Skip to main content
Cell Transplantation logoLink to Cell Transplantation
. 2023 Sep 9;32:09636897231196493. doi: 10.1177/09636897231196493

Injectable Decorin/Gellan Gum Hydrogel Encapsulating Adipose-Derived Stem Cells Enhances Anti-Inflammatory Effect in Cartilage Injury via Autophagy Signaling

Weiping He 1,*, Yu Wu 2,*, Zhihong Luo 1, Genghua Yang 1, Woquan Ye 1, Xi Chen 3, Jianhua Ren 3, Tangzhao Liang 3, Zhiqiang Liao 1, Shihai Jiang 4, Kun Wang 3,
PMCID: PMC10493051  PMID: 37688441

Abstract

Adipose-derived stem cells (ADSCs) are employed as a promising alternative in treating cartilage injury. Regulating the inflammatory “fingerprint” of ADSCs to improve their anti-inflammatory properties could enhance therapy efficiency. Herein, a novel injectable decorin/gellan gum hydrogel combined with ADSCs encapsulation for arthritis cartilage treatment is proposed. Decorin/gellan gum hydrogel was prepared according to the previous manufacturing protocol. The liquid–solid form transition of gellan gum hydrogel is perfectly suitable for intra-articular injection. Decorin-enriched matrix showing an immunomodulatory ability to enhance ADSCs anti-inflammatory phenotype under inflammation microenvironment by regulating autophagy signaling. This decorin/gellan gum/ADSCs hydrogel efficiently reverses interleukin-1β-induced cellular injury in chondrocytes. Through a mono-iodoacetate-induced arthritis mice model, the synergistic therapeutic effect of this ADSCs-loaded hydrogel, including inflammation attenuation and cartilage protection, is demonstrated. These results make the decorin/gellan gum hydrogel laden with ADSCs an ideal candidate for treating inflammatory joint disorders.

Keywords: ADSCs, decorin, gellan gum, autophagy, arthritis

Introduction

Intra-articular injection of adipose-derived stem cells (ADSCs) has been developed as an important therapeutic approach to address arthritis symptoms distributed from animal models 1 to clinical practice 2 . ADSCs can be harvested easily and repeatedly with minimally invasive liposuctions in clinical practice. Clinical outcomes including functional improvement and pain relief have been revealed by therapeutic applications on osteoarthritis (OA)35, rheumatoid arthritis (RA), and cartilage defect disorder6,7. The existing inflammatory microenvironment is the main cause of cartilage degradation and pain since the inflammatory cytokines induce an increase in metalloproteases expression enhancing cartilage degradation8,9. ADSCs regulate inflammatory milieu in the chronic inflammatory joint disorder by modulating pro-inflammatory cytokines interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α) and governing immunomodulated anti-inflammatory effects1012. On the contrary, the articular inflammatory microenvironment also has a huge impact on the inflammatory phenotype of ADSC. A previous study suggested that inflammatory commitment of ADSC induced by IL-1β activates nuclear factor kappa-B signaling in chondrocytes and synoviocytes 13 . Furthermore, TNF-α exposure inhibited glycosaminoglycan (GAG) deposition and chondrogenic gene expression in ADSCs 14 , indicating that the pro-inflammatory microenvironment could hamper the therapeutic efficiency of ADSC by cytokines interaction. Therefore, regulating the inflammatory “fingerprint” of ADSC and maintaining its anti-inflammatory potential is crucial to the success of ADSC-injected therapy.

Mesenchymal stem cells (MSCs) chondrogenesis involved harmonious crosstalk between extracellular matrix (ECM) components and MSCs 15 . Decorin, a small leucine-rich proteoglycan within cartilage ECM 16 , enhanced chondrogenic differentiation of MSCs in gel matrix by inducing GAG and type II collagen deposition 17 . Interaction between sulfate chains on decorin and peripheral cells controlled the biomechanical environment and the biochemical transduction into cartilage cells18,19. Indeed, loss of decorin resulted in greater GAG loss and formation of severer OA symptoms in the post-traumatic OA model 20 , indicating a protective role of decorin in late-stage cartilage degeneration 21 . Interestingly, patients with spondyloarthropathies showed upregulated decorin antibody levels in synovial fluid compared with OA patients 22 , suggesting an involved role of decorin in regulating inflammatory rheumatic diseases. Of note, alleviation of antigen-induced arthritis by corticosteroid administration elevated decorin mRNA levels in connective tissues 23 . Furthermore, co-culture MSCs could reduce the pro-inflammatory effect of TNFα and IL-1β on cartilage by increasing decorin secretion 24 , in line with the fact that decorin may interact with extracellular tyrosine kinase receptors for immunol signaling transduction in inflammatory diseases 25 . Given the fact that decorin-overexpressed MSCs significantly attenuate inflammation by regulating immune responses 26 , we therefore wonder whether pericellular decorin would enhance the anti-inflammatory property of ADSCs that facilitate cartilage repair.

Recent studies from the L.M. Grover research group developed decorin-encapsulated gellan gum hydrogel for sustained release of decorin and delivering anti-inflammatory effect27,28. The transition of solid to fluid-forming hydrogel under shear stress makes it as ideal for injection into the intra-articular space. Integrated MSCs with gellan gum hydrogel have been shown effective for osteochondral repair 29 . In addition, decorin shows regulating immune cell dynamics 30 via mediating outside-in innate autophagy signaling 31 . Inhibiting autophagy is associated with cartilage matrix degeneration 32 . In particular, autophagy is required to maintain the anti-inflammatory potential of ADSCs33,34, whereas inhibition of autophagy mediated stress-induced apoptosis in ADSCs 35 . Despite the previous study indicating that decorin induces autophagy signaling in different cell types36,37, its precise role play in ADSC phenotype during inflammation is unclear.

In this study, we intend to evaluate the introduction of ADSCs within decorin-loaded gellan gum hydrogel on articular therapy in inflammatory joint disorder. This formulation not only aimed at improving the ADSCs regenerative function at the articular cartilage but also took advantage of the immunomodulatory properties of decorin. The immuno-modulation role of decorin functions not only at ADSCs in the inflammatory microenvironment but also in the chondrocytes via a sustained release. Our results here may represent an easy and effective route to improve the anti-inflammatory capacity of ADSCs for intra-articular injection therapy.

Materials and Methods

Isolation of Mouse ADSCs

Isolation of ADSCs was performed according to previous reports38,39. Briefly, the inguinal subcutaneous fat was collected from C57BL/6 mice (male, 10 weeks old). The adipose was minced into pieces less than 1 mm in diameter. Digestion was performed with 0.1% type I collagenase (Gibco, United States) at 37°C for 45 min. After neutralization, the collected cells were seeded to a final concentration of 5 × 106 cells/ml. The cells were cultured in a 37°C incubator supplied with 5% CO2 and 95% humidity. The third- or fourth-passage cells were used for the following experiments. Characterization of the ADSCs through flow cytometry and differentiation assay has been performed according to our previous study 39 .

Decorin/Gellan Gum Hydrogel Preparation and ADSCs Encapsulation

Decorin/gellan gum hydrogel was performed according to previous report 27 with slight modification. Briefly, gellan powder was dissolved in deionized water for 1% (w/v) solution with heating to 70°C. After dissolving, gellan sol was added to the rotational rheometer (Fig. 1A, Brookfield, United States) and cooled to 50°C. Decorin from bovine articular cartilage (Sigma, Germany) and sodium chloride (0.2 M) were added to gellan sol for final concentrations of 0.9% (w/v) gellan, 0.24 mg/ml Decorin, and 10 mM sodium chloride (NaCl). Finally, the whole mixture was cooled at a rate of 1°C/min to a final temperature of 24°C under shear (450/s). In the case of a hydrogel loaded with ADSCs, suspension of ADSCs at a final density of 1 × 106 cells/ml was mixed into the mixture under shear (100/s) in 1 min followed by further experiments.

Figure 1.

Figure 1.

Decorin-laden gellan gum hydrogel production and physiochemical property analysis of the decorin/gellan gum/ADSCs hydrogel. (A) Representative figure of the rotational rheometer used in this study. Viscosity characterization of the decorin/gellan hydrogel by applying shear (n = 2). (B) Optical images of the post-shearing hydrogel with syringe injection. (C) Cumulative release curve of the decorin-enriched hydrogel over 180 min. (D) Scanning electron microscopy (SEM) characterization image of the decorin/gellan/ADSCs hydrogel illustrating the forming gel structure. Scare bars: 100 μm. (E) Representative image of 4′,6-diamidino-2-phenylindole (DAPI) staining identifies ADSCs encapsulation in hydrogel on day 1. (F) Swelling degree test of three groups of hydrogels (n = 3). ADSCs: adipose-derived stem cells.

Scanning Electron Microscopy Analysis

Solid form ADSCs/decorin/gellan gum hydrogel was immersed in 2.5% glutaraldehyde for fixation for about 1 h. Dehydration was performed with graded ethanol (50%, 70%, 80%, 90%, 95%, and 100%) and lyophilized for 12 h to eliminate residual water. All samples were sputter-coated with platinum and imaged with an ULTRA 55 scanning electron microscope (SEM; Carl ZEISS AG, Germany).

In Vitro Decorin Release

Cumulatively release levels of decorin from the hydrogel were determined according to a previous study 27 . A total of 1 ml of the solid decorin/gellan gum hydrogel was soaked in 2 ml of Dulbecco’s Modified Eagle Medium (DMEM) and incubated at 37°C. At a certain time point, the medium was removed for measurement by enzyme-linked immunosorbent assay (ELISA). Decorin cumulatively released was quantified using Bovine Decorin ELISA (Sigma, RAB1185, Germany) following the manufacturer’s protocol.

Swelling Assay

The swelling behavior of hydrogels was performed as described before 40 . Briefly, 200 mg (M0) of lyophilized hydrogel was soaked in 20 ml of normal saline solution. At every time point, the hydrogels were removed and immediately weighed (M1). The swelling behavior was calculated as Swelling (%) =((M1M0)/M0)×100 .

Live/Dead Assay

A Live/Dead cell imaging kit (Invitrogen, R37601) was used to determine the effect of IL-β (20 ng/mL) with/without decorin on ADSC viability according to the manufacturer’s protocol. Images were taken with a fluorescence microscope (Leica, Germany) at ex 488 nm and ex 570 nm.

Cell Proliferation Assay Under Inflammation

The ADSCs were seeded onto the hydrogels at the density of 1 × 106 cells/ml. Then IL-1β (20 ng/ml) was added to each well and incubated for 24 h. Cell proliferation assay was evaluated using Cell Counting Kit-8 (CCK-8, Sigma-Aldrich) according to the manufacturer’s instructions.

Co-culture System

The Transwell co-culture system was performed as described before 41 . A number of 5 × 104 primary chondrocytes were seeded into a 24-well plate. Cells were incubated with/without 20 ng/ml IL-1β for 12 h. After induction of inflammation, ADSCs/decorin/gellan gum hydrogel was added into transwell inserts for co-culture with inflamed chondrocytes for another 12 h. ADSCs or chondrocytes were collected and used in the following Quantitative Real-time Polymerase Chain Reaction (qRT-PCR) analysis.

Quantitative Real-time Polymerase Chain Reaction

Total RNA from ADSCs or chondrocytes was extracted using TRIzol (Invitrogen, Germany). The qRT-PCR assay was performed using the ABsolute Real Time PCR SYBR Master Mix (Invitrogen, AB1162B) on a 7000 PCR System according to the manufacturer’s instructions. The fold change in the expression of the sample was calculated using the ΔΔCt comparative method. Primers used in this study are listed in Table 1.

Table 1.

Mus Musculus Primers Used in This Study.

GENE Forward Reverse
SOX9 AGTACCCGCATCTGCACAAC ACGAAGGGTCTCTTCTCGCT
Col2a1 GGGAATGTCCTCTGCGATGAC GAAGGGGATCTCGGGGTTG
Aggrecan CCTGCTACTTCATCGACCCC AGATGCTGTTGACTCGAACCT
Runx2 GACTGTGGTTACCGTCATGGC ACTTGGTTTTTCATAACAGCGGA
MMP-13 CTTCTTCTTGTTGAGCTGGACTC CTGTGGAGGTCACTGTAGACT
ADAMTS5 GGAGCGAGGCCATTTACAAC CGTAGACAAGGTAGCCCACTTT
TNF-α GGTGCCTATGTCTCAGCCTCTT GCCATAGAACTGATGAGAGGGAG
IL-6 TAGTCCTTCCTACCCCAATTTCC TTGGTCCTTAGCCACTCCTTC
GAPDH AGGTCGGTGTGAACGGATTTG TGTAGACCATGTAGTTGAGGTCA
β-Actin ACTCTTCCAGCCTTCCTTC ATCTCCTTCTGCATCCTGTC

ADAMTS5: ADAM metallopeptidase with thrombospondin type I Motif 5; Col2a1: collagen type II alpha 1 chain; IL-6: interleukin-6; MMP-13: matrix metalloproteinase 13; Runx2: Runt-related transcription factor 2; SOX9: SRY-box transcription factor 9; TNF-α: tumor necrosis factor-alpha; GAPDH: Glycerinaldehyd-3-phosphat-Dehydrogenase.

Western Blotting

ADSCs were harvested and homogenized in Radioimmunoprecipitation assay (RIPA) lysis buffer. Protein concentration was determined using a bicinchoninic acid (BCA) assay (Invitrogen, Germany). After gel running, samples were blocked with 5% skim milk and incubated with primary antibodies at 4°C overnight: LC3A/B (CST, 1:1,000, 4108), p62 (CST, 1:1,000, 23214), and β-actin (CST, 1:1,000, 3700). Samples were rinsed three times with PBST and incubated with indicated secondary antibodies for 2 h at room temperature. Samples were developed with Immobilon Western Chemilum HRP Substrate (Millipore, WBKLS0500) for 1–2 min. The band density for each sample was determined relative to β-actin.

Animal Experiments

The mono-iodoacetate (MIA)-induced OA animal study was conducted under the guideline of the National Institutes of Health Guide for the Care and Use of Laboratory Animals and ARRIVE guidelines. A total of 38 adult male C57BL/6 mice (10 weeks old) were randomly divided into the following groups: (1) SHAM = 3; (2) SHAM + gellan/deocrin/ADSCs hydrogel = 3; (3) MIA = 6; (4) MIA + gellan gum hydrogel = 6; (5) MIA + gellan/decorin hydrogel = 6; (6) MIA + gellan/ADSCs hydrogel = 6; and (7) MIA + gellan/decorin/ADSCs hydrogel = 8. PBS intra-articular injection serves as the SHAM group. The MIA OA model was generated according to the previous report with slight modification 42 . Briefly, under anesthesia, an intra-articular injection of 0.25 mg of MIA was performed into the left knee. In the treatment groups, OA induction was followed by their respective treatments: Intra-articular injection of hydrogel for once on 7 days post-MIA injection. Tissue was collected at 14 days post-MIA injection.

Masson’s Trichrome Stain Assessment, Imaging, and OARSI Scoring

The collected specimens of the knee joint were fixed in 4% paraformaldehyde, decalcified in 10% EDTA decalcification solution for 2 weeks, and embedded in paraffin. The specimens were sectioned at 5 μm thickness in the sagittal plane and prepared for Masson’s trichrome stain analysis. Sections were hydrated to distilled water and mordant in Bouins’s solution for 5 min. After washing three times, they were kept in modified Weigert’s iron hematoxylin for 5 min. Then Biebrich Scarlet-Acid Fuchsin solution was used for 5 min, followed by 1% phosphomolybdic acid solution for 3 min and Aniline Blue solution for 10 min. Sections were imaged on a Zeiss Axioscanner fluorescent microscope. The Osteoarthritis Research Society International (OARSI) scoring was performed as a recommended scoring system for mice 43 .

Cytokines Evaluations

The synovial membranes were collected and lysed. The ELISA kits (MTA00B, MLB00C, D6050, R&D, United States) were employed to test the cytokines (TNF-α, IL-1β, and IL-6) in synovium samples according to the manufacturer’s instructions.

Statistical Analysis

All statistical analysis was performed using GraphPad Prism version 7.0 (GraphPad Software, La Jolla, CA, United States). Statistical significance was determined at p < 0.05. Statistical differences between the two groups were using Student’s t-test. Statistical differences among groups were analyzed using one-way variance with Tukey’s post hoc tests. For the data that were not normally distributed, a non-parametric Mann–Whitney t-test was used.

Results

Decorin-Laden Gellan Gum Hydrogel Formulation and ADSCs Encapsulation Properties

First, we studied the viscosity–shear rate profiles of decorin-laden hydrogel. As shown in Fig. 1A, time-dependent viscosity profiles highlight the degree of thixotropy of decorin-laden hydrogel. The viscosities of hydrogel were around 10,000 mPa·s at a shear rate of 1/s. Increasing the shear force exerted on the hydrogel results in shear-thinning behavior, indicating that the post-sheared hydrogel exhibited typical liquid-like property which is suitable for applying syringe (27G) injection (Fig. 1B). After hydrogel formulation, we evaluated the rate of decorin release by determining the cumulative release curve (Fig. 1C). Release of decorin from the hydrogel was calculated approximately linear over time (y = 0.0752+0.103x−0.0001x2, R2 = 0.9851). After shearing and cooling, “rope-like” gelled entities were synthesized and were analyzed by SEM image (Fig. 1D). As shown in Fig. 1E, DAPI staining in hydrogel on day 1 could observe encapsulated ADSCs in decorin/gellan gum hydrogel. We further found that the swelling ratio of the hydrogel declined with the encapsulation of ADSCs (Fig. 1F). Proper hydrogel formulation could ameliorate the survival of ADSCs and preserve functional property. Taken together, these results indicated that ADSCs could be successfully and effectively encapsulated into decorin/gellan gum hydrogel.

Biocompatibility and Chondrogenesis Regulation of ADSCs by Decorin-Laden Hydrogel Under Inflammatory Stress

ADSCs encapsulated in different hydrogels can still proliferate and differentiate into cartilage cells. To evaluate the biocompatibility of ADSCs in the deocrin/gellan gum hydrogel under inflammatory stress, IL-1β (20 ng/mL) was treated with or without decorin loading. Live/dead cell staining analysis indicated ADSCs encapsulated in decorin-laden hydrogel after culture for 7 days displayed a high rate of cell viability under IL-1β stimulation when compared with ADSCs encapsulated in gellan hydrogel (Fig. 2A, B). Of note, the cell viabilities of ADSCs encapsulated in decorin/gellan gum hydrogel showed significant differences with increased IL-1β concentration (from 10 to 40 ng/ml) while compared with non-decorin loading (Fig. 2C), suggesting a protection role of decorin against inflammation-induced cell death on ADSCs.

Figure 2.

Figure 2.

ADSCs in vitro biocompatibility evaluation in the decorin/gellan gum hydrogel with IL-1β. (A, B) Representative images of live/dead images of ADSCs cultured in the decorin/gellan gum hydrogel on days 1 and 7 with IL-1β (20 ng/mL) stimulation (n = 3). Scare bars: 200 μm. (C) CCK-8 assay identified ADSCs viabilities encapsulated in the decorin/gellan gum hydrogel with increased IL-1β concentration (from 10 to 40 ng/ml, n = 3). ADSCs = adipose-derived stem cells; CCK-8 = Cell Counting Kit-8; IL-1β = interleukin-1β.

We then compared the ability of decorin-laden hydrogels to regulate the chondrogenic differentiation of ADSCs under inflammatory stress by measuring the expression of chondrogenic-related genes, including SRY-box transcription factor 9 (SOX9), collagen type II alpha 1 chain (Col2a1), aggrecan (ACAN), and Runt-related transcription factor 2 (Runx2). After in vitro chondrogenic differentiation for 14 days, the mRNA expression levels of SOX9, Col2a1, and ACAN significantly increased in ADSCs encapsulated decorin hydrogel under IL-1β stimulation, while the expression of the OA-related gene Runx2 showed significant decreased (Fig. 3). These results suggested that the decorin/gellan gum hydrogel could protect the chondrogenic differentiation ability of ADSCs in the inflammation microenvironment.

Figure 3.

Figure 3.

The chondrogenic differentiation analysis of ADSCs under inflammatory stress. Quantitative real-time PCR analysis of the expression of SOX9, Col2a1, ACAN, and Runx2 in ADSCs encapsulated in the decorin/gellan gum hydrogel with or without IL-1β stimulation (n = 4). ADSCs = adipose-derived stem cells; IL-1β = interleukin-1β.

Decorin/Gellan Hydrogel Modulate Autophagy Signaling in ADSCs Under Inflammatory Stress

Decorin was known to induce autophagy signaling in different cell types 44 . During autophagy, cytoplasmic LC3 protein (LC3-I) is conjugated to phosphatidylethanolamine for lipidation to form LC3-phosphatidylethanolamine conjugate (LC3-II) and then recruited to the autophagosome membranes45,46. The ubiquitin-associated protein p62 binds to LC3 and entraps in autophagosome; therefore, decreased expression of p62 is commonly considered upregulated autophagy signaling. IL-1β-induced inflammation interferes with autophagy signaling in ADSCs, as indicated by upregulated LC3-II and p62 protein expression (Fig. 4). Of note, enhanced p62 expression in acute inflammation via impaired proteasomal degradation is associated with IL-1β production 47 . Importantly, decorin-loaded hydrogels restored the autophagy signaling in ADSCs, as suggested by upregulated p62 protein expression (Fig. 4) when compared with IL-1β treatment. A previous study has shown that 200 nM decorin stimulation on endothelial cells for 12 h effectively activates the LC3-I/LC3-II conversion 48 and increases p62 degradation 49 . However, loading with gellan gum hydrogel only did not show a significant change in p62 protein expression. These above results proved that the decorin/gellan gum hydrogel could attenuate the autophagy destruction in ADSCs with IL-1β-induced inflammation challenges.

Figure 4.

Figure 4.

Decorin/gellan gum hydrogel regulates autophagy signaling in ADSCs under IL-1β stress. WB analysis of the expression of autophagy-related genes LC3-I, LC3-II, and p62 in ADSCs encapsulated in the decorin/gellan gum hydrogel with or without IL-1β stimulation (n = 3). ADSCs = adipose-derived stem cells; IL-1β = interleukin-1β.

In Vitro Chondrocytes Protection and In Vivo Cartilage Repair Assays of Decorin/Gellan/ADSCs Hydrogel

To determine the protected effect of decorin/gellan/ADSCs hydrogel on chondrocytes, a co-culture transwell system was used. IL-1β stimulation significantly induced mRNA expression levels of inflammatory genes in chondrocytes, including matrix metalloproteinase 13 (MMP-13), ADAM metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS5), TNF-α, and IL-6 (Fig. 5). However, placement of decorin/gellan/ADSCs hydrogel in upper transwell chamber decreased the mRNA expression levels of OA-related genes, indicating that decorin/gellan/ADSCs hydrogel may provide protection on chondrocytes through cytokines paracrine signaling or decorin released.

Figure 5.

Figure 5.

In vitro evaluation of chondrocytes protection in a co-culture system. The qRT-PCR of the expression of OA-related genes MMP-13, ADAMTS5, TNF-α, and IL-6 in chondrocytes with or without the decorin/gellan/ADSCs hydrogel treatment under IL-1β stimulation (n = 4). ADSCs = adipose-derived stem cells; IL-1β = interleukin-1β; OA = osteoarthritis; TNF-α = tumor necrosis factor-alpha.

After comparatively analyzing the ability to provide chondrocyte protection in vitro, we tested the ability of the hydrogel to attenuate cartilage injury in vivo. Hydrogels loaded with different components were injected into mice knee joint with MIA-induced cartilage damage (Fig. 6A). Masson’s trichrome staining showed that apparent cartilage damage in the MIA group and MIA + gellan gum group (MIA + G), as reflected by GAG denaturation on the cartilage surface and collagen matrix disorganization (red staining). As expected, injection of gellan/decorin hydrogel (MIA + G/D) slightly suppressed cartilage degeneration in the MIA model, while injection of gellan/ADSCs hydrogel (MIA + G/A) significantly decreased cartilage injury, as shown by improvement of the structural integrity in cartilage. Interestingly, a synergistic therapeutic effect of decorin and ADSCs was displayed by better protection on tibial cartilages injury (Fig. 6A). In line with this result, analysis of the OARSI score showed that the decorin/gellan gum/ADSCs hydrogel injection group displayed significantly lower scores than gellan gum/ADSCs hydrogel injection group (Fig. 6B). Furthermore, MIA-induced inflammatory factor (TNF-α, IL-1β, and IL-6) upregulation in the synovial membrane was significantly attenuated by intra-articular application of decorin/gellan gum/ADSCs hydrogel (Fig. 6C). Taken together, the above results indicated that the decorin/gellan gum/ADSCs hydrogel presented better in vivo cartilage protection ability in inflammatory arthritis when compared with intra-articular injection of decorin/gellan gum/ hydrogel or ADSCs/gellan gum/ hydrogel.

Figure 6.

Figure 6.

In vivo evaluation of cartilage protection by the decorin/gellan gum/ADSCs hydrogel in the MIA models. (A) Representative Masson’s trichrome staining images of mouse cartilage injuries in different hydrogel groups. (B) OARSI histological assessment score in different hydrogel groups. (C) The ELISA assay was employed to test the cytokines TNF-α, IL-1β, and IL-6 in synovium membranes from different hydrogel groups (G, gellan gum; D, decorin; A, ADSCs). ADSCs = adipose-derived stem cells; ELISA = enzyme-linked immunosorbent assay; IL-1β = interleukin-1β; MIA = mono-iodoacetate; TNF-α = tumor necrosis factor-alpha.

Discussion

This study aimed to determine the regulated effect of decorin-rich gellan gum hydrogel on ADSC regenerative phenotype under the inflammatory microenvironment. Our results demonstrate that encapsulation ADSCs within a decorin-enriched gellan gum not only regulated their proliferation activity under inflammation but also supported their chondrogenesis in favor of cartilage regeneration. The underlying mechanism may be related to the regulation of ADSCs autophagy signaling in the inflammatory microenvironment. Of note, a synergistic effect of ADSCs on articular therapy in inflammatory joint disorder was achieved by combining with decorin-loaded gellan hydrogel, suggesting the regenerative potential of ADSCs/decorin/gellan hydrogel for cartilage injury repair in preclinical medicine (Fig. 7).

Figure 7.

Figure 7.

Schematic diagram of the injectable ADSCs-loaded decorin/gellan gum hydrogel for arthritis inflammatory microenvironment regulation. Integration of ADSCs and decorin in hydrogel results in cartilage ECM biomimetic and anti-inflammatory properties. ADSCs: adipose-derived stem cells; ECM: extracellular matrix.

ADSCs are multipotent and able to differentiate into chondrocytes. Arthroscopy injection of ADSCs improved cartilage regeneration and reduced pain in patients with OA. The efficacy of articular cartilage regeneration by MSCs-hydrogel composite was demonstrated by clinical trial 50 . Ideal hydrogel matrix should be injectable for intra-articular implantation 51 , biomimetic for maintaining stem cell phenotype for chondrogenesis 52 , and particularly immunomodulated for inflammation attenuation 53 . Naturally derived polymers, including gellan gum, can form hydrogels for MSCs encapsulation 54 . Gellan gum hydrogel eye drop formulation was first developed by Hill et al 27 . Our results confirmed the transition between solid and liquid states of gellan gum hydrogel, allowing loading of ADSCs followed by needle injection. We believe that the resting solid state of gellan gum hydrogel after intra-articular injection is beneficial not only for stem cell retention but also for application for three-dimensional tissue bioprinting manufacturing.

Three-dimensional culture of MSCs in the ECM motifs-modified gellan gum hydrogel presented higher proliferation and metabolic secrete factors when compared with commercially available hydrogel 55 , indicating the importance of mimicking the ECM microenvironment for MSCs to maintain inflammation attenuation phenotype 53 . Interestingly, trehalose-assisted gellan gum encapsulation of ADSC retained their differentiation phenotype under cryopreservation stress 56 . The outcomes of our study confirmed that decorin supported the chondrogenesis of ADSCs in inflammatory circumstances. The decreased proliferation rate induced by IL-1β was also retained by decorin-assisted gellan gum hydrogel. A similar study on BMSCs pointed out that stiffness varying can form a suitable network for cartilage differentiation of BMSCs 57 . There is evidence that ADSCs are more likely to differentiate into cartilage than BMSCs; therefore, optimization of hydrogel matrix stiffness favors for MSC chondrogenic differentiation was practicable 58 . Considering decorin was necessary for maintaining fibril structure and mechanical stiffness of tendon and cartilage59,60, we assumed that application of decorin in gellan gum could serve as viscous substrates to modulate stiffness in a microenvironment, which preserves ADSCs regenerative potential under inflammation.

On the other hand, decorin is known for its ability to adhere to the aggrecan network and mediate biomechanical transduction of cartilage 61 , which explains that decorin delays the loss of sulfated GAGs in chondrocyte induced by IL-1β stimulation 20 . Here, we demonstrated that decorin-loaded gellan gum hydrogel increases the aggrecan expression in ADSCs with IL-1β activation, indicating that decorin regulates the integrity of the aggrecan network in the pericellular microenvironment 62 . Therefore, the chondromimetic matrix formulated by decorin gellan hydrogel may control the chondrogenic differentiation of ADSCs which is in line with other data 63 . In addition, the decorin-based biomimetic scaffold has been shown suitable for promoting chondrogenic differentiation of ADSCs 64 , suggesting the crucial role decorin plays in assembling chondromimetic hydrogel for cartilage regeneration. Taken together, combination of ADSCs and the extracellular decorin matrix may favor better participation in suppressing cartilage injury.

A previous study showed the possibility of the immuno-modulation capacity of decorin 65 . Not surprisingly, IL-1β stimulation induces decorin production in human stromal cells 66 , indicating that decorin acts as a feedback regulation under the inflammatory microenvironment. Interestingly, decorin boosts pro-inflammatory signaling via Toll-like receptors in macrophages 67 . However, Toll-like receptors are not dominantly expressed in ADSCs, suggesting that a different immunomodulatory role may decorin play in the ADSCs. Topical decorin reduces the infiltration of inflammatory macrophages and neutrophils in corneal inflammation68,69, indicating the inflammation attenuating aspect of decorin on other cell types. Moreover, decorin overexpressing MSCs regulate inflammation in injuries via increasing the expression of anti-inflammatory cytokines 26 . Our results showed a synergistic effect of decorin and ADSCs in inflammatory joint disorder; therefore, we assumed that decorin in the matrix could modulate ADSCs immunol phenotype to enhance the anti-inflammatory effect via cytokines paracrine action on chondrocytes. In line with these data, a co-culture-secreted protein study of MSCs and cartilage explants showed a protective effect of MSCs on IL-1β induced cartilage catabolic injury by increasing the decorin secretion 24 .

ADSCs increase the autophagy flux in response to stress stimulations 70 , probably via the Sirtuin-1-related autophagy pathway to attenuate oxidative stress-induced cell death 71 . Enhancing autophagy signaling in stress-challenged ADSCs plays a protective role in cell survival 72 . In addition, enhanced autophagy of ADSCs improves its stemness 73 and therefore promotes therapeutic effect under inflammation 74 . Our results showed that decorin-laden hydrogel may increase autophagy signaling in ADSCs under IL-1β challenging. Previous studies revealed that proteoglycan could regulate the autophagy pathway in endothelial cells via outside-in signaling31,75. Recent studies have shown that decorin can also induce autophagy and mitophagy. Decorin directly interacts with vascular endothelial growth factor receptor 2 to induce autophagy 76 . We assumed that decorin-laden hydrogel may increase autophagy signaling in ADSCs via interacting with receptor tyrosine kinases under an inflammatory microenvironment. In fact, preconditioning induction of autophagy in ADSCs alters the immune phenotype and increases the expression of cytokines 77 . Therefore, regulating the inflammatory “fingerprint” of ADSC by decorin may upregulate its anti-inflammatory potential. Interestingly, ADSCs itself could activate autophagy in chondrocytes for attenuating catabolism in an inflammation microenvironment 78 , suggesting that released decorin may have a direct therapeutic effect via contacting chondrocytes.

There are some limitations in this study: (i) Comparison of the immune phenotype of the different magnitude of cell numbers in hydrogels could enhance the novelty of this study; (ii) using human-driven cells or decorin would complement the translational potential of this study; (iii) the underlying mechanism of decorin regulate autophagy in ADSCs is unknown; and (iv) tracking the distribution of ADSCs after intra-articular injection of the hydrogel would strengthen the research depth of this study. However, we could not underestimate the scientific novelty of this study which is the synergistic therapeutic effect of decorin/gellan gum/ADSCs hydrogel in favor of cartilage protection in inflammatory arthritis.

In summary, decorin/gellan gum hydrogel enhances the anti-inflammatory properties of encapsulating ADSCs. The regulating mechanism may be complemented by the ability of decorin to induce autophagy signaling and therefore modulates the immune response of ADSC in the inflammation microenvironment. Further research aimed at exploring a promising intra-articular injection candidate for the anti-inflammatory drug of decorin/gellan gum/ADSCs hydrogel in inflammatory joint disorder is warranted.

Acknowledgments

The authors thank the technician colleagues for their help in the study. The schematic diagram figure was drawn by Figdraw (https://www.figdraw.com/static/index.html).

Footnotes

Author Contributions: Conceptualization, W.H. and K.W.; methodology, W.H., Y.W., Z.L., G.Y., X.C., J.R., S.J., and K.W.; validation, W.H., Y.W., Z.L., G.Y., T.L., Z.L., and S.J.; formal analysis, W.H. and Y.W.; investigation, W.H., Y.W., Z.L., G.Y., W.Y., X.C., J.R., T.L., Z.L., S.J., and W.K.; resources, W.H., S.J., and K.W.; data curation, W.H., Y.W., W.Y., J.R., T.L., Z.L., S.J., and K.W.; writing—original draft preparation, W.H., Y.W., and S.J.; writing—review and editing, W.H., Z.L., S.J., and K.W.; supervision, W.H. and K.W.; project administration, W.H., S.J., and K.W.; funding acquisition, W.H. and K.W. All authors have read and agreed to the published version of the manuscript.

Availability of data and material: Data are available within the paper and on request from the corresponding authors.

Ethical Approval: This study was reviewed by the Dongguan Hospital of Integrated Traditional Chinese and Western Medicine Ethics and Institutional Review Committee, Dongguan, China.

Statement of Human and Animal Rights: All of the procedures involving animals were conducted in accordance with the Institutional Animal Care guidelines of Dongguan Hospital of Integrated Traditional Chinese and Western Medicine, Dongguan, China (2020-0112). This article does not contain any studies with human subjects.

Statement of Informed Consent: There are no human subjects in this article and informed consent is not applicable.

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was funded by the Medical Scientific Research Foundation of Guangdong Province, China (A2020393) and the Dongguan Science and Technology of Social Development Program (20221800906152 and 20211800904092). This research was partly supported by the Guangzhou Key Research and Development Project (No. 2023B03J0211) and the Science and Technology Planning Project of Guangdong Province (No. 2023A1515012621).

References

  • 1. Veronesi F, Maglio M, Tschon M, Aldini NN, Fini M. Adipose-derived mesenchymal stem cells for cartilage tissue engineering: state-of-the-art in in vivo studies. J Biomed Mater Res A. 2014;102(7):2448–66. [DOI] [PubMed] [Google Scholar]
  • 2. Fodor PB, Paulseth SG. Adipose derived stromal cell (ADSC) injections for pain management of osteoarthritis in the human knee joint. Aesthet Surg J. 2016;36(2):229–36. [DOI] [PubMed] [Google Scholar]
  • 3. Jo CH, Lee YG, Shin WH, Kim H, Chai JW, Jeong EC, Kim JE, Shim H, Shin JS, Shin IS, Ra JC, et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells. 2014;32(5):1254–66. [DOI] [PubMed] [Google Scholar]
  • 4. Delanois RE, Etcheson JI, Sodhi N, Henn RF, 3rd, Gwam CU, George NE, Mont MA. Biologic therapies for the treatment of knee osteoarthritis. J Arthroplasty. 2019;34(4):801–13. [DOI] [PubMed] [Google Scholar]
  • 5. Kuwasawa A, Okazaki K, Noda K, Fukushima T, Nihei K. Intra-articular injection of culture-expanded adipose tissue-derived stem cells for knee osteoarthritis: assessments with clinical symptoms and quantitative measurements of articular cartilage volume. J Orthop Sci. 2023;28(2):408–15. [DOI] [PubMed] [Google Scholar]
  • 6. Djouad F, Bouffi C, Ghannam S, Noël D, Jorgensen C. Mesenchymal stem cells: innovative therapeutic tools for rheumatic diseases. Nat Rev Rheumatol. 2009;5(7):392–99. [DOI] [PubMed] [Google Scholar]
  • 7. Ueyama H, Okano T, Orita K, Mamoto K, Ii M, Sobajima S, Iwaguro H, Nakamura H. Local transplantation of adipose-derived stem cells has a significant therapeutic effect in a mouse model of rheumatoid arthritis. Sci Rep. 2020;10(1):3076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Tossetta G, Fantone S, Licini C, Marzioni D, Mattioli-Belmonte M. The multifaced role of HtrA1 in the development of joint and skeletal disorders. Bone. 2022;157:116350. [DOI] [PubMed] [Google Scholar]
  • 9. Udomsinprasert W, Jittikoon J, Honsawek S. Interleukin-34 as a promising clinical biomarker and therapeutic target for inflammatory arthritis. Cytokine Growth Factor Rev. 2019;47:43–53. [DOI] [PubMed] [Google Scholar]
  • 10. Bora P, Majumdar AS. Adipose tissue-derived stromal vascular fraction in regenerative medicine: a brief review on biology and translation. Stem Cell Res Ther. 2017;8(1):145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Dubey NK, Wei HJ, Yu SH, Williams DF, Wang JR, Deng YH, Tsai FC, Wang PD, Deng WP. Adipose-derived stem cells attenuates diabetic osteoarthritis via inhibition of glycation-mediated inflammatory cascade. Aging Dis. 2019;10(3):483–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Chang TH, Wu CS, Chiou SH, Chang CH, Liao HJ. Adipose-derived stem cell exosomes as a novel anti-inflammatory agent and the current therapeutic targets for rheumatoid arthritis. Biomedicines. 2022;10(7):1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Cavallo C, Merli G, Zini N, D’Adamo S, Cattini L, Guescini M, Grigolo B, Di Martino A, Santi S, Borzi RM, Filardo G. Small extracellular vesicles from inflamed adipose derived stromal cells enhance the NF-kappaB-dependent inflammatory/catabolic environment of osteoarthritis. Stem Cells Int. 2022;2022:9376338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Skalska U, Kontny E, Prochorec-Sobieszek M, Maśliński W. Intra-articular adipose-derived mesenchymal stem cells from rheumatoid arthritis patients maintain the function of chondrogenic differentiation. Rheumatology (Oxford). 2012;51(10):1757–64. [DOI] [PubMed] [Google Scholar]
  • 15. Djouad F, Delorme B, Maurice M, Bony C, Apparailly F, Louis-Plence P, Canovas F, Charbord P, Noël D, Jorgensen C. Microenvironmental changes during differentiation of mesenchymal stem cells towards chondrocytes. Arthritis Res Ther. 2007;9(2):R33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Wilda M, Bächner D, Just W, Geerkens C, Kraus P, Vogel W, Hameister H. A comparison of the expression pattern of five genes of the family of small leucine-rich proteoglycans during mouse development. J Bone Miner Res. 2000;15(11):2187–96. [DOI] [PubMed] [Google Scholar]
  • 17. Salinas CN, Anseth KS. Decorin moieties tethered into PEG networks induce chondrogenesis of human mesenchymal stem cells. J Biomed Mater Res A. 2009;90(2):456–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Lewis JL, Krawczak DA, Oegema TR, Jr, Westendorf JJ. Effect of decorin and dermatan sulfate on the mechanical properties of a neocartilage. Connect Tissue Res. 2010;51(2):159–70. [DOI] [PubMed] [Google Scholar]
  • 19. Wilusz RE, Sanchez-Adams J, Guilak F. The structure and function of the pericellular matrix of articular cartilage. Matrix Biol. 2014;39:25–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Li Q, Han B, Wang C, Tong W, Wei Y, Tseng WJ, Han LH, Liu XS, Enomoto-Iwamoto M, Mauck RL, Qin L, et al. Mediation of cartilage matrix degeneration and fibrillation by decorin in post-traumatic osteoarthritis. Arthritis Rheumatol. 2020;72(8):1266–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Bock HC, Michaeli P, Bode C, Schultz W, Kresse H, Herken R, Miosge N. The small proteoglycans decorin and biglycan in human articular cartilage of late-stage osteoarthritis. Osteoarthritis Cartilage. 2001;9(7):654–63. [DOI] [PubMed] [Google Scholar]
  • 22. Polgar A, Falus A, Koo E, Ujfalussy I, Sesztak M, Szuts I, Konrad K, Hodinka L, Bene E, Meszaros G, Ortutay Z, et al. Elevated levels of synovial fluid antibodies reactive with the small proteoglycans biglycan and decorin in patients with rheumatoid arthritis or other joint diseases. Rheumatology (Oxford). 2003;42(4):522–27. [DOI] [PubMed] [Google Scholar]
  • 23. Kydd AS, Reno CR, Tsao HW, Hart DA. Early inflammatory arthritis in the rabbit: the influence of intraarticular and systemic corticosteroids on mRNA levels in connective tissues of the knee. J Rheumatol. 2007;34(1):130–39. [PubMed] [Google Scholar]
  • 24. Dechêne L, Colin M, Demazy C, Fransolet M, Niesten A, Arnould T, Serteyn D, Dieu M, Renard P. Characterization of the proteins secreted by equine muscle-derived mesenchymal stem cells exposed to cartilage explants in osteoarthritis model. Stem Cell Rev Rep. 2023;19(2):550–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Dong Y, Zhong J, Dong L. The role of decorin in autoimmune and inflammatory diseases. J Immunol Res. 2022;2022:1283383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Liu D, Kong F, Yuan Y, Seth P, Xu W, Wang H, Xiao F, Wang L, Zhang Q, Yang Y, Wang H. Decorin-modified umbilical cord mesenchymal stem cells (MSCs) attenuate radiation-induced lung injuries via regulating inflammation, fibrotic factors, and immune responses. Int J Radiat Oncol Biol Phys. 2018;101(4):945–56. [DOI] [PubMed] [Google Scholar]
  • 27. Hill LJ, Moakes RJA, Vareechon C, Butt G, Ng A, Brock K, Chouhan G, Vincent RC, Abbondante S, Williams RL, Barnes NM, et al. Sustained release of decorin to the surface of the eye enables scarless corneal regeneration. NPJ Regen Med. 2018;3:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Chouhan G, Moakes RJA, Esmaeili M, Hill LJ, deCogan F, Hardwicke J, Rauz S, Logan A, Grover LM. A self-healing hydrogel eye drop for the sustained delivery of decorin to prevent corneal scarring. Biomaterials. 2019;210:41–50. [DOI] [PubMed] [Google Scholar]
  • 29. Xing J, Peng X, Li A, Chen M, Ding Y, Xu X, Yu P, Xie J, Li J. Gellan gum/alginate-based Ca-enriched acellular bilayer hydrogel with robust interface bonding for effective osteochondral repair. Carbohydr Polym. 2021;270:118382. [DOI] [PubMed] [Google Scholar]
  • 30. Wu M, Downie LE, Hill LJ, Chinnery HR. The effect of topical decorin on temporal changes to corneal immune cells after epithelial abrasion. J Neuroinflammation. 2022;19(1):90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Gubbiotti MA, Iozzo RV. Proteoglycans regulate autophagy via outside-in signaling: an emerging new concept. Matrix Biol. 2015;48:6–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Lin Z, Miao J, Zhang T, He M, Wang Z, Feng X, Bai L. JUNB-FBXO21-ERK axis promotes cartilage degeneration in osteoarthritis by inhibiting autophagy. Aging Cell. 2021;20(2):e13306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Li C, Pan J, Ye L, Xu H, Wang B, Xu H, Xu L, Hou T, Zhang D. Autophagy regulates the therapeutic potential of adipose-derived stem cells in LPS-induced pulmonary microvascular barrier damage. Cell Death Dis. 2019;10(11):804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Hu Y, Shao J, Shen L, Wang S, Xu K, Mao J, Shen J, Chen W. Protection of adipose-derived mesenchymal stromal cells during acute lung injury requires autophagy maintained by mTOR. Cell Death Discov. 2022;8(1):481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Li Q, Yin Y, Zheng Y, Chen F, Jin P. Inhibition of autophagy promoted high glucose/ROS-mediated apoptosis in ADSCs. Stem Cell Res Ther. 2018;9(1):289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Neill T, Chen CG, Buraschi S, Iozzo RV. Catabolic degradation of endothelial VEGFA via autophagy. J Biol Chem. 2020;295(18):6064–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Zhang TW, Li ZF, Ding W, Wang HR, Ding SL, Han GJ, Li XL, Dong J, Jiang LB. Decorin inhibits nucleus pulposus apoptosis by matrix-induced autophagy via the mTOR pathway. J Orthop Res. 2021;39(8):1777–88. [DOI] [PubMed] [Google Scholar]
  • 38. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Luo Y, Yi X, Liang T, Jiang S, He R, Hu Y, Bai L, Wang C, Wang K, Zhu L. Autograft microskin combined with adipose-derived stem cell enhances wound healing in a full-thickness skin defect mouse model. Stem Cell Res Ther. 2019;10(1):279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Mousavi SS, Keshvari H, Daemi H. Partial sulfation of gellan gum produces cytocompatible, body temperature-responsive hydrogels. Int J Biol Macromol. 2023;235:123525. [DOI] [PubMed] [Google Scholar]
  • 41. Shah S, Esdaille CJ, Bhattacharjee M, Kan HM, Laurencin CT. The synthetic artificial stem cell (SASC): shifting the paradigm of cell therapy in regenerative engineering. Proc Natl Acad Sci U S A. 2022;119(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. He BH, Christin M, Mouchbahani-Constance S, Davidova A, Sharif-Naeini R. Mechanosensitive ion channels in articular nociceptors drive mechanical allodynia in osteoarthritis. Osteoarthritis Cartilage. 2017;25(12):2091–99. [DOI] [PubMed] [Google Scholar]
  • 43. Okun A, Liu P, Davis P, Ren J, Remeniuk B, Brion T, Ossipov MH, Xie J, Dussor GO, King T, Porreca F. Afferent drive elicits ongoing pain in a model of advanced osteoarthritis. Pain. 2012;153(4):924–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Neill T, Schaefer L, Iozzo RV. Decorin as a multivalent therapeutic agent against cancer. Adv Drug Deliv Rev. 2016;97:174–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Pugsley HR. Assessing autophagic flux by measuring LC3, p62, and LAMP1 Co-localization using multispectral imaging flow cytometry.J Vis Exp. 2017(125):55637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Tanida I, Ueno T, Kominami E. LC3 and autophagy. Methods Mol Biol. 2008;445:77–88. [DOI] [PubMed] [Google Scholar]
  • 47. Choe JY, Jung HY, Park KY, Kim SK. Enhanced p62 expression through impaired proteasomal degradation is involved in caspase-1 activation in monosodium urate crystal-induced interleukin-1b expression. Rheumatology (Oxford). 2014;53(6):1043–53. [DOI] [PubMed] [Google Scholar]
  • 48. Buraschi S, Neill T, Goyal A, Poluzzi C, Smythies J, Owens RT, Schaefer L, Torres A, Iozzo RV. Decorin causes autophagy in endothelial cells via Peg3. Proc Natl Acad Sci USA. 2013;110(28):E2582–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Yao T, Zhang CG, Gong MT, Zhang M, Wang L, Ding W. Decorin-mediated inhibition of the migration of U87MG glioma cells involves activation of autophagy and suppression of TGF-beta signaling. FEBS Open Bio. 2016;6(7):707–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Park YB, Ha CW, Lee CH, Yoon YC, Park YG. Cartilage regeneration in osteoarthritic patients by a composite of allogeneic umbilical cord blood-derived mesenchymal stem cells and hyaluronate hydrogel: results from a clinical trial for safety and proof-of-concept with 7 years of extended follow-up. Stem Cells Transl Med. 2017;6(2):613–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Zeng J, Huang L, Xiong H, Li Q, Wu C, Huang Y, Xie H, Shen B. Injectable decellularized cartilage matrix hydrogel encapsulating urine-derived stem cells for immunomodulatory and cartilage defect regeneration. NPJ Regen Med. 2022;7(1):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Antich C, Jiménez G, de Vicente J, López-Ruiz E, Chocarro-Wrona C, Griñán-Lisón C, Carrillo E, Montañez E, Marchal JA. Development of a biomimetic hydrogel based on predifferentiated mesenchymal stem-cell-derived ECM for cartilage tissue engineering. Adv Healthc Mater. 2021;10(8):e2001847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Zhu H, Wu X, Liu R, Zhao Y, Sun L. ECM-inspired hydrogels with ADSCs encapsulation for rheumatoid arthritis treatment. Adv Sci (Weinh). 2023;10(9):e2206253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Gasperini L, Mano JF, Reis RL. Natural polymers for the microencapsulation of cells. J R Soc Interface. 2014;11(100):20140817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Silva NA, Moreira J, Ribeiro-Samy S, Gomes ED, Tam RY, Shoichet MS, Reis RL, Sousa N, Salgado AJ. Modulation of bone marrow mesenchymal stem cell secretome by ECM-like hydrogels. Biochimie. 2013;95(12):2314–19. [DOI] [PubMed] [Google Scholar]
  • 56. Ng JY, Tan KYF, Ee PLR. Sugar-assisted cryopreservation of stem cell-laden gellan gum-collagen interpenetrating network hydrogels. Biomacromolecules. 2022;23(7):2803–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Park A, Choi JH, Lee S, Been S, Song JE, Khang G. Application of double network of gellan gum and pullulan for bone marrow stem cells differentiation towards chondrogenesis by controlling viscous substrates. J Tissue Eng Regen Med. 2020;14(11):1592–603. [DOI] [PubMed] [Google Scholar]
  • 58. Ai C, Liu L, Wong K, Tan XH, Goh JCH. The effect of chondroitin sulfate concentration and matrix stiffness on chondrogenic differentiation of mesenchymal stem cells. Biomater Sci. 2023;11:4557–73. [DOI] [PubMed] [Google Scholar]
  • 59. Robinson KA, Sun M, Barnum CE, Weiss SN, Huegel J, Shetye SS, Lin L, Saez D, Adams SM, Iozzo RV, Soslowsky LJ, et al. Decorin and biglycan are necessary for maintaining collagen fibril structure, fiber realignment, and mechanical properties of mature tendons. Matrix Biol. 2017;64:81–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Gronau T, Krüger K, Prein C, Aszodi A, Gronau I, Iozzo RV, Mooren FC, Clausen-Schaumann H, Bertrand J, Pap T, Bruckner P, et al. Forced exercise-induced osteoarthritis is attenuated in mice lacking the small leucine-rich proteoglycan decorin. Ann Rheum Dis. 2017;76(2):442–49. [DOI] [PubMed] [Google Scholar]
  • 61. Han B, Li Q, Wang C, Patel P, Adams SM, Doyran B, Nia HT, Oftadeh R, Zhou S, Li CY, Liu XS, et al. Decorin regulates the aggrecan network integrity and biomechanical functions of cartilage extracellular matrix. ACS Nano. 2019;13(10):11320–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Chery DR, Han B, Zhou Y, Wang C, Adams SM, Chandrasekaran P, Kwok B, Heo SJ, Enomoto-Iwamoto M, Lu XL, Kong D, et al. Decorin regulates cartilage pericellular matrix micromechanobiology. Matrix Biol. 2021;96:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Ansboro S, Hayes JS, Barron V, Browne S, Howard L, Greiser U, Lalor P, Shannon F, Barry FP, Pandit A, Murphy JM. A chondromimetic microsphere for in situ spatially controlled chondrogenic differentiation of human mesenchymal stem cells. J Control Release. 2014;179:42–51. [DOI] [PubMed] [Google Scholar]
  • 64. Recha-Sancho L, Semino CE. Chondroitin sulfate- and decorin-based self-assembling scaffolds for cartilage tissue engineering. Plos One. 2016;11(6):e0157603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Zwambag DP, Molladavoodi S, Guerreiro MJ, DeWitte-Orr SJ, Gregory DE. Immuno-stimulatory capacity of decorin in the rat tail intervertebral disc and the mechanical consequence of resultant inflammation. Eur Spine J. 2020;29(7):1641–48. [DOI] [PubMed] [Google Scholar]
  • 66. Halari CD, Renaud SJ, Lala PK. Molecular mechanisms in IL-1beta-mediated decorin production by decidual cells. Mol Hum Reprod. 2021;27(12):gaab068. [DOI] [PubMed] [Google Scholar]
  • 67. Merline R, Moreth K, Beckmann J, Nastase MV, Zeng-Brouwers J, Tralhao JG, Lemarchand P, Pfeilschifter J, Schaefer RM, Iozzo RV, Schaefer L. Signaling by the matrix proteoglycan decorin controls inflammation and cancer through PDCD4 and MicroRNA-21. Sci Signal. 2011;4(199): ra75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Wu M, Downie LE, Hill LJ, Chinnery HR. Topical decorin reduces corneal inflammation and imparts neuroprotection in a mouse model of benzalkonium chloride-induced corneal neuropathy. Invest Ophthalmol Vis Sci. 2023;64(2):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Wu M, Downie LE, Grover LM, Moakes RJA, Rauz S, Logan A, Jiao H, Hill LJ, Chinnery HR. The neuroregenerative effects of topical decorin on the injured mouse cornea. J Neuroinflammation. 2020;17(1):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Yang CM, Huang YJ, Hsu SH. Enhanced autophagy of adipose-derived stem cells grown on chitosan substrates. Biores Open Access. 2015;4(1):89–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Liu T, Ma X, Ouyang T, Chen H, Lin J, Liu J, Xiao Y, Yu J, Huang Y. SIRT1 reverses senescence via enhancing autophagy and attenuates oxidative stress-induced apoptosis through promoting p53 degradation. Int J Biol Macromol. 2018;117:225–34. [DOI] [PubMed] [Google Scholar]
  • 72. Li C, Chen K, Jia M, Ding X, Jiang Z, Li L, Zhang D. AMPK promotes survival and adipogenesis of ischemia-challenged ADSCs in an autophagy-dependent manner. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(12):1498–510. [DOI] [PubMed] [Google Scholar]
  • 73. Chinnapaka S, Yang KS, Flowers Q, Faisal M, Nerone WV, Rubin JP, Ejaz A. Metformin improves stemness of human adipose-derived stem cells by downmodulation of mechanistic target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK) signaling. Biomedicines. 2021;9(12):1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Yin Y, Chen F, Li J, Yang J, Li Q, Jin P. AURKA enhances autophagy of adipose derived stem cells to promote diabetic wound repair via targeting FOXO3a. J Invest Dermatol. 2020;140(8):1639–49. [DOI] [PubMed] [Google Scholar]
  • 75. Gubbiotti MA, Neill T, Frey H, Schaefer L, Iozzo RV. Decorin is an autophagy-inducible proteoglycan and is required for proper in vivo autophagy. Matrix Biol. 2015;48:14–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Buraschi S, Neill T, Iozzo RV. Decorin is a devouring proteoglycan: remodeling of intracellular catabolism via autophagy and mitophagy. Matrix Biol. 2019;75–76:260–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Wise RM, Al-Ghadban S, Harrison MAA, Sullivan BN, Monaco ER, Aleman SJ, Donato UM, Bunnell BA. Short-term autophagy preconditioning upregulates the expression of COX2 and PGE2 and alters the immune phenotype of human adipose-derived stem cells in vitro. Cells. 2022;11(9):1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Jiang LB, Lee S, Wang Y, Xu QT, Meng DH, Zhang J. Adipose-derived stem cells induce autophagic activation and inhibit catabolic response to pro-inflammatory cytokines in rat chondrocytes. Osteoarthritis Cartilage. 2016;24(6):1071–81. [DOI] [PubMed] [Google Scholar]

Articles from Cell Transplantation are provided here courtesy of SAGE Publications

RESOURCES