 3). p < 0.05, p < 0.01, p < 0.001
Page 8 of 17
Dengetal. Collagen and Leather (2024) 6:14
large molecular size, intermolecular entanglement, and
relatively low solubility, exhibited a high retention rate
of approximately 80% after decellularization (Fig. 1E).
GAG quantification showed that after the decellulariza-
tion process, approximately 45% of GAG was retained
(Fig.1F).
3.2 Preparation andcharacterization ofHE hydrogels
HAMA was synthesized by grafting methacrylic anhy-
dride onto HA, and the chemical structure was deter-
mined by 1H NMR as shown in Fig. S1. Distinct new
resonant peaks at 5.8 and 6.2ppm in the HAMA spectra
confirmed the existence of the methacryloyl group sug-
gesting that the hyaluronic acid has been modified by
methacrylic anhydride. In combination of HA and car-
tilage-derived dECM, it formed HE hydrogels to mimic
the physiological extracellular matrix (Fig. 2A). e
microstructure of hydrogels was observed by scanning
electron microscopy (Fig. 2B, C). ey both presented
an interconnected porous structure, which facilitated
nutrient transfer and cell migration and proliferation.
e addition of dECM significantly decreased the pore
size and porosity of the HE hydrogels compared to the
control. DMA results showed that the storage and loss
modulus of the HE hydrogels were higher than those of
the HA group as the frequency increased. is indicated
a significant enhancement in mechanical properties due
to the incorporation of dECM (Fig.2D, E). e degrada-
tion behavior of hydrogels was evaluated by two enzyme
solutions (Fig.2F, G). In the hyaluronidase solution, the
rate of HA loss was higher, suggesting that dECM could
slow down the degradation rate of the material and effec-
tively alleviate the problem of short retention time of HA
in intra-articular injection therapy. In PBS buffer, the
hydrogels rapidly reached solubilization equilibrium and
exhibited excellent water absorption and retention prop-
erties (Fig. 2H). e dual-network hydrogel structure
formed by the addition of dECM to HA was conducive
to delaying the release of the active ingredients of dECM,
especially the large molecular weight components, such
as GAG and collage, realizing its sustained effect on
the recovery of chondrocyte phenotype and cartilage
reconstruction.
3.3 Preparation andcharacterization ofHE microgels
To improve intra-articular injectability for the treatment
of osteoarthritis, the HE microgels were fabricated using
microfluidics devices. As shown in Fig.3A, the continu-
ous phase enters the channel from both sides, generating
and encapsulating the dispersed-phase droplets under
the shear forces of the continuous phase. ese droplets
are then cured by a UV light source above the collection
channel. Upon determining the material concentration,
the flow rate ratio of the continuous and dispersed phases
was adjusted by a syringe pump, resulting in a corre-
sponding change in the diameter of the microgels. In this
study, the flow rate ratio of the continuous and dispersed
phases was set at 10:1. is resulted in HE microgels with
a uniform size distribution and an average particle size
ranging from 80–85 μm. is ensured that the micro-
gels could pass through a 1mL syringe (with an inner
diameter of 0.25mm) without disrupting their morphol-
ogy, thereby ensuring good injectability (Fig.3B). Under
microscopic observation, the microgels were dispersed
in a regular spherical shape. For better observation, a
dye solution was added dropwise to the solution, reveal-
ing that the dECM was uniformly distributed within the
microgels. e microscopic morphology of the microgels
was further studied using scanning electron microscopy,
as shown Fig.3C. e incorporation of dECM enhanced
the composition of microgels with a diverse range of
growth factors, as well as GAG, and collagens. ese
components play crucial roles in the cellular behavior
of chondrocytes, such as proliferation, morphology and
matrix secretion. e release profiles of key growth fac-
tors, such as IGF-1, TGF-1β, and BMP-2, and the carti-
lage-specific structural macromolecules and proteins,
including GAG and collagen, in both microgels were
evaluated. e release curves demonstrated that approxi-
mately 60% of the small molecular weight growth factors
were released within 7days (Fig.3D-F), while the water-
soluble GAG exhibited a slower release trend due to the
formation of a network structure through HA cross-link-
ing (Fig.3G). Consequently, the structural collagen with
high molecular weight demonstrated the slowest release
rate (Fig.3H), and its releasing behavior in hyaluronidase
condition suggested that its gradual release is depend-
ent on the degradation of microgels (Fig.3I). e release
of growth factors, structural ECM macromolecules and
proteins showed a significant temporal correlation, indi-
cating that microgels possess biomimetic properties for
actively modulating the dynamic microenvironment dur-
ing cartilage formation.
3.4 HE microgels enhance proliferation andmorphology
ofOA chondrocytes
To observe the effects of both microgels on cell proliferation
and morphology, transwell chambers were utilized to co-
culture microgels with OA chondrocytes invitro (Fig.4A).
e viability and proliferation of OA chondrocytes were
assessed by live/dead staining result (Fig. 4B) and CCK-8
assay (Fig. 4C). e results collectively demonstrated
that the addition of HE microgels significantly increased
the proliferative activity of OA chondrocytes. Notably,
at 3days, the difference between the HE group and the
remaining groups was the most significant, accompanied
Page 9 of 17
Dengetal. Collagen and Leather (2024) 6:14
by a pronounced increase in cell number. is may be
attributed to the rapid release of growth factors such as
IGF-1, TGF-1β, and BMP-2 from HE hydrogels, which
recover OA chondrocytes’ function to boost proliferation.
OA chondrocytes exhibited irregular cell morphology and
hypertrophy in invitro culture. e ability of HE microgels
to promote morphological recovery of OA chondrocytes
with increasing co-culture time was indicated by rhoda-
mine/DAPI staining (Fig.4D), as well as semi-quantitatively
by spreading area (Fig.4E1) and cell roundness (Fig.4E2).
At 3days, with the rapid release of growth factors from the
HE microgels, the larger cell spreading area of OA chon-
drocytes corresponded to enhanced proliferation, consist-
ent with the CCK-8 data. e decrease in spreading area
and cell roundness of OA chondrocytes in the HE group
at 7days may indicate the synergetic effects of the continu-
ously released growth factors and the slow-released dECM
macromolecules on the recovery of cell morphology.
Fig. 2 (A) Scheme of hydrogel constituents. (B) Hydrogel microstructure (SEM). (C) Pore size and porosity of hydrogels. (D) Storage and E loss
modulus of hydrogels. (F-G) Degradation behavior of hydrogels in two enzyme solutions. (H) Solubilization properties of hydrogels. Error bars
represent SD (n ≥ 3). p < 0.05, p < 0.01, p < 0.001
Page 10 of 17
Dengetal. Collagen and Leather (2024) 6:14
Fig. 3 (A) Schematic diagram of the protocol for the preparation of microgels by microfluidics. (B) Particle size distribution of two kinds
of microgels. (C) SEM images as well as representative light microscopy images of the two microgels. Release curve of D IGF-1, E TGF-1β, F BMP-2,
G GAG, and H, I collagen from microgels
Page 11 of 17
Dengetal. Collagen and Leather (2024) 6:14
3.5 HE microgels promote cartilage matrix secretion ofOA
chondrocytes
GAG is a crucial component of cartilage ECM and serves
as an indicator for assessing the chondrocytes’ function
in cartilage regeneration and homeostasis. e ability of
HE microgels to regulate cartilage ECM secretion in OA
chondrocytes was further investigated. e HE group
exhibited superior pro-chondrogenic matrix secretion in
OA chondrocytes compared to the HA group (Fig.5A),
as evidenced by stronger positive staining from tolui-
dine blue (TB) staining after 3 and 7days of co-culture,
corroborated by semi-quantitative data (Fig. 5B). HE
microgels contributed to an increase in cell number
and enhancement of cartilage-specific polysaccharide
synthesis, as shown in the quantitative analysis of DNA
and GAG (Fig.5C-E). e regulatory effect of HE micro-
gels on cartilage matrix synthesis in OA chondrocytes
was not significantly evident at 3 days, suggesting that
the observed effect during this time period may be pri-
marily attributed to enhanced proliferation based on the
CCK-8 data. However, the GAG/DNA values of the HE
group significantly surpassed those of the other groups at
7days, providing further evidence for the enhancement
of matrix synthesis by growth factors and degradation
products. In addition, the qPCR assessed the expression
of various cartilage-specific genes in OA chondrocytes
after 5 days of co-culture with microgels (Fig.5F): the
expression of COL I
Fig. 4 (A) Schematic diagram of co-culture method assessing interactions between porous microgels and cells in vitro. (B) Live/dead staining
images of OA cells after 3 and 7 days of co-culture. (C) Measurement of OA cell proliferation by CCK-8 kit. (D) Cytoskeleton staining after 3
and 7 days of co-culture, cell spreading area (E1), cell circularity (E2). Error bars represent SD (n ≥ 3). p < 0.05, p < 0.01, p < 0.001
Page 12 of 17
Dengetal. Collagen and Leather (2024) 6:14
gene was associated with chondrogenic fibrosis, COL
II and AGG represented ECM synthesis, COL X gene was
expressed in chondrocyte hypertrophy, and SOX9 was
the essential cartilage-promoting factor, which can alle-
viate the progression of OA. HE microgels significantly
decreased the expression level of COL I and COL X genes
and significantly increased the expression of COL II, AGG,
and SOX9. is finding was further supported by the close
relationship among the three key growth factors and the
structural ECM macromolecules towards the chondro-
cyte’s phenotype, based on the gene interaction network
map generated using the STRING database (Fig.5G). In
summary, HE microgels demonstrated enhanced secre-
tion of cartilage matrix and expression of cartilage-specific
genes in OA chondrocytes invitro, as evidenced by his-
tological staining, GAG/DNA analysis, and gene expres-
sion levels. is can be attributed to the synergistic effect
of spatiotemporal release of cartilage-specific molecules,
which rapidly recover the function of OA chondrocytes
with the short-term release of cartilage-specific growth
factors, and continuously support the cartilage regen-
eration and homeostasis with the long-term release of
Fig. 5 (A) Toluidine blue staining images and B semi-quantitative results of OA chondrocytes after 3 and 7 days of co-culture. (C) GAG,
D DNA, and E GAG/DNA quantification. (F) Expression levels of cartilage matrix-related genes in OA chondrocytes co-cultured for 5 days. (G)
Plot of the relationship between ECM proteins genes, growth factors genes and cartilage phenotype related genes. Error bars represent SD
(n ≥ 3). p < 0.05, p < 0.01, p < 0.001
Page 13 of 17
Dengetal. Collagen and Leather (2024) 6:14
structural ECM macromolecules and proteins, such as
GAGs and collagens, as the HE microgels degrades.
3.6 In vivoevaluation ofHE microgels forOA alleviation
e therapeutic potential of HE microgels for OA alle-
viation was evaluated using a rat OA model established
by ACLT surgery. Different samples were injected into the
joint cavity four weeks post-surgery (Fig.6A), and the joints
were reconstructed using micro-computed tomography
(micro-CT) to assess the OA progression. e subchondral
bone treated with PBS was found to be severely deformed,
with a concave and uneven surface. In contrast, the joints
Fig. 6 (A) Scheme of evaluating the effect of microgels in OA rat models. (B) micro-CT, (C) joint space width and D knee width of samples.
(E) gross appearance of the keen joints four weeks after sample injection and F gross morphology score. Error bars represent SD
(n = 6). p < 0.05, p < 0.01, p < 0.001
Page 14 of 17
Dengetal. Collagen and Leather (2024) 6:14
treated with HE microgels more closely resembled the
morphology and structure of healthy rat joints (Fig. 6B).
According to semi-quantitative knee analysis (Fig.6C, D),
there was no statistical difference between the HE micro-
gel treatment and the native group (NA). A decrease in
joint space width reduction and maintenance of knee
width suggest that HE microgels attenuate osteoarthritic
degeneration after four weeks of injection [37]. e gross
view of the knee joint (Fig.6E) and the gross morphology
score (Fig.6F) were consistent with the joint visualization
results, suggesting the effectiveness of the treatment.
e H&E staining of the natural joints showed neat and
orderly chondrocyte arrangement, a homogeneous matrix,
and a smooth surface of the cartilage layer (Fig. 7A). In
Fig. 7 Four weeks after intra-articular injection, tissue sections of rat OA knee joints were stained for A H&E, B TB, C Masson, D Sirius Red, E COL II,
and F COL I. Semi-quantitative analysis of G TB, H COL II, and I COL I staining, J Mankin histologic scores for cartilage structure, chondrocyte, matrix
staining and tidal line integrity (n = 6). p < 0.05, p < 0.01, p < 0.001
Page 15 of 17
Dengetal. Collagen and Leather (2024) 6:14
contrast, the PBS group exhibited significant tissue fibro-
sis, disorganized cell arrangement, and an uneven carti-
lage layer. Despite the lubricating effect of HA leading to a
smoother surface, the HA-treated group displayed a reduc-
tion in thickness and lighter staining color matrix layer
along with irregularly arranged cells. is suggests that
HA’s ability to effectively repair damaged cartilage tissue
is limited. On the other hand, the HE group showed a sig-
nificant improvement in both cell arrangement and matrix,
which can be attributed to the combined lubricating prop-
erties of HA and the cartilage matrix synthesizing function
of dECM. Furthermore, the TB (Fig.7B), Masson (Fig.7C),
Sirius Red (Fig.7D) staining and semi-quantitative analysis
(Fig.7G) allowed for a general assessment of GAG levels.
Among the experimental groups, representative images of
the HE group demonstrated the superior results by best
preserving the GAGs and minimizing the fibrocartilage
production in the regenerated cartilage matrix. In addition,
COL I and II, classic indicators representing the quality of
regenerated cartilage, were assessed by immunohistochem-
ical staining (Fig.7E, F) and semiquantitative data (Fig.7H,
I) to further evaluate the quality of neocartilage tissues.
e HE group exhibited significantly higher production of
COL II with minimal level of COL I, indicating its supe-
riority in the regeneration of healthier hyaline cartilage.
Based on above observations, Mankin histological scores
(Fig.7J) were utilized to illustrate the differences between
the experimental groups and the natural group in terms of
cartilage structure, chondrocytes, matrix staining, and tide-
mark integrity. e results suggested that the HE groups
had achieved the significant improvements over the PBS
and HA groups in the overall Mankin score, with distinct
amelioration in OA cartilage structure and matrix stain-
ing. Collectively, HE microgels effectively promoted the
repair of articular cartilage and significantly alleviated the
degree of OA, evidenced by radiological observation, gross
appearance, histological/immunohistochemical staining,
and analysis in an OA rat model invivo.
4 Conclusion
is study successfully pioneered a cell-free OA therapy
approach, utilizing intra-articular injectable dECM-
enhanced HA microgels. ese microgels, characterized
by their uniform size and mechanical strength, were fab-
ricated using microfluidics and photopolymerization.
ey provided a spatiotemporal cascade effect, enabling
the rapid release of cartilage-specific growth factors such
as IGF-1, TGF-β, and BMP-2. is facilitated the swift
recovery of OA chondrocytes’ function. Simultaneously,
the slower release of cartilage structural ECM macromole-
cules and proteins, including GAGs and collagens, contin-
uously supplied essential elements for supporting cartilage
regeneration and homeostasis as the microgels degraded.
is enhancement was observed in terms of cell prolifera-
tion, morphology, matrix synthesis, and cartilage-specific
gene expression. Furthermore, HE microgels effectively
promoted the regeneration of collagen II-enriched hya-
line cartilage and significantly alleviated the degree of OA,
evidenced by radiological observation, gross appearance,
histological/immunohistochemical staining, and analysis
in an OA rat model invivo. Together, it underscores the
potential of HE microgels in dECM combined with HA as
a promising therapeutic approach for OA.
Abbreviations
OA Osteoarthritis
HE DECM-enhanced hyaluronic
NSAIDs Non-steroidal anti-inflammatory drugs
HA Hyaluronic acid
ECM Extracellular matrix
dECM Decellularized ECM
TGF-β Transforming growth factor-β
IGF-1 Insulin-like growth factor-1
BMP Bone morphogenetic protein
FGF Fibroblast growth factor
EGF Epidermal growth factor
MA Methacrylic anhydride
CHAPS 3-[(3-Cholamidopropyl) dimethylamino] propanesulfonic acid inter-
nal salt
HAMA Hyaluronic acid methacryloyl
GAG Glycosaminoglycan
H&E Hematoxylin–eosin
TB Toluidine blue
Supplementary Information
The online version contains supplementary material available at https:// doi.
org/ 10. 1186/ s42825- 024- 00158-6.
Supplementary Material 1.
Acknowledgements
We would like to thank Sichuan University Analysis & Testing Center for techni-
cal assistance with Micro-CT.
Authors’ contributions
SYD designed this project and revised the manuscript. SYD, HFC, YL and WQS
performed experiments and analyzed data. MYC and XLC performed experi-
ments and draw the scheme. JL, YJF, QGW and XDZ reviewed the manuscript.
All authors read and approved the final manuscript.
Funding
This work was supported by the National Key Research Program of China
(2023YFB4605800), the Natural Science Foundation of China (32071353) and
the 111 Project (B16033).
Availability of data and materials
All data generated or analyzed during this study are presented in this article.
Declarations
Ethics approval and consent to participate
Human OA articular cartilage tissue was obtained from total joint replacement
surgery under the ethical support of the Biomedical Ethics Review Committee
of West China Hospital, Sichuan University (2022200). The animal experiments
were approved by the Ethical Committee of Sichuan University (KS2020308).
All the animals were purchased from Laboratory Animal Center of Sichuan
University. The animal experiment guidance from the ethical committee and
Page 16 of 17
Dengetal. Collagen and Leather (2024) 6:14
the guide for care and use of laboratory animals of Sichuan University were
followed during the whole experiment course.
Competing interests
The authors declare that they have no known competing financial interests
or personal relationships that could have appeared to influence the work
reported in this paper.
Author details
1 National Engineering Research Center for Biomaterials, Sichuan University,
Chengdu, Sichuan 610065, China. 2 College of Biomedical Engineering,
Sichuan University, Chengdu, Sichuan 610065, China. 3 School of Medicine,
The Chinese University of Hong Kong, Shenzhen 518172, China. 4 Department
of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineer-
ing & Nanomedicine, University of Otago, Christchurch 8011, New Zealand.
5 Sichuan Testing Center for Biomaterials and Medical Devices Co.Ltd, 29
Wangjiang Road, Chengdu, Sichuan, China.
Received: 18 January 2024 Revised: 21 March 2024 Accepted: 24 March
2024
References
1. Cui A, Li H, Wang D, Zhong J, Chen Y, Lu H. Global, regional prevalence,
incidence and risk factors of knee osteoarthritis in population-based
studies. EClinicalMedicine. 2020;29–30:100587.
2. da Costa BR, Pereira TV, Saadat P, Rudnicki M, Iskander SM, Bodmer NS,
et al. Effectiveness and safety of non-steroidal anti-inflammatory drugs
and opioid treatment for knee and hip osteoarthritis: network meta-
analysis. Br Med J. 2021;375:n2321.
3. Zhang W, Moskowitz RW, Nuki G, Abramson S, Altman RD, Arden N,
et al. OARSI recommendations for the management of hip and knee
osteoarthritis, Part II: OARSI evidence-based, expert consensus guide-
lines. Osteoarthritis Cartilage. 2008;16(2):137–62.
4. Tommaso I, Daniele L, Beniamino P. Intra-articular injections for the
treatment of osteoarthritis focus on the clinical use of hyaluronic acid.
Drugs R D. 2011;11(1):13–27.
5. CatNTC C. Vascular and upper gastrointestinal effects of non-steroidal
anti-inflammatory drugs: meta-analyses of individual participant data
from randomised trials. Lancet. 2013;382(9894):769–79.
6. Kou L, Xiao S, Sun R, Bao S, Yao Q, Chen R. Biomaterial-engineered
intra-articular drug delivery systems for osteoarthritis therapy. Drug
Deliv. 2019;26(1):870–85.
7. Watterson JR, Esdaile JM. Viscosupplementation: therapeutic mecha-
nisms and clinical potential in osteoarthritis of the knee. J Am Acad
Orthop Surg. 2000;8(5):277–84.
8. Lucke M, Messner W, Kim ESH, Husni ME. The impact of identifying
carotid plaque on addressing cardiovascular risk in psoriatic arthritis.
Arthritis Res Ther. 2016;18:1733.
9. Fakhari A, Berkland C. Applications and emerging trends of hyaluronic
acid in tissue engineering, as a dermal filler and in osteoarthritis treat-
ment. Acta Biomater. 2013;9(7):7081–92.
10. Maudens P, Meyer S, Seemayer CA, Jordan O, Allémann E. Self-assembled
thermoresponsive nanostructures of hyaluronic acid conjugates for
osteoarthritis therapy. Nanoscale. 2018;10(4):1845–54.
11. Stellavato A, dE Novellis F, Reale S, De Rosa M, Schiraldi C. Hybrid com-
plexes of high and low molecular weigth: Evaluation using an in vitro
model of osteoarthritis. J Biol Regul Homeost Agents. 2016;30(4):7–15.
12. Lin X, Tsao CT, Kyomoto M, Zhang M. Injectable natural polymer
hydrogels for treatment of knee osteoarthritis. Adv Healthc Mater.
2021;11(9):2101479.
13. Atwal A, Dale TP, Snow M, Forsyth NR, Davoodi P. Injectable hydrogels:
An emerging therapeutic strategy for cartilage regeneration. Adv Colloid
Interface Sci. 2023;321:103030.
14. Tezel A, Fredrickson GH. The science of hyaluronic acid dermal fillers. J
Cosmet Laser Ther. 2008;10(1):35–42.
15. Seong Y-J, Lin G, Kim BJ, Kim H-E, Kim S, Jeong S-H. Hyaluronic acid-based
hybrid hydrogel microspheres with enhanced structural stability and
high injectability. ACS Omega. 2019;4(9):13834–44.
16. Lei Y, Wang Y, Shen J, Cai Z, Zhao C, Hong C, et al. Injectable hydrogel
microspheres with self-renewable hydration layers alleviate osteoarthritis.
Sci Adv. 2022;8(5):eab16449.
17. Yu H, Huang C, Kong X, Ma J, Ren P, Chen J, et al. Nanoarchitectonics of
cartilage-targeting hydrogel microspheres with reactive oxygen species
responsiveness for the repair of osteoarthritis. ACS Appl Mater Interfaces.
2022;14(36):40711–23.
18. Cunniffe GM, Díaz-Payno PJ, Sheehy EJ, Critchley SE, Almeida HV,
Pitacco P, et al. Tissue-specific extracellular matrix scaffolds for the
regeneration of spatially complex musculoskeletal tissues. Biomaterials.
2019;188:63–73.
19. Jiang W, Zhang X, Yu S, Yan F, Chen J, Liu J, et al. Decellularized
extracellular matrix in the treatment of spinal cord injury. Exp Neurol.
2023;368:114506.
20. Wang X, Lu Y, Wang W, Wang Q, Liang J, Fan Y, et al. Effect of different
aged cartilage ECM on chondrogenesis of BMSCs in vitro and in vivo.
Regen Biomater. 2020;7(6):583–95.
21. Peng Z, Sun H, Bunpetch V, Koh Y, Wen Y, Wu D, et al. The regulation of
cartilage extracellular matrix homeostasis in joint cartilage degeneration
and regeneration. Biomaterials. 2021;268:120555.
22. Grogan SP, Chen X, Sovani S, Taniguchi N, Colwell CW, Lotz MK, et al. Influ-
ence of cartilage extracellular matrix molecules on cell phenotype and
neocartilage formation. Tissue Eng Part A. 2014;20(1–2):264–74.
23. Chen Y, Mehmood K, Chang Y-F, Tang Z, Li Y, Zhang H. The molecular
mechanisms of glycosaminoglycan biosynthesis regulating chondrogen-
esis and endochondral ossification. Life Sci. 2023;335:122243.
24. Rosenzweig DH, Solar-Cafaggi S, Quinn TM. Functionalization of dynamic
culture surfaces with a cartilage extracellular matrix extract enhances
chondrocyte phenotype against dedifferentiation. Acta Biomater.
2012;8(9):3333–41.
25. Kock L, van Donkelaar CC, Ito K. Tissue engineering of functional articular
cartilage: the current status. Cell Tissue Res. 2011;347(3):613–27.
26. Lin S, Li H, Wu B, Shang J, Jiang N, Peng R, et al. TGF-β1 regu-
lates chondrocyte proliferation and extracellular matrix synthesis
via circPhf21a-Vegfa axis in osteoarthritis. Cell Commun Signal.
2022;20(1):1–18.
27. Park H, Temenoff JS, Holland TA, Tabata Y, Mikos AG. Delivery of TGF-β1
and chondrocytes via injectable, biodegradable hydrogels for cartilage
tissue engineering applications. Biomaterials. 2005;26(34):7095–103.
28. Shi S, Kelly BJ, Wang C, Klingler K, Chan A, Eckert GJ, et al. Human IGF-I
propeptide A promotes articular chondrocyte biosynthesis and employs
glycosylation-dependent heparin binding. Biochim Biophys Acta-Gen
Subj. 2018;1862(3):567–75.
29. Wen C, Xu L, Xu X, Wang D, Liang Y, Duan L. Insulin-like growth factor-1
in articular cartilage repair for osteoarthritis treatment. Arthritis Res Ther.
2021;23(1):277.
30. Liu X. BMP2 promotes chondrocyte proliferation via the Wnt/β-catenin
signaling pathway. Mol Med Rep. 2011;4(4):621–6.
31. Gründer T, Gaissmaier C, Fritz J, Stoop R, Hortschansky P, Mollenhauer J,
et al. Bone morphogenetic protein (BMP)-2 enhances the expression of
type II collagen and aggrecan in chondrocytes embedded in alginate
beads. Osteoarthritis Cartilage. 2004;12(7):559–67.
32. Lu Y, Wang Y, Zhang H, Tang Z, Cui X, Li X, et al. Solubilized cartilage ecm
facilitates the recruitment and chondrogenesis of endogenous bmscs
in collagen scaffolds for enhancing microfracture treatment. ACS Appl
Mater Interfaces. 2021;13(21):24553–64.
33. Cao H, Wang X, Chen M, Liu Y, Cui X, Liang J, et al. Childhood carti-
lage ecm enhances the chondrogenesis of endogenous cells and
subchondral bone repair of the unidirectional collagen–decm scaf-
folds in combination with microfracture. ACS Appl Mater Interfaces.
2021;13(48):57043–57.
34. Cao H, Chen M, Cui X, Liu Y, Liu Y, Deng S, et al. Cell-Free Osteoarthritis
Treatment with Sustained-Release of Chondrocyte-Targeting Exosomes
from Umbilical Cord-Derived Mesenchymal Stem Cells to Rejuvenate
Aging Chondrocytes. ACS Nano. 2023;17(14):13358–76.
Page 17 of 17
Dengetal. Collagen and Leather (2024) 6:14
35. Pritzker KPH, Gay S, Jimenez SA, Ostergaard K, Pelletier JP, Revell PA, et al.
Osteoarthritis cartilage histopathology: grading and staging. Osteoarthri-
tis Cartilage. 2006;14(1):13–29.
36. Kawecki M, Łabuś W, Klama-Baryla A, Kitala D, Kraut M, Glik J, et al. A
review of decellurization methods caused by an urgent need for quality
control of cell-free extracellular matrix’ scaffolds and their role in regen-
erative medicine. J Biomed Mater Res Part B. 2017;106(2):909–23.
37. Lei Y, Wang Y, Shen J, Cai Z, Zhao C, Chen H, et al. Injectable hydrogel
microspheres with self-renewable hydration layers alleviate osteoarthritis.
Sci Adv. 2022;8(5):eabl6449.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The impairment of articular cartilage due to traumatic incidents or osteoarthritis has posed significant challenges for healthcare practitioners, researchers, and individuals suffering from these conditions. Due to the absence of an approved treatment strategy for the complete restoration of cartilage defects to their native state, the tissue condition often deteriorates over time, leading to osteoarthritic (OA). However, recent advancements in the field of regenerative medicine have unveiled promising prospects through the utilization of injectable hydrogels. This versatile class of biomaterials, characterized by their ability to emulate the characteristics of native articular cartilage, offers the distinct advantage of minimally invasive administration directly to the site of damage. These hydrogels can also serve as ideal delivery vehicles for a diverse range of bioactive agents, including growth factors, anti-inflammatory drugs, steroids, and cells. The controlled release of such biologically active molecules from hydrogel scaffolds can accelerate cartilage healing, stimulate chondrogenesis, and modulate the inflammatory microenvironment to halt osteoarthritic progression. The present review aims to describe the methods used to design injectable hydrogels, expound upon their applications as delivery vehicles of biologically active molecules, and provide an update on recent advances in leveraging these delivery systems to foster articular cartilage regeneration.
Article
Full-text available
Background The transforming growth factor-beta (TGF-β) signaling pathway is an important pathway associated with the pathogenesis of osteoarthritis (OA). This study was to investigate the involvement of circRNAs in the TGF-β signaling pathway. Methods Cell Counting Kit-8 (CCK-8) assay and 5-ethynyl-2′-deoxyuridine (EdU) assay were used to detect the proliferation of primary mouse chondrocytes (PMCs). RNA-sequencing together with bioinformatics analysis were used to systematically clarify TGF-β1 induced alternations of circRNAs in PMCs. The regulatory and functional role of circPhf21a was examined in PMCs. Downstream targets of circPhf21a were explored by RNA-sequencing after overexpression of circPhf21a and verified by RT-qPCR in PMCs. Finally, the role and mechanism of circPhf21a in OA were explored in mouse models. Results We found that TGF-β1 promoted the proliferation of PMCs. Meanwhile, RT-qPCR and western blotting indicated that TGF-β1 promoted extracellular matrix (ECM) anabolism. RNA-sequencing revealed that a total of 36 circRNAs were differentially expressed between PMCs treated with and without TGF-β1. Of these, circPhf21a was significantly decreased by TGF-β1. Furthermore, circPhf21a knockdown promoted the proliferation and ECM synthesis of PMCs, whereas overexpression of circPhf21a showed the opposite effects. Mechanically, the expression profiles of the mRNAs revealed that Vegfa may be the target of circPhf21a. Additionally, we found that circPhf21a was significantly upregulated in the mouse OA model, and inhibition of circPhf21a significantly relieved the progression of OA. Conclusions Our results found that TGF-β1 promoted the proliferation and ECM synthesis of PMCs via the circPhf21a-Vegfa axis, which may provide novel therapeutic targets for OA treatment.
Article
Full-text available
Introducing hydration layers to hydrogel microspheres (HMs) by coating the surface with liposomes can effectively reduce friction. However, the lubrication can be inactivated when the surface coatings are damaged. To endow HMs with the ability to form self-renewable hydration layers and maintain cellular homeostasis, rapamycin-liposome-incorporating hyaluronic acid-based HMs (RAPA@Lipo@HMs) were created using microfluidic technology and photopolymerization processes. The RAPA@Lipo@HMs improve joint lubrication by using a smooth rolling mechanism and continuously exposing liposomes on the outer surface to form self-renewable hydration layers via frictional wear. In addition, the released autophagy activator (rapamycin)-loaded cationic liposomes can target negatively charged cartilage through electrostatic interactions and maintain cellular homeostasis by increasing autophagy. Furthermore, the in vivo data showed that the RAPA@Lipo@HMs can alleviate joint wear and delay the progression of osteoarthritis. The RAPA@Lipo@HMs can provide efficient lubrication and potentially alleviate friction-related diseases such as osteoarthritis.
Article
Full-text available
Articular cartilage repair is a critical issue in osteoarthritis (OA) treatment. The insulin-like growth factor (IGF) signaling pathway has been implicated in articular cartilage repair. IGF-1 is a member of a family of growth factors that are structurally closely related to pro-insulin and can promote chondrocyte proliferation, enhance matrix production, and inhibit chondrocyte apoptosis. Here, we reviewed the role of IGF-1 in cartilage anabolism and catabolism. Moreover, we discussed the potential role of IGF-1 in OA treatment. Of note, we summarized the recent progress on IGF delivery systems. Optimization of IGF delivery systems will facilitate treatment application in cartilage repair and improve OA treatment efficacy.
Article
Full-text available
Osteoarthritis (OA) is a serious chronic and degenerative disease that increasingly occurs in the aged population. Its current clinical treatments are limited to symptom relief and cannot regenerate cartilage. Although a better understanding of OA pathophysiology has been facilitating the development of novel therapeutic regimen, delivery of therapeutics to target sites with minimal invasiveness, high retention, and minimal side effects remains a challenge. Biocompatible hydrogels have been recognized to be highly promising for controlled delivery and release of therapeutics and biologics for tissue repair. In this review, the current approaches and the challenges in OA treatment, and unique properties of injectable natural polymer hydrogels as delivery system to overcome the challenges are presented. The common methods for fabrication of injectable polysaccharide‐based hydrogels and the effects of their composition and properties on the OA treatment are detailed. The strategies of the use of hydrogels for loading and release cargos are also covered. Finally, recent efforts on the development of injectable polysaccharide‐based hydrogels for OA treatment are highlighted, and their current limitations are discussed.
Article
Functional limitation caused by spinal cord injury (SCI) has the problem of significant clinical and economic burden. Damaged spinal axonal connections and an inhibitory environment severely hamper neuronal function. Regenerative biomaterials can fill the cavity and produce an optimal microenvironment at the site of SCI, inhibiting apoptosis, inflammation, and glial scar formation while promoting neurogenesis, axonal development, and angiogenesis. Decellularization aims to eliminate cells from the ultrastructure of tissues while keeping tissue-specific components that are similar to the structure of real tissues, making decellularized extracellular matrix (dECM) a suitable scaffold for tissue engineering. dECM has good biocompatibility, it can be widely obtained from natural organs of different species, and can be co-cultured with cells for 3D printing to obtain the target scaffold. In this paper, we reviewed the pathophysiology of SCI, the characteristics of dECM and its preparation method, and the application of dECM in the treatment of SCI. Although dECM has shown its therapeutic effect at present, there are still many indicators that need to be taken into account, such as the difficulty in obtaining materials and standardized production mode for large-scale use, the effect of decellularization on the physical and chemical properties of dECM, and the study on the synergistic effect of dECM and cells.
Article
Clinically, intra-articular administration can hardly achieve the truly targeted therapy, and the drugs are usually insufficient to show local and long-term therapeutic effects because of their rapid clearance. Herein, inspired by the phenomenon that bees track the scent of flowers to collect nectar, we developed cartilage-targeting hydrogel microspheres with reactive oxygen species (ROS)-responsive ability via combining the microfluidic method and photopolymerization processes to integrate cartilage-targeting peptides and ROS-responsive nanoparticles in the hydrogel matrix. The hydrogel microspheres with cartilage-targeting properties promoted better retention in the joint cavity and enhanced cellular uptake of the nanoparticles. Moreover, the ROS-responsive nanoparticles could react with osteoarthritis (OA)-induced intracellular ROS, resulting in the depolymerization of nanoparticles, which could not only eliminate excess ROS and reduce inflammation but also promote the release of dexamethasone (Dex) and kartogenin (KGN) in situ, realizing effective OA therapy. It was demonstrated that this hydrogel microsphere showed favorable ROS-responsive ability and enhanced chondrogenic differentiation as well as the downregulation of pro-inflammatory factors in vitro. Additionally, the hydrogel microspheres, similar to bees, could target and effectively repair cartilage in the OA model. Thus, the injectable hydrogel microspheres exerted an excellent potential to repair OA and may also provide an effective avenue for inflammatory bowel disease therapy.
Article
Objective: To assess the effectiveness and safety of different preparations and doses of non-steroidal anti-inflammatory drugs (NSAIDs), opioids, and paracetamol for knee and hip osteoarthritis pain and physical function to enable effective and safe use of these drugs at their lowest possible dose. Design: Systematic review and network meta-analysis of randomised trials. Data sources: Cochrane Central Register of Controlled Trials (CENTRAL), Medline, Embase, regulatory agency websites, and ClinicalTrials.gov from inception to 28 June 2021. Eligibility criteria for selecting studies: Randomised trials published in English with ≥100 patients per group that evaluated NSAIDs, opioids, or paracetamol (acetaminophen) to treat osteoarthritis. Outcomes and measures: The prespecified primary outcome was pain. Physical function and safety outcomes were also assessed. Review methods: Two reviewers independently extracted outcomes data and evaluated the risk of bias of included trials. Bayesian random effects models were used for network meta-analysis of all analyses. Effect estimates are comparisons between active treatments and oral placebo. Results: 192 trials comprising 102 829 participants examined 90 different active preparations or doses (68 for NSAIDs, 19 for opioids, and three for paracetamol). Five oral preparations (diclofenac 150 mg/day, etoricoxib 60 and 90 mg/day, and rofecoxib 25 and 50 mg/day) had ≥99% probability of more pronounced treatment effects than the minimal clinically relevant reduction in pain. Topical diclofenac (70-81 and 140-160 mg/day) had ≥92.3% probability, and all opioids had ≤53% probability of more pronounced treatment effects than the minimal clinically relevant reduction in pain. 18.5%, 0%, and 83.3% of the oral NSAIDs, topical NSAIDs, and opioids, respectively, had an increased risk of dropouts due to adverse events. 29.8%, 0%, and 89.5% of oral NSAIDs, topical NSAIDs, and opioids, respectively, had an increased risk of any adverse event. Oxymorphone 80 mg/day had the highest risk of dropouts due to adverse events (51%) and any adverse event (88%). Conclusions: Etoricoxib 60 mg/day and diclofenac 150 mg/day seem to be the most effective oral NSAIDs for pain and function in patients with osteoarthritis. However, these treatments are probably not appropriate for patients with comorbidities or for long term use because of the slight increase in the risk of adverse events. Additionally, an increased risk of dropping out due to adverse events was found for diclofenac 150 mg/day. Topical diclofenac 70-81 mg/day seems to be effective and generally safer because of reduced systemic exposure and lower dose, and should be considered as first line pharmacological treatment for knee osteoarthritis. The clinical benefit of opioid treatment, regardless of preparation or dose, does not outweigh the harm it might cause in patients with osteoarthritis. Systematic review registration: PROSPERO number CRD42020213656.