933
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Exosomes derived from berberine-treated bone marrow mesenchymal stem cells ameliorate inflammatory arthritis in rats with collagen-induced rheumatoid arthritis

, , , & ORCID Icon
Article: 2220566 | Received 02 Mar 2023, Accepted 25 May 2023, Published online: 06 Jul 2023

ABSTRACT

To evaluate the effect of exosomes secreted by berberine-treated bone marrow-derived mesenchymal stem cells (BMSCs) on synovial inflammation in collagen-induced arthritis (CIA) model. Exosomes were isolated from berberine-treated BMSCs (Ber-BMSC-EXs) and untreated BMSCs (BMSC-EXs). CIA rats were developed and treated with Ber-BMSC-EXs or BMSC-EXs. Clinical arthritis index was measured, activation of fibroblast-like synoviocytes (FLSs) were evaluated, and response and frequency of T helper 17 (Th17) and regulatory T (Treg) cells as well as cytokine levels in synovial fluid were determined. Ber-BMSC-EXs (with a superior effect than BMSC-EXs) significantly alleviated RA clinical score and paw inflammation, balanced Th17 and Treg cells’ responses and population, modulated levels of pro-inflammatory and anti-inflammatory cytokines in joint synovial fluid, and inhibited FLS activation in CIA rats. Ber-BMSC-EXs (with a significantly greater effect than BMSC-EXs) could significantly ameliorate synovial joint inflammation and clinical symptoms of arthritis in CIA rats, showing that berberine can reinforce anti-arthritic effects of BMSC-EXs.

1. Introduction

Rheumatoid arthritis (RA) is a chronic progressive inflammatory disease that predominantly attacks the synovial tissues and joints. RA is mainly characterized by chronic synovial inflammation (synovitis) and hyperplastic synovial cells (synovial hyperplasia), caused by inflammatory cell infiltration in the synovium, which leads to progressive and irreversible erosion of the bone and cartilage. Indeed, the inflammatory milieu in the synovial cavity provokes a vigorous tissue response, mainly activation of fibroblast-like synoviocytes (FLS) with a tumour-like inflammatory proliferation and invasive phenotype, which contribute to synovial inflammation and hyperplasia. Activated FLSs secrete numerous inflammatory mediators and proteases, which lead to the formation of synovial pannus and the penetration of the inflamed hyperplastic synovium into the neighbour articular structures, causing cartridge and bone damage. Pro-inflammatory and tissue-damaging cellular responses in synovitis are integrated by cytokines, such as tumour necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-17, which are produced by activated FLSs and various infiltrated immune cells (Bustamante et al., Citation2017; Tu et al., Citation2018). Among immune cells, CD4+ T cells including T helper 17 (Th17) and regulatory T (Treg) cells have the key roles in the RA pathogenesis. Th17 cells are pro-inflammatory cells producing IL-17 cytokine that exacerbates the synovial inflammation and the joint destruction, while Treg cells, which are weakly differentiated during RA, are anti-inflammatory cells producing IL-10 and transforming growth factor beta (TGF-β) cytokines that suppress Th17-mediated responses by reducing differentiation and homing of Th17 cells at the inflammatory joints (Banerjee et al., Citation2021; Baroroh et al., Citation2021; Shetty et al., Citation2021; You et al., Citation2021; Zhai et al., Citation2019).

The inflammation inhibition is the main mechanism underlying the preventive effects of the available RA therapeutics on the joint damage and bone loss. Currently, prescribed therapeutics used for the RA treatment, such as steroidal and non-steroidal anti-inflammatory drugs, disease-modifying anti-rheumatic drugs, and biologic agents, often provide relatively limited efficacy together with a momentary relief, and result in many severe side effects with long-term use (Crossfield et al., Citation2021; Leman et al., Citation2021). It is thus needed to explore alternative anti-arthritic medications with higher effectiveness and reliability.

Exosomes have been known as effectual players in the RA progression (Tavasolian et al., Citation2020). Exosomes are nano-sized (below 150 nm) extracellular vesicles that are actively secreted by various types of cells and act as mediators in cell-to-cell communications by transferring functional cargoes, such as proteins, lipids, and genetic materials, thereby affecting the behaviour of recipient cells by modulating biological processes and cellular signallings (Tavasolian et al., Citation2020; Zhai et al., Citation2022). Mesenchymal stem cells (MSCs), such as bone marrow-derived mesenchymal stem cells (BMSCs), have been found to release a mass of exosomes with the immunomodulatory-mediated therapeutic activity (Burrello et al., Citation2016; Cosenza et al., Citation2017; Yeo et al., Citation2013). Of note, recent studies showed the interest of using BMSC-derived exosomes (BMSC-EXs) to alleviate clinical symptoms of RA. An early study indicated that BMSC-EXs exert the anti-inflammatory effect on B and T lymphocytes in experimental models of inflammatory arthritis (Cosenza et al., Citation2018). Subsequent studies have shown that BMSC-EXs can inactivate RA-related FLSs (RA-FLSs), whereby reducing clinical symptoms of RA in collagen-induced arthritis (CIA) inflammatory model (Chen et al., Citation2018; Meng et al., Citation2020; Meng & Qiu, Citation2020; Su et al., Citation2021).

There is increasing evidence that some naturally occurring compounds, such as berberine, can improve the function of exosomes in various disease conditions (Dong et al., Citation2022; Kalani & Chaturvedi, Citation2017; Li, Stöckl, et al., Citation2021; Osterman et al., Citation2015; Qiu et al., Citation2020; Taverna et al., Citation2015, Citation2016; Wang et al., Citation2011; Wu, Zhou, et al., Citation2016; Zhai et al., Citation2021). Berberine is a well-studied and major bioactive ingredient of various medicinal herbs/plants, such as Berberis aquifolium, Phellodendron japonicum, and Coptis chinensis, which have been widely used in traditional Chinese medicine. Berberine has also been found to exert therapeutic impacts on a wide range of diseases in preclinical studies, including microbial infections and periodontitis (Mohammadian Haftcheshmeh & Momtazi-Borojeni, Citation2021), liver fibrosis (Feng et al., Citation2009), hyperlipidaemia and diabetes (Fatahian et al., Citation2020; Lan et al., Citation2015; Ma et al., Citation2018), autoimmunity (Ehteshamfar et al., Citation2020), cancer (Ayati et al., Citation2017; Mortazavi et al., Citation2020; Rauf et al., Citation2021), and RA (Huang et al., Citation2021; Shen et al., Citation2020). Berberine is mainly known with thanks to its anti-inflammatory/immunomodulatory activities (Ehteshamfar et al., Citation2020; Haftcheshmeh et al., Citation2022; Kang et al., Citation2002; Li et al., Citation2016; Mo et al., Citation2014; Sharma et al., Citation2020; Sujitha et al., Citation2018). In CIA rat model, berberine was found to exert disease-modifying activity that was suggested to be attributable to the suppression of inflammation (Wang et al., Citation2014). Similarly, other studies indicated that berberine can alleviate the clinical signs of arthritis in CIA rats, and suggested that anti-arthritic impact of berberine was mediated by inhibition of proliferation in RA-FLSs and induction of apoptosis in dendritic cells as well as balancing Th17/Treg ratio (Dinesh & Rasool, Citation2019; Hu et al., Citation2011; Sujitha et al., Citation2020; Wang et al., Citation2017, Citation2018; Yue et al., Citation2017). Of note, a recent study showed that berberine can induce chondrogenic differentiation of BMSCs by enhancing the activity of exosomes derived from platelet-rich plasma (Dong et al., Citation2022). The impact of berberine on the function of exosomes can also be supported by another study that demonstrated berberine can exert protective impact on diabetic nephropathy through inhibiting the function of destructive exosomes released by high glucose-induced glomerular mesangial cells (Wang et al., Citation2011).

With reference to these findings and also similar effects of berberine and BMSC-EXs on RA in the CIA model, we encouraged to determine whether berberine affects anti-arthritic effect of BMSC-EXs in CIA rats. To this end, we isolated exosomes secreted by BMSCs treated with and without berberine and evaluated their effects on in vitro inflammatory proliferation of FLSs isolated from CIA rats (CIA-FLSs) and also on clinical symptoms and joint inflammation in CIA rats.

2. Materials and methods

2.1. Cell culture

Rat BM-MSCs were obtained from the Cell Bank of Chinese Academy of Sciences (Shanghai, China) received with the fifth passage and utilized within the seventh to the eighth passages in accordance with the manufacturer’s protocols. The cells were cultured in low-sugar Dulbecco’s modified Eagle’s medium/F12 (DMEM; Gibco) containing 10% exosome-free fetal bovine serum (FBS; Gibco), 2 mM l-glutamine (Sigma), 100 mg/ml streptomycin and 100 U/ml penicillin (Gibco), at 37°C in an incubator with 95% humidity and 5% CO2.

2.2. Exosome isolation

BM-MSCs (2.5 × 106 cells) were seeded in a T175 flask and the culture supernatant was collected after 48 h of treatment with or without berberine (10 μM). Differential centrifugation and ultracentrifugation approach was used to isolate exosomes released by treated or untreated cells, as previously reported (Li, Stöckl, et al., Citation2021; Osterman et al., Citation2015; Qiu et al., Citation2020; Wu, Zhou, et al., Citation2016). Briefly, the cell debris/dead cells and other contaminations in the cell culture supernatant were eliminated by sequential steps of centrifugation; 10 min at 300g, 15 min at 2500g, followed by 25 min at 12,000g, all at 4°C. The supernatant were then ultra-centrifuged for 80 min at 110,000g to separate exosomes, which were then dispersed in the Phosphate-buffered saline (PBS) buffer, filtered through a sterile 22-μm filter, and ultra-centrifuged again for another 80 min at 110,000g. Finally, the pellet was resuspended in the PBS buffer containing 25 mM Trehalose and stored at −80°C. To measure the concentration of isolated exosomes, a bicinchoninic acid assay (BCA) protein assay kit was employed in accordance with the manufacturer’s protocols (Abcam, Cambridge, UK).

2.3. Identification of isolated exosomes

2.3.1. Acetylcholinesterase activity assay

Acetylcholinesterase is a typical enzyme presented in the exosomes and has been frequently used as a biomarker to identify exosomes (Lancaster & Febbraio, Citation2005; Rieu et al., Citation2000; Savina et al., Citation2003). In the present study, an acetylcholinesterase activity assay was carried out in accordance with already reported instructions (Lancaster & Febbraio, Citation2005; Savina et al., Citation2003) to verify the presence of exosomes in berberine-treated BMSCs (Ber-BMSC-EXs) and untreated BMSCs (BMSC-EXs). In brief, each isolated sample, acetylthiocholine (Sigma), and 5,5′-dithio-bis[2-nitrobenzoic acid] (Sigma) were mixed and loaded into a 96-well plate. Then, absorbance values were detected at 412 nm in 5 min intervals for 30 min, using a microplate reader (Bio-Teck).

2.3.2. Dynamic light scattering

The size distribution of vesicles in exosome isolates (Ber-BMSC-EXs and BMSC-EXs) was determined using dynamic light scattering (DLS) by a Zetasizer (Malvern Instruments, Malvern, UK). Exosome isolates were diluted in the PBS buffer (1:200) and measurements were recorded for 30 s at 25°C. Three replicates of each sample were measured and averaged to present the size and concentration values.

2.3.3. Transmission electron microscopy

The size and morphology of vesicles in exosome isolates (BMSC-EXs and Ber-BMSC-EXs) were monitored by transmission electron microscopy (TEM). Vesicles were decorated by negative staining to be visualized by a TEM instrument (Philips CM10 TEM; ×200,000 magnification). In brief, freshly isolated exosomes were dispersed in the cold PBS buffer and loaded onto parafilm. The loaded fluid was covered using a formvar (polyvinyl formal)-carbon-coated 400 copper mesh grid for 10 min at 25°C, followed by 1 min of incubation with 2% phosphotungstic acid and drying at 25°C for 15 min.

2.4. Animals

Male Sprague–Dawley rats (5–7 weeks old, 160–180 g) were obtained from Experimental Animal Centre of Shandong University. Animals were kept in the pathogen-free condition with the controlled temperature (24 ± 2°C) and humidity (55 ± 5%), under a 12 h dark/light cycle, with ad libitum access to tap water and standard chow diet. The priority was to decrease the number of animals used and to minimize animals’ suffering.

2.5. Development and assessment of collagen-induced RA (CIA) in rat

CIA was developed in rats according to the previously published method (Brand et al., Citation2007; Rosloniec et al., Citation2010), with some modifications. In brief, type II collagen (CII; 4 mg/ml; Sigma) was emulsified in complete Freund’s adjuvant (CFA; 4 mg/ml; Sigma) at 1:1 ratio. On day zero, rats were immunized with 200 µl CII/CFA emulsion via a slow subcutaneous injection at 1.5 cm distal from the base of the tail, followed by a booster immunization on day 7 with 100 µl emulsion at a tail area proximal to the primary injection site. Rats were daily checked for signs and symptoms of arthritis by two blinded, expert examiner. The arthritis severity was determined by scoring the inflamed paw according to the clinical arthritis index graded from 0 to 4, in which 0 = no swelling or erythema, 1 = swelling or erythema of one digit, 2 = swelling or erythema of two digits, 3 = swelling or erythema of more than two digits and footpad, and 4 = swelling or erythema of the entire paw and joint rigidity with incapacity to bend the knee (Choi et al., Citation2016; Tong et al., Citation2015). The total score of four paws with a maximum score of 16 for each rat was employed to determine the overall severity and progression of RA. The incidence of disease was denoted as the appearance of inflamed paws with a score ≥2 among four paws (Choi et al., Citation2016). The swelling volume was determined in paws with a foot volume plethysmometer once a week after CII/CFA immunization.

2.6. Treatment and grouping

The primary signs of arthritis in CIA rats were observed in knee joints after 4 weeks. Rats were allocated into four experimental groups (n = 8) on day 28, including the normal control group, the CIA model group, CIA + BMSC-EXs group, and CIA + Ber-BMSC-EXs group. In the two latter groups, the knee joints of CIA rats were injected intra-articularly with BMSC-EXs (100 µg/100 µl) and Ber-BMSC-EXs (100 µg/100 µl), respectively, on a weekly basis for 4 weeks from day 28, under anaesthesia. In the two former groups, normal rats and CIA rats received intra-articular injection of PBS buffer (100 µl) in the same schedule. Of note, the treatment dose, administration route, and power of study were selected in accordance with prior reports that demonstrated beneficial in vivo effects in rodents (Qi et al., Citation2016; Zhang et al., Citation2016).

One week after the last treatment, all rats were euthanized using an extra dosage of chloral hydrate hydrochloride, and subsequent experimental studies on the knee joints and blood samples were conducted as explained in the following sections.

2.7. Isolation and culture of CIA-FLSs

CIA-FLSs were isolated by enzymatic digestion method from synovial tissues of CIA rats with reference to an already published protocol (Zhao et al., Citation2016). On day 28 after CIA and under sterile conditions, synovial tissues were dissected form the knee joints, cut into small sections (<1 mm), washed gently in the sterile PBS buffer, and digested through incubation in the PBS containing type I collagenase (1 mg/ml) at 37°C for 2 h. The obtained cell suspension was incubated overnight in low-sugar DMEM medium containing 10% FBS, 2 mM l-glutamine, 100 mg/ml streptomycin, and 100 U/ml penicillin, in 95% air humidity with 5% CO2 at 37°C. Afterward, non-adherent cells and cell debris were eliminated and a homogenous population of adherent CIA-FLSs was cultured under a similar condition for subsequent experiments.

2.8. Purity assessment of CIA-FLSs

The purity of cultured CIA-FLSs (passage 3) was evaluated using flow cytometry analysis of the two fibroblast markers, CD90.1 (Thy-1) and CD54 Intercellular adhesion molecule (ICAM-1), in accordance with manufacture’s instructions (Biolegend). Briefly, 1 × 105 cells were stained with phycoerythrin (PE)-tagged anti-rat CD90.2 and anti-rat CD54 monoclonal antibodies (Biolegend), through incubation in the stain buffer (PBS containing 1% bovine serum albumin (BSA)) at 4°C for 40 min. PE-tagged mouse IgG1 isotype antibody (Biolegend) was also used as the negative control. Subsequently, a flow cytometry instrument (FACS Calibur; BD Biosciences, San Jose, USA) was employed to evaluate cell fluorescence, and FlowJo software was then used to analyse the obtained data.

2.9. Cell viability/proliferation assay

To evaluate the effect of Ber-BMSC-EXs and BMSC-EXs on the proliferation of isolated CIA-FLSs, the cell viability assay was carried out by the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide] assay (Sigma-Aldrich). In brief, cells were seeded in the 96-well plates at a density of 1 × 104 cells/well, and incubated in 5% CO2 at 37°C for 24 h. Thereafter, the cultured cells were treated with Ber-BMSC-EXs (100 µg/ml) or BMSC-EXs (100 µg/ml) or vehicle control (PBS), and incubated for 24, 48, and 72 h. Each treatment was repeated six times. The cells were then incubated with MTT solution (5 mg/ml) for 4 h. Afterward, the produced formazan crystals were dissolved in dimethyl sulfoxide, and absorbance at 560 nm was detected using an ELISA plate reader (Bio-Tek). The results were presented in terms of cell viability % in comparison with control cells as the mean ± standard deviation (SD) of three independent assays.

2.10. Assessment of synovial inflammation

To determine the effect of Ber-BMSC-EXs and BMSC-EXs on the synovial inflammation in CIA rats, we evaluated Th17 and Treg cells (using flow cytometry analysis) as well as cytokine levels (using ELISA assay) in the synovial fluids of knee joints at the end of study (week 8) and cytokine production by isolated CIA-FLSs as explained in the following subsections.

2.10.1. Collection of synovial fluid

To collect the synovial fluid of the knee joint, the already reported protocols were used with some modifications as follows. After sacrificing rats, the skin and the patellar ligaments were excised to uncover the synovial membrane of the knee. The needle (30-gauge) of a 0.3 ml syringe filled with 250 μl of heparinized PBS was inserted into the membrane, and the articular cavity was washed via slow and repetitive infusions and aspirations with PBS to collect synovial lavage ingredients. The process was twicely repeated to collect 500 μl of synovial fluid sample.

2.10.2. Isolation of synovial fluid mononuclear cells

To evaluate the presence of Th17 and Treg cells in knee joints, mononuclear cells were isolated from synovial fluids in accordance with previously published protocol, as follows. Freshly isolated synovial fluids were centrifuged at 700g for 8 min at 23°C (supernatant was collected for the cytokine assay by the ELISA). Cell pellets were resuspended in Roswell park memorial institute (RPMI) 1640 supplemented with density gradient cell separation solution (Sigma) and were centrifuged at 1500g for 30 min at 22°C. The cells were then washed by PBS buffer through centrifugation (repeated three times) at 400g for 8 min at 23°C. The isolated cells were then resuspended in the RPMI 1640 medium, and live cells were counted by the trypan blue-staining and analysed by flow cytometry.

2.10.3. Flow cytometry analysis

The presence of Th17 cells (CD4+ IL-17+ T cells) and Treg cells (CD4+ CD25+ Foxp3+ T cells) in synovial fluids was determined using flow cytometry analysis. In brief, the isolated synovial fluid mononuclear cells were incubated for 4 h at 37°C in the complete RPMI 1640 medium containing phorbol myristate acetate (50 ng/ml), ionomycin (500 ng/ml), and Brefeldin A (5 μg/ml). Afterward, cells were stained by fluorescent-tagged monoclonal antibodies against surface markers, including fluorescein isothio-cyanate-tagged anti CD4, PE-tagged anti CD25 (BD Biosciences). For intracellular staining, cells were first fixed and permeabilized by the Cytofix/Cytoperm kit (BD Biosciences) according to manufacturer’s instructions, then stained with PE-tagged anti IL-17 and PE-Cy5-tagged anti FoxP3 antibodies (BD Biosciences). Finally, stained cells were washed with the wash buffer (BD Biosciences), suspended in stain buffer (BD Biosciences), and analysed by a FACS-Calibur flow cytometer (BD Biosciences, San Jose, USA).

2.10.4. ELISA

To assay cytokines in the knee joint, the supernatants obtained after centrifugation of synovial fluid samples were used. The levels of pro-inflammatory cytokines including TNF-α, IL-1β, IL-6, and IL-17 as well as anti-inflammatory cytokines including IL-10 and TGF-β in synovial fluids were assayed using specific ELISA kits (MyBioSource or R&D Systems) in accordance with the manufacturer’s protocols. To evaluate cytokine production by the CIA-FLSs, cells (5 × 104 cells/well) were seeded in a 12-well plate, followed by 24-h incubation in 5% CO2 at 37°C, and then treated with Ber-BMSC-EXs (100 µg/ml) or BMSC-EXs (100 µg/ml). After 24-h of treatment, the levels of TNF-α, IL-1β, and IL-6 were assayed in the culture medium using ELISA in accordance with the instructions of the kits.

2.11. Statistical analysis

The results were analysed using GraphPad Prism software (Version 9). The values were presented as mean ± SD or median [interquartile range] for normally and non-normally distributed data, respectively. The statistical analysis was carried out using a one-way analysis of variance followed by Tukey’s post hoc test to assess the significance of the differences among the experimental groups. Data with p-value < 0.05 show the statistical significance.

3. Results

3.1. Exosome identification

The result of the acetylcholinesterase activity assay demonstrated a significantly high enzyme activity in samples isolated from berberine-treated and untreated BMSCs, whereas it was non-detectable in the control sample, verifying the presence of exosome particles in the isolates ((A)). It was further verified via evaluation of the size and the morphology of the particles in the isolates using DLS and TEM analysis. The particles in both samples isolated from berberine-treated and untreated BMSCs had a circular disk-like shape within the size range of exosomes, with mean diameters around 80–110 nm ((B–D)).

Figure 1. Exosome identification. Acetylcholinesterase activity was markedly increased in medium samples isolated from berberine-treated BMSCs and untreated BMSCs, verifying the presence of exosome particles (A). The size distribution analysis indicated the presence of vesicles in the size range of exosomes (80–110 nm) (B). Representative photomicrographs indicate the morphology and size of Ber-BMSC-EXs and BMSC-EXs visualized by TEM (×200,000 magnification; scale bar, 200 nm) (C and D). No significant differences were detected in acetylcholinesterase activity and size distribution of vesicle between Ber-BMSC-EXs and BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

Figure 1. Exosome identification. Acetylcholinesterase activity was markedly increased in medium samples isolated from berberine-treated BMSCs and untreated BMSCs, verifying the presence of exosome particles (A). The size distribution analysis indicated the presence of vesicles in the size range of exosomes (80–110 nm) (B). Representative photomicrographs indicate the morphology and size of Ber-BMSC-EXs and BMSC-EXs visualized by TEM (×200,000 magnification; scale bar, 200 nm) (C and D). No significant differences were detected in acetylcholinesterase activity and size distribution of vesicle between Ber-BMSC-EXs and BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

3.2. Purity of isolated CIA-FLSs

The flow cytometry analysis of isolated synovial cells indicated the presence of a high percentage of cells positive for fibroblast markers CD90.2 (91.7%) or CD54 (88.3%) (). The results confirmed the isolation of FLSs with a high purity from knee joints of CIA rats.

Figure 2. Purity assessment of isolated CIA-FLSs by flow cytometry. Fibroblast markers CD90.2 and CD54 tagged, respectively, 91.7% and 88.3% of FLSs isolated from knee joints of CIA rats, confirming a high purity of the isolated cells. CD, cluster of differentiation; CIA, collagen-induced rheumatoid arthritis; FLSs, fibroblast-like synoviocytes.

Figure 2. Purity assessment of isolated CIA-FLSs by flow cytometry. Fibroblast markers CD90.2 and CD54 tagged, respectively, 91.7% and 88.3% of FLSs isolated from knee joints of CIA rats, confirming a high purity of the isolated cells. CD, cluster of differentiation; CIA, collagen-induced rheumatoid arthritis; FLSs, fibroblast-like synoviocytes.

3.3. Ber-BMSC-EXs inhibit the activated CIA-FLSs

The impact of Ber-BMSC-EXs on the proliferation of CIA-FLSs was assessed using the MTT colorimetric assay. The isolated CIA-FLSs (at passage 3) were treated with Ber-BMSC-EXs, BMSC-EXs, or the vehicle control for different durations (24, 48, and 72 h). The results indicated that there was a significant difference (p < 0.001) between the proliferation of CIA-FLSs in exosome groups and control groups ((A)). Of note, both Ber-BMSC-EXs and BMSC-EXs significantly (p < 0.001) decreased the percentage of viable CIA-FLSs, when compared to the vehicle control, in a time-dependent manner ((A)). However, a significantly higher decrease (p < 0.001 or p = 0.005) in the percentage of viable cells was found in Ber-BMSC-EXs group than BMSC-EXs group during all treatment time points ((A)). Furthermore, the production of pro-inflammatory cytokines by CIA-FLSs treated with the Ber-BMSC-EXs or the BMSC-EXs were also measured. It was found that treatment with Ber-BMSC-EXs or BMSC-EXs could significantly decreased the production of pro-inflammatory cytokines including TNF-α, IL-6, and IL-1β by CIA-FLSs, when compared with the control treatment ((B–D)). Significantly, the modulatory effect of Ber-BMSC-EXs on the cytokine production was found to be more profound than that of BMSC-EXs.

Figure 3. The inhibitory effect of Ber-BMSC-EXs on the activated CIA-FLSs. Ber-BMSC-EXs reduced significantly the proliferation of CIA-FLSs during 72-h post-treatment compared with the control (A). Ber-BMSC-EXs decreased significantly the production of inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in CIA-FLSs compared with the control (B–D). Of note, the inhibitory effects of Ber-BMSC-EXs on the activated CIA-FLSs were significantly higher than that of BMSC-EXs. Results are presented as mean ± SD of three independent experiments. CIA, collagen-induced rheumatoid arthritis; FLSs, fibroblast-like synoviocytes.

Figure 3. The inhibitory effect of Ber-BMSC-EXs on the activated CIA-FLSs. Ber-BMSC-EXs reduced significantly the proliferation of CIA-FLSs during 72-h post-treatment compared with the control (A). Ber-BMSC-EXs decreased significantly the production of inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in CIA-FLSs compared with the control (B–D). Of note, the inhibitory effects of Ber-BMSC-EXs on the activated CIA-FLSs were significantly higher than that of BMSC-EXs. Results are presented as mean ± SD of three independent experiments. CIA, collagen-induced rheumatoid arthritis; FLSs, fibroblast-like synoviocytes.

3.4. Ber-BMSC-EXs inhibit the progression of inflammatory arteritis in CIA rats

To investigate the anti-RA impacts of Ber-BMSC-EXs, we employed the CIA rat model administrated with four intra-articular injections of Ber-BMSC-EXs (100 µg/rat) in weekly intervals from week 4 to 7 after the first immunization. The progression of RA was monitored until week 8. Clinical symptoms of RA were assessed in the paws. Ber-BMSC-EXs treatment was started after the initiation of severe CIA, 4 weeks after the first immunization. In CIA rats, the clinical scores of RA ((A)) and the degrees of paw swelling ((B)) were significantly elevated compared to control rats. However, 4 weeks treatment with weakly administration of Ber-BMSC-EXs or BMSC-EXs significantly reduces the RA clinical score ((A)) and paw swelling volume ((B)) in CFA rats. Notably, these were significantly decreased in CIA + Ber-BMSC-EXs rats when compared to CIA + BMSC-EXs rats ((A,B)).

Figure 4. The inhibitory effect of Ber-BMSC-EXs on the RA progression in CFA rats. Ber-BMSC-EXs significantly decreased score of the arthritis index (A) and the volume of the paw swelling (B) in CIA rats compared with control rats. Of note, the inhibitory effects of Ber-BMSC-EXs on the activated CIA-FLSs were significantly higher than that of BMSC-EXs. The changes of arthritis scores (A) and the paw swelling (B) were evaluated at weekly intervals from week 4 to week 8 after the first immunization in different experimental groups. The results of the paw swelling volume assessment are represented as the mean ± SD (n = 8). The results of the arthritis index are expressed as the median [25% percentile − 75% percentile] (n = 8). *p = 0.02 statistical difference between CIA group and CIA + Ber-EXs group, $p < 0.001 statistical difference between CIA group and CIA + Ber-EXs group, ***p < 0.001 statistical difference between CIA group and CIA + EXs group, and p < 0.001 statistical difference between CIA + EXs group and CIA + Ber-EXs, #p = 0.02 statistical difference between CIA + EXs group and CIA + Ber-EXs.

Figure 4. The inhibitory effect of Ber-BMSC-EXs on the RA progression in CFA rats. Ber-BMSC-EXs significantly decreased score of the arthritis index (A) and the volume of the paw swelling (B) in CIA rats compared with control rats. Of note, the inhibitory effects of Ber-BMSC-EXs on the activated CIA-FLSs were significantly higher than that of BMSC-EXs. The changes of arthritis scores (A) and the paw swelling (B) were evaluated at weekly intervals from week 4 to week 8 after the first immunization in different experimental groups. The results of the paw swelling volume assessment are represented as the mean ± SD (n = 8). The results of the arthritis index are expressed as the median [25% percentile − 75% percentile] (n = 8). *p = 0.02 statistical difference between CIA group and CIA + Ber-EXs group, $p < 0.001 statistical difference between CIA group and CIA + Ber-EXs group, ***p < 0.001 statistical difference between CIA group and CIA + EXs group, and p < 0.001 statistical difference between CIA + EXs group and CIA + Ber-EXs, #p = 0.02 statistical difference between CIA + EXs group and CIA + Ber-EXs.

3.5. Ber-BMSC-EXs modulate the levels of cytokines in the joint synovial fluid

A characteristic feature of CIA is a remarkable increase in the expression of pro-inflammatory cytokines. At the end of study, the levels of pro-inflammatory cytokines and anti-inflammatory cytokines in the joint synovial fluid were assayed by ELISA. The levels of pro-inflammatory cytokines, including IL-1β (p < 0.01), IL-6 (p < 0.01), IL-17 (p < 0.001), and TNF-α (p < 0.001), were significantly decreased in the CIA + Ber-BMSC-EXs group and CIA + BMSC-EXs group, when compared with the CIA group ((A–D)). Notably, the levels of pro-inflammatory cytokines in the CIA + Ber-BMSC-EXs group were significantly lower than those in the CIA + BMSC-EXs group (p < 0.001) ((A–D)). In case of anti-inflammatory cytokines, the levels of IL-10 and TGF-β were significantly higher in the CIA + Ber-BMSC-EXs group and the CIA + BMSC-EXs group compared to the CIA group ((E,F)). However, the levels of the anti-inflammatory cytokines in the CIA + Ber-BMSC-EXs group were significantly higher than those in the CIA + BMSC-EXs group ((E,F)).

Figure 5. Ber-BMSC-EXs modulate the abnormal levels of cytokines in the joint synovial fluid of CIA rats. The levels of pro-inflammatory cytokines, including IL-1β, IL-6, IL-17, and TNF-α, were significantly decreased in CIA + Ber-BMSC-EXs when compared with the control rats (A–D). However, the levels of anti-inflammatory cytokines, including IL-10 and TGF-β, were significantly decreased in CIA + Ber-BMSC-EXs compared with those in the control rats (D, E). The modulatory effect of Ber-BMSC-EXs on anti- and pro-inflammatory cytokines was more profound than that of BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

Figure 5. Ber-BMSC-EXs modulate the abnormal levels of cytokines in the joint synovial fluid of CIA rats. The levels of pro-inflammatory cytokines, including IL-1β, IL-6, IL-17, and TNF-α, were significantly decreased in CIA + Ber-BMSC-EXs when compared with the control rats (A–D). However, the levels of anti-inflammatory cytokines, including IL-10 and TGF-β, were significantly decreased in CIA + Ber-BMSC-EXs compared with those in the control rats (D, E). The modulatory effect of Ber-BMSC-EXs on anti- and pro-inflammatory cytokines was more profound than that of BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

3.6. Ber-BMSC-EXs modulate the Th17/Treg cells balance

The Th-17/Treg imbalance is a well-known impaired immune response in the RA pathogenesis; Th17 cells have a high frequency and exert a destructive role, whereas Treg cells have a protective role and an impaired function, leading to the joint inflammation and destruction. To evaluate the effect of intra-articular administration of exosome formulations on the cell responses and populations of Treg and Th17 cells in the joint synovial fluid of the CIA rats, ELISA and flow cytometry analysis were employed. Ber-BMSC-EXs or BMSC-EXs treatments significantly increased the proportion of CD4+ CD25+ Foxp3+ Treg cells () and the level of IL-10 and TGF-β, as markers of Treg response ((E,F)), in the synovial fluid of CIA rats compared to the control treatment. On the other hand, the population of IL-7-producing CD4+ () and the level of IL-17 () were significantly decreased in the synovial fluid of CIA + Ber-BMSC-EXs or CIA + BMSC-EXs groups, when compared with the control group. Of note, the Ber-BMSC-EXs treatment showed a higher influence on the frequency and responses of Treg and Th17 cells in the CIA rats than the BMSC-EXs treatment.

Figure 6. Ber-BMSC-EXs increase the population of Treg cells in the joint synovial fluid of CIA rats. (A) The representative flow cytometry data of CD25+ Foxp3+ Treg cells. (B) Percentage of CD25+ Foxp3+ Treg cells in the synovial fluid. Ber-BMSC-EXs show the superior effect than BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

Figure 6. Ber-BMSC-EXs increase the population of Treg cells in the joint synovial fluid of CIA rats. (A) The representative flow cytometry data of CD25+ Foxp3+ Treg cells. (B) Percentage of CD25+ Foxp3+ Treg cells in the synovial fluid. Ber-BMSC-EXs show the superior effect than BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

Figure 7. Ber-BMSC-EXs decrease the population of Th17 cells in the joint synovial fluid of CIA rats. (A) The representative flow cytometry data of CD4+ IL-17+ T cells. (B) Percentage of CD4+ IL-17+ T cells in the synovial fluid. Ber-BMSC-EXs show the superior effect than BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

Figure 7. Ber-BMSC-EXs decrease the population of Th17 cells in the joint synovial fluid of CIA rats. (A) The representative flow cytometry data of CD4+ IL-17+ T cells. (B) Percentage of CD4+ IL-17+ T cells in the synovial fluid. Ber-BMSC-EXs show the superior effect than BMSC-EXs. Results are presented as mean ± SD of three independent experiments.

4. Discussion

A growing number of experimental studies has shown that berberine and MSC-derived exosomes (Chang et al., Citation2022; Chen et al., Citation2018; Cosenza et al., Citation2018; Hajavi et al., Citation2019; Li, Fang, et al., Citation2021; Liu et al., Citation2020; Meng & Qiu, Citation2020; Su et al., Citation2021; Zhang et al., Citation2022) are able to effectively alleviate inflammatory arthritis in the experimental models of RA. In the present study, we evaluated and compared for the first time the effect of the local intra-articular administration of exosomes derived from BMSCs (BMSC-EXs) and berberine-treated BMSCs (Ber-BMSC-EXs) on the in vitro proliferation of CIA-FLSs and on the inflammatory arthritis in the CIA model. The resultant data indicated that either BMSC-EXs or Ber-BMSC-EXs could significantly ameliorate the synovial joint inflammation and the clinical symptoms of arthritis in CIA rats. However, Ber-BMSC-EXs exerted a significantly greater effect than BMSC-EXs, showing that berberine can reinforce anti-arthritic effects of BMSC-EXs.

FLSs play a key role in the RA/CIA pathogenesis. In RA/CIA, FLSs are abnormally activated and exhibit tumour-like proliferation and invasion, which contribute to synovial hyperplasia, causing the synovial pannus formation and cartridge degeneration (Bustamante et al., Citation2017; Tu et al., Citation2018). Thus, inhibiting excessive proliferation of activated FLSs may attenuate the disease severity and provide an effective approach in the RA treatment. Several independent experimental investigations suggested that berberine or BMSC-EXs (Meng et al., Citation2020; Meng & Qiu, Citation2020; Su et al., Citation2021; Zhang et al., Citation2022) could exert the anti-arthritic effect through proliferation inhibition and apoptosis induction of the activated arthritis-FLSs. Of note, we found that Ber-BMSC-EXs, with a higher effect than BMSC-EXs, reduced the in vitro proliferation of CIA-FLSs. Moreover, activated FLSs secrete a huge amount of pro-inflammatory cytokines, causing synovial inflammation and further intensifying joint destruction, and eventually worsening the arthritis progression (Bustamante et al., Citation2017; Tu et al., Citation2018). TNF-α provokes the synovial inflammation and joint destruction during RA via inducing the adhesion and migration of inflammatory cells and promoting the synthesis of other pro-inflammatory cytokines (Bertolini et al., Citation1986; Edrees et al., Citation2005; Matsuno et al., Citation2002). IL-1β plays a critical role in synovial inflammation and pannus formation. In association with other inflammatory cytokines such as TNF-α and IL-6, IL-1β exacerbates the progression of joint inflammation and concomitant cartilage and bone erosion in RA/CIA (Joosten et al., Citation1996, Citation1999; Nikfar et al., Citation2018; van den Berg et al., Citation1994; Yang et al., Citation2019). Increased levels of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 have been found in the synovial fluids from RA patients and CIA models. It was previously reported that BMSC-EXs decreased the production of inflammatory cytokines TNF-α and IL-1β in mouse FLSs, and inhibited the RA progression in the CIA mice (Li, Fang, et al., Citation2021). Interestingly, we found that Ber-BMSC-EXs, with a superior effect than BMSC-EXs, decreased the production of inflammatory cytokines including TNF-α, IL-1β, and IL-6 by CIA-FLS cells, in vitro. Consistently, we detected a significant reduction in the levels of these cytokines in the synovial fluid of the knee joints in the CIA rats treated with Ber-BMSC-EXs or BMSC-EXs; however, Ber-BMSC-EXs showed a greater effect than BMSC-EXs. Furthermore, clinical observations showed that the paw inflammation, a typical clinical sign of inflammatory CIA characterized by swelling and/or erythema, was significantly decreased in CIA rats treated with Ber-BMSC-EXs or BMSC-EXs; however, a higher effect was found with Ber-BMSC-EXs than BMSC-EXs.

The inflammatory cytokine milieu in the synovial cavity can also promote infiltration/differentiation of CD4+ T cells that affect development and progression of synovial inflammation (Amadi-Obi et al., Citation2007; Beal et al., Citation2012; Chen & Konkel, Citation2015; Wu, Yan, et al., Citation2016). The Th17 subtype of CD4+ T cells has the key role in synovial inflammation and subsequent bone erosion and cartilage degeneration, mainly by the production of IL-17 cytokine (Hernández-Palma et al., Citation2019; Lubberts et al., Citation2002, Citation2004; Misra et al., Citation2022). High levels of Th17 cells and IL-17 have been detected in the RA/CIA synovial fluid and tissues (Barton et al., Citation2007; Hajavi et al., Citation2019; Meehan et al., Citation2021; Misra et al., Citation2022; Paradowska-Gorycka et al., Citation2020). IL-17 exacerbates bone erosion (by inducing osteoclastogenesis) (Adamopoulos et al., Citation2010; Koenders et al., Citation2005; Kotake et al., Citation1999) and cartilage degeneration (by inducing production of metalloproteinases and breakdown of proteoglycans) (Chabaud et al., Citation2000, Citation2001). Another subtype of CD4+ T cells that are weakly differentiated during the RA progression is Treg cells that play an inhibitory role in the RA pathogenesis (Kondo et al., Citation2018; Paradowska-Gorycka et al., Citation2020). Treg cells produce anti-inflammatory cytokines IL-10 and TGF-β, which can suppress pro-inflammatory responses and impede the destructing impact of Th17 cells by decreasing their differentiation and homing at the inflammatory joints (Kondo et al., Citation2018). Hence, the Th17/Treg imbalance aggravates the development and progression of RA and CIA.

A previous study showed that BMSC-EXs can improve Th17/Treg balance and efficiently alleviate inflammation and arthritis signs in the CIA mice (Cosenza et al., Citation2018). Our results indicated that Ber-BMSC-EXs or BMSC-EXs can regulate the balance between Treg and Th17 cells. Ber-BMSC-EXs, with a higher effect than BMSC-EXs, were found to decrease the population of T17 cells (IL-17-producing CD4+ T cells) and increase the population of Treg cells (CD25+ Foxp3+ T cells) in the joint synovial fluid of the CIA rats. It was further supported by the results that showed a reduction of Th17-related cytokine (IL-17) and an increase of Treg-related cytokine (IL-10) in the synovial fluids. Thus, our results indicate that Ber-BMSC-EXs or BMSC-EXs can modulate the phenotype of the activated CD4+ T cells from IL-17-producing cells towards Foxp3+ Treg cells in the joint synovial fluid of CIA rats. These may be due to the modulatory effect of the exosome formulations on the cytokine levels in the synovial fluid. Of note, the presence of IL-17, IL-1β, and IL-6 in the synovial fluid can promote differentiation of the Th17 cells (Amadi-Obi et al., Citation2007; Wu, Yan, et al., Citation2016), while TGF-β can promote Treg cell differentiation (Beal et al., Citation2012; Chen & Konkel, Citation2015); notably, Ber-BMSC-EXs or BMSC-EXs increased the levels of TGF-β and decreased the levels of IL17, IL-1β, and IL-6 in the joint synovial fluid.

Importantly, the improving effects of berberine on anti-arthritic effects of BMSC-EXs can be due to anti-inflammatory properties of berberine. Of note, there have been a large number of investigations showing anti-inflammatory properties of berberine in various diseases like RA (Ehteshamfar et al., Citation2020; Haftcheshmeh et al., Citation2022; Mohammadian Haftcheshmeh & Momtazi-Borojeni, Citation2021; Shen et al., Citation2020). Thus, it can be suggested that berberine may alter cargo content of BMSC-EXs, such as miRs and proteins/enzymes that are responsible for anti-inflammatory properties of these exosomes, thereby improving the anti-arthritic feature of BMSC-EXs.

To the best of our knowledge, there is no study reporting the therapeutic efficacy of Ber-BMSC-EXs in the experimental inflammatory arthritis and, thus, the results of present study endow a novel therapeutic opportunity for BMSC-EXS in the RA treatment. However, although the results of our study clearly indicate a potential future therapy by berberine-primed exosomes, a number of limitations need to be acknowledged in the future studies. The impact of berberine on the cargo content of BMSC-EXS as well as the underlying molecular mechanisms should be evaluated. Moreover, the histopathological alterations in knee joints and the synovial tissue should also be assessed. Furthermore, it is essential to do in vivo toxicity assessments to determine non-toxic dose of Ber-BMSC-EXS in RA.

Ethics approval

This study was performed in line with the Guideline of the National Institute of Health for the Care and Use of Laboratory Animals. Approval was granted by the Ethics Committee of the Central Hospital Affiliated to Shandong First Medical University, Jinan, China (No. SFMU202127).

Author contributions

ZY contributed to the conception and design of the study. FS and YM conducted the experimental work, performed data acquisition, and prepared the first draft. FL and CL performed data analysis and interpretation of data. ZY revised the draft critically for important intellectual content. All authors read and approved the final version to be submitted.

Disclosure statement

The authors have no relevant financial or non-financial interests to disclose.

Data availability statement

All data generated or analysed during this study are included in this article. Further enquiries can be directed to the corresponding author.

Correction Statement

This article has been corrected with minor changes. These changes do not impact the academic content of the article.

Additional information

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

References

  • Adamopoulos, I. E., Chao, C. C., Geissler, R, Laface, D., Blumenschein, W., Iwakura, Y., McClanahan, T., & Bowman, E. P. (2010). Interleukin-17A upregulates receptor activator of NF-κB on osteoclast precursors. Arthritis Research & Therapy, 12(1), R29. https://doi.org/10.1186/ar2936
  • Amadi-Obi, A., Yu, C. R., Liu, X., Mahdi, R. M., Clarke, G. L., Nussenblatt, R. B., Gery, I., Lee, Y. S., & Egwuagu, C. E. (2007). TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nature Medicine, 13(6), 711–718. https://doi.org/10.1038/nm1585
  • Ayati, S. H., Fazeli, B., Momtazi-Borojeni, A. A., Cicero, A. F. G., Pirro, M., & Sahebkar, A. (2017). Regulatory effects of berberine on microRNome in cancer and other conditions. Critical Reviews in Oncology/Hematology, 116(2017), 147–158. https://doi.org/10.1016/j.critrevonc.2017.05.008
  • Banerjee, P., Kumar, T., Sarangi, S. C., Meetei, U. D., Devi, A. S., & Kumar, R. (2021). Anti-inflammatory potential of aqueous extract of Elsoltzia stachyodes on experimental models of inflammation in rats. Journal of Natural Science, Biology and Medicine, 12(1), 104.
  • Baroroh, H. N., Nugroho, A. E., Lukitaningsih, E., & Nurrochmad, A. (2021). Immune-enhancing effect of Bengkoang (Pachyrhizus erosus (L.) Urban) fiber fractions on mouse peritoneal macrophages, lymphocytes, and cytokines. Journal of Natural Science, Biology and Medicine, 12(1), 84–92. https://doi.org/10.4103/jnsbm.JNSBM_53_20
  • Barton, N. J., Stevens, D. A., Hughes, J. P., Rossi, A. G., Chessell, I. P., Reeve, A. J., & McQueen, D. S. (2007). Demonstration of a novel technique to quantitatively assess inflammatory mediators and cells in rat knee joints. Journal of Inflammation (London, England), 4(1), 13. https://doi.org/10.1186/1476-9255-4-13
  • Beal, A. M., Ramos-Hernández, N., Riling, C. R., Nowelsky, E. A., & Oliver, P. M. (2012). TGF-β induces the expression of the adaptor Ndfip1 to silence IL-4 production during iTreg cell differentiation. Nature Immunology, 13(1), 77–85. https://doi.org/10.1038/ni.2154
  • Bertolini, D. R., Nedwin, G. E., Bringman, T. S., Smith, D. D., & Mundy, G. R. (1986). Stimulation of bone resorption and inhibition of bone formation in vitro by human tumour necrosis factors. Nature, 319(6053), 516–518. https://doi.org/10.1038/319516a0
  • Brand, D. D., Latham, K. A., & Rosloniec, E. F. (2007). Collagen-induced arthritis. Nature Protocols, 2(5), 1269–1275. https://doi.org/10.1038/nprot.2007.173
  • Burrello, J., Monticone, S., Gai, C., Gomez, Y., Kholia, S., & Camussi, G. (2016). Stem cell-derived extracellular vesicles and immune-modulation. Frontiers in Cell and Developmental Biology, 4(83), 83–92.
  • Bustamante, M. F., Garcia-Carbonell, R., Whisenant, K. D., & Guma, M. (2017). Fibroblast-like synoviocyte metabolism in the pathogenesis of rheumatoid arthritis. Arthritis Research & Therapy, 19(1), 1–12. https://doi.org/10.1186/s13075-017-1303-3
  • Chabaud, M., Garnero, P., Dayer, J. M., Guerne, P. A., Fossiez, F., & Miossec, P. (2000). Contribution of interleukin 17 to synovium matrix destruction in rheumatoid arthritis. Cytokine, 12(7), 1092–1099. https://doi.org/10.1006/cyto.2000.0681
  • Chabaud, M., Lubberts, E., Joosten, L., van Den Berg, W., & Miossec, P. (2001). IL-17 derived from juxta-articular bone and synovium contributes to joint degradation in rheumatoid arthritis. Arthritis Research, 3(3), 168–177. https://doi.org/10.1186/ar294
  • Chang, T. H., Wu, C. S., Chiou, S. H., Chang, C. H., & Liao, H. J. (2022). Adipose-derived stem cell exosomes as a novel anti-inflammatory agent and the current therapeutic targets for rheumatoid arthritis. Biomedicines, 10(7). https://doi.org/10.3390/biomedicines10071725
  • Chen, W., & Konkel, J. E. (2015). Development of thymic Foxp3 + regulatory T cells: TGF-β matters. European Journal of Immunology, 45(4), 958–965. https://doi.org/10.1002/eji.201444999
  • Chen, Z., Wang, H., Xia, Y., Yan, F., & Lu, Y. (2018). Therapeutic potential of mesenchymal cell-derived miRNA-150-5p-expressing exosomes in rheumatoid arthritis mediated by the modulation of MMP14 and VEGF. The Journal of Immunology, 201(8), 2472–2482. https://doi.org/10.4049/jimmunol.1800304
  • Choi, J. K., Kim, S. W., Kim, D. S., Lee, J. Y., Lee, S., Oh, H. M., Ha, Y. S., Yoo, J., Park, P. H., Shin, T. Y., & Kwon, T. K. (2016). Oleanolic acid acetate inhibits rheumatoid arthritis by modulating T cell immune responses and matrix-degrading enzymes. Toxicology and Applied Pharmacology, 290(1), 1–9. https://doi.org/10.1016/j.taap.2015.11.005
  • Cosenza, S., Ruiz, M., Maumus, M., Jorgensen, C., & Noël, D. (2017). Pathogenic or therapeutic extracellular vesicles in rheumatic diseases: Role of mesenchymal stem cell-derived vesicles. International Journal of Molecular Sciences, 18(4), 889. https://doi.org/10.3390/ijms18040889
  • Cosenza, S., Toupet, K., Maumus, M., Luz-Crawford, P., Blanc-Brude, O., Jorgensen, C., & Noël, D. (2018). Mesenchymal stem cells-derived exosomes are more immunosuppressive than microparticles in inflammatory arthritis. Theranostics, 8(5), 1399. https://doi.org/10.7150/thno.21072
  • Crossfield, S. S. R., Buch, M. H., Baxter, P., Kingsbury, S. R., Pujades-Rodriguez, M., & Conaghan, P. G. (2021). Changes in the pharmacological management of rheumatoid arthritis over two decades. Rheumatology, 60(9), 4141–4151. https://doi.org/10.1093/rheumatology/keaa892
  • Dinesh, P., & Rasool, M. (2019). Berberine mitigates IL-21/IL-21R mediated autophagic influx in fibroblast-like synoviocytes and regulates Th17/Treg imbalance in rheumatoid arthritis. Apoptosis, 24(7), 644–661. https://doi.org/10.1007/s10495-019-01548-6
  • Dong, B., Liu, X., Li, J., Wang, B., Yin, J., Zhang, H., & Liu, W. (2022). Berberine encapsulated in exosomes derived from platelet-rich plasma promotes chondrogenic differentiation of the bone marrow mesenchymal stem cells via the Wnt/β-catenin pathway. Biological and Pharmaceutical Bulletin, 45(10), 1444–1451. https://doi.org/10.1248/bpb.b22-00206
  • Edrees, A. F., Misra, S. N., & Abdou, N. I. (2005). Anti-tumor necrosis factor (TNF) therapy in rheumatoid arthritis: Correlation of TNF-alpha serum level with clinical response and benefit from changing dose or frequency of infliximab infusions. Clinical and Experimental Rheumatology, 23(4), 469–474.
  • Ehteshamfar, S. M., Akhbari, M., Afshari, J. T., Seyedi, M., Nikfar, B., Shapouri-Moghaddam, A., Ghanbarzadeh, E., & Momtazi-Borojeni, A. A. (2020). Anti-inflammatory and immune-modulatory impacts of berberine on activation of autoreactive T cells in autoimmune inflammation. Journal of Cellular and Molecular Medicine, 24(23), 13573–13588. https://doi.org/10.1111/jcmm.16049
  • Fatahian, A., Haftcheshmeh, S. M., Azhdari, S., Farshchi, H. K., Nikfar, B., & Momtazi-Borojeni, A. A. (2020). Promising anti-atherosclerotic effect of berberine: Evidence from in vitro, in vivo, and clinical studies. Reviews of Physiology, Biochemistry and Pharmacology, 178, 83–110. https://doi.org/10.1007/112_2020_42
  • Feng, Y., Cheung, K.-F., Wang, N., Liu, P., Nagamatsu, T., & Tong, Y. (2009). Chinese medicines as a resource for liver fibrosis treatment. Chinese Medicine, 4(1), 1–11. https://doi.org/10.1186/1749-8546-4-16
  • Haftcheshmeh, S. M., Abedi, M., Mashayekhi, K., Mousavi, M. J., Navashenaq, J. G., Mohammadi, A., & Momtazi-Borojeni, A. A. (2022). Berberine as a natural modulator of inflammatory signaling pathways in the immune system: Focus on NF-κB, JAK/STAT, and MAPK signaling pathways. Phytotherapy Research, 36(3), 1216–1230. https://doi.org/10.1002/ptr.7407
  • Hajavi, J., Esmaeili, S. A., Varasteh, A. R., Vazini, H., Atabati, H., Mardani, F., Momtazi-Borojeni, A. A., Hashemi, M., Sankian, M., & Sahebkar, A. (2019). The immunomodulatory role of probiotics in allergy therapy. Journal of Cellular Physiology, 234(3), 2386–2398. https://doi.org/10.1002/jcp.27263
  • Hernández-Palma, L.A., García-Arellano, S., Bucala, R., Llamas-Covarrubias, MA., De la Cruz-Mosso, U., Oregon-Romero, E., Cerpa-Cruz, S., Parra-Rojas, I., Plascencia-Hernández, A., & Muñoz-Valle, J. F. (2019). Functional MIF promoter haplotypes modulate Th17-related cytokine expression in peripheral blood mononuclear cells from control subjects and rheumatoid arthritis patients. Cytokine, 115(3), 89–96. https://doi.org/10.1016/j.cyto.2018.11.014
  • Hu, Z., Jiao, Q., Ding, J., Liu, F., Liu, R., Shan, L., Zeng, H., Zhang, J., & Zhang, W. (2011). Berberine induces dendritic cell apoptosis and has therapeutic potential for rheumatoid arthritis. Arthritis & Rheumatism, 63(4), 949–959. https://doi.org/10.1002/art.30202
  • Huang, D.-N., Wu, F.-F., Zhang, A.-H., Sun, H., & Wang, X.-J. (2021). Efficacy of berberine in treatment of rheumatoid arthritis: From multiple targets to therapeutic potential. Pharmacological Research, 169(5), 105667. https://doi.org/10.1016/j.phrs.2021.105667
  • Joosten, L. A. B., Helsen, M. M., Saxne, T., van de Loo, F. A. J., Heinegård, D., & van den Berg, W. B. (1999). IL-1αβ blockade prevents cartilage and bone destruction in murine type II collagen-induced arthritis, whereas TNF-α blockade only ameliorates joint inflammation. The Journal of Immunology, 163(9), 5049–5055. https://doi.org/10.4049/jimmunol.163.9.5049
  • Joosten, L. A. B., Helsen, M. M., van de Loo, F. A., & van den Berg, W. B. (1996). Anticytokine treatment of established type II collagen-induced arthritis in DBA/1 mice. A comparative study using anti-TNF alpha, anti-IL-1 alpha/beta, and IL-1Ra. Arthritis and Rheumatism, 39(5), 797–809. https://doi.org/10.1002/art.1780390513
  • Kalani, A., & Chaturvedi, P. (2017). Curcumin-primed and curcumin-loaded exosomes: Potential neural therapy. Neural Regeneration Research, 12(2), 205. https://doi.org/10.4103/1673-5374.200799
  • Kang, B. Y., Chung, S. W., Cho, D., & Kim, T. S. (2002). Involvement of p38 mitogen-activated protein kinase in the induction of interleukin-12 p40 production in mouse macrophages by berberine, a benzodioxoloquinolizine alkaloid. Biochemical Pharmacology, 63(10), 1901–1910. https://doi.org/10.1016/S0006-2952(02)00982-6
  • Koenders, M. I., Lubberts, E., Oppers-Walgreen, B., van den Bersselaar, L., Helsen, M. M., Di Padova, F. E., Boots, A. M., Gram, H., Joosten, L. A., & van den Berg, W. B. (2005). Blocking of interleukin-17 during reactivation of experimental arthritis prevents joint inflammation and bone erosion by decreasing RANKL and interleukin-1. The American Journal of Pathology, 167(1), 141–149. https://doi.org/10.1016/S0002-9440(10)62961-6
  • Kondo, Y., Yokosawa, M., Kaneko, S., Furuyama, K., Segawa, S., Tsuboi, H., Matsumoto, I., & Sumida, T. (2018). Transcriptional regulation of CD 4+ T cell differentiation in experimentally induced arthritis and rheumatoid arthritis. Arthritis & Rheumatology, 70(5), 653–661. https://doi.org/10.1002/art.40398
  • Kotake, S., Udagawa, N., Takahashi, N., Matsuzaki, K., Itoh, K., Ishiyama, S., Saito, S., Inoue, K., Kamatani, N., Gillespie, M. T., & Martin, T. J. (1999). IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. The Journal of Clinical Investigation, 103(9), 1345–1352. https://doi.org/10.1172/JCI5703
  • Lan, J., Zhao, Y., Dong, F., Yan, Z., Zheng, W., Fan, J., & Sun, G. (2015). Meta-analysis of the effect and safety of berberine in the treatment of type 2 diabetes mellitus, hyperlipemia and hypertension. Journal of Ethnopharmacology, 161(4), 69–81. https://doi.org/10.1016/j.jep.2014.09.049
  • Lancaster, G. I., & Febbraio, M. A. (2005). Exosome-dependent trafficking of HSP70: A novel secretory pathway for cellular stress proteins. Journal of Biological Chemistry, 280(24), 23349–23355. https://doi.org/10.1074/jbc.M502017200
  • Leman, M. A., Claramita, M., & Rahayu, G. R. (2021). Predicting factors on modeling health behavior: A systematic review. American Journal of Health Behavior, 45(2), 268–278. https://doi.org/10.5993/AJHB.45.2.7
  • Li, G. Q., Fang, Y. X., Liu, Y., Meng, F. R., Wu, X., Zhang, C. W., Zhang, Y., Liu, Y. Q., & Liu, D. (2021). MicroRNA-21 from bone marrow mesenchymal stem cell-derived extracellular vesicles targets TET1 to suppress KLF4 and alleviate rheumatoid arthritis. Therapeutic Advances in Chronic Disease, 12(4), 20406223211007369. https://doi.org/10.1177/20406223211007369
  • Li, S., Stöckl, S., Lukas, C., Herrmann, M., Brochhausen, C., König, M. A., Johnstone, B., & Grässel, S. (2021). Curcumin-primed human BMSC-derived extracellular vesicles reverse IL-1β-induced catabolic responses of OA chondrocytes by upregulating miR-126-3p. Stem Cell Research & Therapy, 12(1), 252. https://doi.org/10.1186/s13287-021-02317-6
  • Li, Z., Zheng, J., Zhang, N., & Li, C. (2016). Berberine improves airway inflammation and inhibits NF-kappaB signaling pathway in an ovalbumin-induced rat model of asthma. Journal of Asthma, 53(10), 999–1005. https://doi.org/10.1080/02770903.2016.1180530
  • Liu, H., Li, R., Liu, T., Yang, L., Yin, G., & Xie, Q. (2020). Immunomodulatory effects of mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles in rheumatoid arthritis. Frontiers in Immunology, 11(8), 1912. https://doi.org/10.3389/fimmu.2020.01912
  • Lubberts, E., Joosten, L. A., van de Loo, F. A., Schwarzenberger, P., Kolls, J., & van den Berg, W. B. (2002). Overexpression of IL-17 in the knee joint of collagen type II immunized mice promotes collagen arthritis and aggravates joint destruction. Inflammation Research, 51(2), 102–104. https://doi.org/10.1007/BF02684010
  • Lubberts, E., Koenders, M. I., Oppers-Walgreen, B., Van Den Bersselaar, L., Coenen-de Roo, C. J., Joosten, L. A., & Van Den Berg, W. B. (2004). Treatment with a neutralizing anti-murine interleukin-17 antibody after the onset of collagen-induced arthritis reduces joint inflammation, cartilage destruction, and bone erosion. Arthritis and Rheumatism, 50(2), 650–659. https://doi.org/10.1002/art.20001
  • Ma, X., Chen, Z., Wang, L., Wang, G., Wang, Z., Dong, X., Wen, B., & Zhang, Z. (2018). The pathogenesis of diabetes mellitus by oxidative stress and inflammation: Its inhibition by berberine. Frontiers in Pharmacology, 9(7), 782. https://doi.org/10.3389/fphar.2018.00782
  • Matsuno, H., Yudoh, K., Katayama, R., Nakazawa, F., Uzuki, M., Sawai, T., Yonezawa, T., Saeki, Y., Panayi, G. S., Pitzalis, C., & Kimura T (2002). The role of TNF-α in the pathogenesis of inflammation and joint destruction in rheumatoid arthritis (RA): A study using a human RA/SCID mouse chimera. Rheumatology, 41(3), 329–337. https://doi.org/10.1093/rheumatology/41.3.329
  • Meehan, R. T., Regan, E. A., Hoffman, E. D., Wolf, M. L., Gill, M. T., Crooks, J. L., Parmar, P. J., Scheuring, R. A., Hill, J. C., Pacheco, K. A., & Knight V. (2021). Synovial fluid cytokines, chemokines and MMP levels in osteoarthritis patients with knee pain display a profile similar to many rheumatoid arthritis patients. Journal of Clinical Medicine, 10(21), 5027. https://doi.org/10.3390/jcm10215027
  • Meng, H. Y., Chen, L. Q., & Chen, L. H. (2020). The inhibition by human MSCs-derived miRNA-124a overexpression exosomes in the proliferation and migration of rheumatoid arthritis-related fibroblast-like synoviocyte cell. BMC Musculoskeletal Disorders, 21(1), 150. https://doi.org/10.1186/s12891-020-3159-y
  • Meng, Q., & Qiu, B. (2020). Exosomal MicroRNA-320a derived from mesenchymal stem cells regulates rheumatoid arthritis fibroblast-like synoviocyte activation by suppressing CXCL9 expression. Frontiers in Physiology, 11(5), 441. https://doi.org/10.3389/fphys.2020.00441
  • Misra, S., Mondal, S., Chatterjee, S., Dutta, S., Sinha, D., Bhattacharjee, D., Lahiri, D., & Ghosh, A. (2022). Interleukin-17 as a predictor of subclinical synovitis in the remission state of rheumatoid arthritis. Cytokine, 153, 155837. https://doi.org/10.1016/j.cyto.2022.155837
  • Mo, C., Wang, L., Zhang, J., Numazawa, S., Tang, H., Tang, X., Han, X., Li, J., Yang, M., Wang, Z., & Wei D. (2014). The crosstalk between Nrf2 and AMPK signal pathways is important for the anti-inflammatory effect of berberine in LPS-stimulated macrophages and endotoxin-shocked mice. Antioxidants & Redox Signaling, 20(4), 574–588. https://doi.org/10.1089/ars.2012.5116
  • Mohammadian Haftcheshmeh, S., & Momtazi-Borojeni, A. A. (2021). Berberine as a promising natural compound for the treatment of periodontal disease: A focus on anti-inflammatory properties. Journal of Cellular and Molecular Medicine, 25(24), 11333–11337. https://doi.org/10.1111/jcmm.17019
  • Mortazavi, H., Nikfar, B., Esmaeili, S. A., Rafieenia, F., Saburi, E., Chaichian, S., Gorji, M. A., & Momtazi-Borojeni, A. A. (2020). Potential cytotoxic and anti-metastatic effects of berberine on gynaecological cancers with drug-associated resistance. European Journal of Medicinal Chemistry, 187(4), 111951. https://doi.org/10.1016/j.ejmech.2019.111951
  • Nikfar, S., Saiyarsarai, P., Tigabu, B. M., & Abdollahi, M. (2018). Efficacy and safety of interleukin-1 antagonists in rheumatoid arthritis: A systematic review and meta-analysis. Rheumatology International, 38(8), 1363–1383. https://doi.org/10.1007/s00296-018-4041-1
  • Osterman, C. J., Lynch, J. C., Leaf, P., Gonda, A., Ferguson Bennit, H. R., Griffiths, D., & Wall, N. R. (2015). Curcumin modulates pancreatic adenocarcinoma cell-derived exosomal function. PLoS One, 10(7), e0132845.
  • Paradowska-Gorycka, A., Wajda, A., Romanowska-Próchnicka, K., Walczuk, E., Kuca-Warnawin, E., Kmiolek, T., Stypinska, B., Rzeszotarska, E., Majewski, D., Jagodzinski, P. P., & Pawlik, A. (2020). Th17/treg-related transcriptional factor expression and cytokine profile in patients with rheumatoid arthritis. Frontiers in Immunology, 11(4), 572858. https://doi.org/10.3389/fimmu.2020.572858
  • Qi, X., Zhang, J., Yuan, H., Xu, Z., Li, Q., Niu, X., Hu, B., Wang, Y., & Li, X. (2016). Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells repair critical-sized bone defects through enhanced angiogenesis and osteogenesis in osteoporotic rats. International Journal of Biological Sciences, 12(7), 836–849. https://doi.org/10.7150/ijbs.14809
  • Qiu, B., Xu, X., Yi, P., & Hao, Y. (2020). Curcumin reinforces MSC-derived exosomes in attenuating osteoarthritis via modulating the miR-124/NF-kB and miR-143/ROCK1/TLR9 signalling pathways. Journal of Cellular and Molecular Medicine, 24(18), 10855–10865. https://doi.org/10.1111/jcmm.15714
  • Rauf, A., Abu-Izneid, T., Khalil, A. A., Imran, M., Shah, Z. A., Emran, T. B., Mitra, S., Khan, Z., Alhumaydhi, F. A., Aljohani, A. S., & Khan, I. (2021). Berberine as a potential anticancer agent: A comprehensive review. Molecules, 26(23), 7368. https://doi.org/10.3390/molecules26237368
  • Rieu, S., Géminard, C., Rabesandratana, H., Sainte-Marie, J., & Vidal, M. (2000). Exosomes released during reticulocyte maturation bind to fibronectin via integrin α4β1. European Journal of Biochemistry, 267(2), 583–590. https://doi.org/10.1046/j.1432-1327.2000.01036.x
  • Rosloniec, E. F., Cremer, M., Kang, A. H., Myers, L. K., & Brand, D. D. (2010). Collagen-induced arthritis. Current Protocols in Immunology, 89(1), 15–15. https://doi.org/10.1002/0471142735.im1505s89
  • Savina, A., Furlán, M., Vidal, M., & Colombo, M. I. (2003). Exosome release is regulated by a calcium-dependent mechanism in K562 cells. Journal of Biological Chemistry, 278(22), 20083–20090. https://doi.org/10.1074/jbc.M301642200
  • Sharma, A., Sharma, R., Kumar, D., & Padwad, Y. (2020). Berberis lycium Royle fruit extract mitigates oxi-inflammatory stress by suppressing NF-κB/MAPK signalling cascade in activated macrophages and Treg proliferation in splenic lymphocytes. Inflammopharmacology, 28(4), 1053–1072. https://doi.org/10.1007/s10787-018-0548-z
  • Shen, P., Jiao, Y., Miao, L., Jh, C., & Momtazi-Borojeni, A. A. (2020). Immunomodulatory effects of berberine on the inflamed joint reveal new therapeutic targets for rheumatoid arthritis management. Journal of Cellular and Molecular Medicine, 24(21), 12234–12245. https://doi.org/10.1111/jcmm.15803
  • Shetty, S. S., Sharma, M., Kabekkodu, S. P., Kumar, N. V. A., Satyamoorthy, K., & Radhakrishnan, R. (2021). Understanding the molecular mechanism associated with reversal of oral submucous fibrosis targeting hydroxylysine aldehyde-derived collagen cross-links. Journal of Carcinogenesis, 20(1), 9.
  • Su, Y., Liu, Y., Ma, C., Guan, C., Ma, X., & Meng, S. (2021). Mesenchymal stem cell-originated exosomal lncRNA HAND2-AS1 impairs rheumatoid arthritis fibroblast-like synoviocyte activation through miR-143-3p/TNFAIP3/NF-κB pathway. Journal of Orthopaedic Surgery and Research, 16(1), 116. https://doi.org/10.1186/s13018-021-02248-1
  • Sujitha, S., Dinesh, P., & Rasool, M. (2018). Berberine modulates ASK1 signaling mediated through TLR4/TRAF2 via upregulation of miR-23a. Toxicology and Applied Pharmacology, 359(11), 34–46. https://doi.org/10.1016/j.taap.2018.09.017
  • Sujitha, S., Dinesh, P., & Rasool, M. (2020). Berberine encapsulated PEG-coated liposomes attenuate Wnt1/β-catenin signaling in rheumatoid arthritis via miR-23a activation. European Journal of Pharmaceutics and Biopharmaceutics, 149(4), 170–191. https://doi.org/10.1016/j.ejpb.2020.02.007
  • Tavasolian, F., Moghaddam, A. S., Rohani, F., Abdollahi, E., Janzamin, E., Momtazi-Borojeni, A. A., Moallem, S. A., Jamialahmadi, T., & Sahebkar, A. (2020). Exosomes: Effectual players in rheumatoid arthritis. Autoimmunity Reviews, 19(6), 102511. https://doi.org/10.1016/j.autrev.2020.102511
  • Taverna, S., Fontana, S., Monteleone, F., Pucci, M., Saieva, L., De Caro, V., Cardinale, V. G., Giallombardo, M., Vicario, E., Rolfo, C., & De Leo, G. (2016). Curcumin modulates chronic myelogenous leukemia exosomes composition and affects angiogenic phenotype via exosomal miR-21. Oncotarget, 7(21), 30420–30439. https://doi.org/10.18632/oncotarget.8483
  • Taverna, S., Giallombardo, M., Pucci, M., Flugy, A., Manno, M., Raccosta, S., Rolfo, C., De Leo, G., & Alessandro, R. (2015). Curcumin inhibits in vitro and in vivo chronic myelogenous leukemia cells growth: A possible role for exosomal disposal of miR-21. Oncotarget, 6(26), 21918–21933. https://doi.org/10.18632/oncotarget.4204
  • Tong, B., Dou, Y., Wang, T., Yu, J., Wu, X., Lu, Q., Chou, G., Wang, Z., Kong, L., Dai, Y., & Xia, Y. (2015). Norisoboldine ameliorates collagen-induced arthritis through regulating the balance between Th17 and regulatory T cells in gut-associated lymphoid tissues. Toxicology and Applied Pharmacology, 282(1), 90–99. https://doi.org/10.1016/j.taap.2014.11.008
  • Tu, J., Hong, W., Zhang, P., Wang, X., Körner, H., & Wei, W. (2018). Ontology and function of fibroblast-like and macrophage-like synoviocytes: How do they talk to each other and can they be targeted for rheumatoid arthritis therapy? Frontiers in Immunology, 9(6), 1467. https://doi.org/10.3389/fimmu.2018.01467
  • van den Berg, W. B., Joosten, L. A., Helsen, M., & van de Loo, F. A. (1994). Amelioration of established murine collagen-induced arthritis with anti-IL-1 treatment. Clinical and Experimental Immunology, 95(2), 237–243. https://doi.org/10.1111/j.1365-2249.1994.tb06517.x
  • Wang, X., He, X., Zhang, C.-F., Guo, C.-R., Wang, C.-Z., & Yuan, C.-S. (2017). Anti-arthritic effect of berberine on adjuvant-induced rheumatoid arthritis in rats. Biomedicine & Pharmacotherapy, 89(5), 887–893. https://doi.org/10.1016/j.biopha.2017.02.099
  • Wang, X.-h., Jiang, S.-m., & Sun, Q.-w. (2011). Effects of berberine on human rheumatoid arthritis fibroblast-like synoviocytes. Experimental Biology and Medicine, 236(7), 859–866. https://doi.org/10.1258/ebm.2011.010366
  • Wang, Y.-Y., Tang, L.-Q., & Wei, W. (2018). Berberine attenuates podocytes injury caused by exosomes derived from high glucose-induced mesangial cells through TGFβ1-PI3K/AKT pathway. European Journal of Pharmacology, 824(4), 185–192. https://doi.org/10.1016/j.ejphar.2018.01.034
  • Wang, Z., Chen, Z., Yang, S., Wang, Y., Huang, Z., Gao, J., Tu, S., & Rao, Z. (2014). Berberine ameliorates collagen-induced arthritis in rats associated with anti-inflammatory and anti-angiogenic effects. Inflammation, 37(5), 1789–1798. https://doi.org/10.1007/s10753-014-9909-y
  • Wu, H., Yan, S., Chen, J., Luo, X., Li, P., Jia, X., Dai, X., Wang, C., Huang, Q., Liu, L., Zhang, Y., Zhou, A., Chang, Y., Zhang, L., & Wei, W. (2016). JAK1-STAT3 blockade by JAK inhibitor SHR0302 attenuates inflammatory responses of adjuvant-induced arthritis rats and decreases Th17 and total B cells. Joint, Bone, Spine: Revue du Rhumatisme, 83(5), 525–532. https://doi.org/10.1016/j.jbspin.2015.09.002
  • Wu, H., Zhou, J., Zeng, C., Wu, D., Mu, Z., Chen, B., Xie, Y., Ye, Y., & Liu, J. (2016). Curcumin increases exosomal TCF21 thus suppressing exosome-induced lung cancer. Oncotarget, 7(52), 87081–87090. https://doi.org/10.18632/oncotarget.13499
  • Yang, J., Wang, J., Liang, X., Zhao, H., Lu, J., Ma, Q., Jing, B., & Tian, F. (2019). IL-1β increases the expression of inflammatory factors in synovial fluid-derived fibroblast-like synoviocytes via activation of the NF-κB-mediated ERK-STAT1 signaling pathway. Molecular Medicine Reports, 20(6), 4993–5001.
  • Yeo, R. W., Lai, R. C., Zhang, B., Tan, S. S., Yin, Y., Teh, B. J., & Lim, S. K. (2013). Mesenchymal stem cell: An efficient mass producer of exosomes for drug delivery. Advanced Drug Delivery Reviews, 65(3), 336–341. https://doi.org/10.1016/j.addr.2012.07.001
  • You, S. H., Yoon, M. Y., & Moon, J. S. (2021). Antioxidant and anti-inflammatory activity study of fulvic acid. Journal of Natural Science, Biology and Medicine, 12(3), 285–289.
  • Yue, M., Xia, Y., Shi, C., Guan, C., Li, Y., Liu, R., Wei, Z., & Dai, Y. (2017). Berberine ameliorates collagen-induced arthritis in rats by suppressing Th17 cell responses via inducing cortistatin in the gut. The FEBS Journal, 284(17), 2786–2801. https://doi.org/10.1111/febs.14147
  • Zhai, K. F., Duan, H., Cui, C. Y., Cao, Y. Y., Si, J. L., Yang, H. J., Wang, Y. C., Cao, W. G., Gao, G. Z., & Wei, Z. J. (2019). Liquiritin from Glycyrrhiza uralensis attenuating rheumatoid arthritis via reducing inflammation, suppressing angiogenesis, and inhibiting MAPK signaling pathway. Journal of Agricultural and Food Chemistry, 67(10), 2856–2864. https://doi.org/10.1021/acs.jafc.9b00185
  • Zhai, K. F, Duan, H., Shi, Y., Zhou, Y. R., Chen, Y., Zhang, Y. S., Gong, Z. P., Cao, W. G., Wu, J., & Wang, J. J. (2022). miRNAs from plasma extracellular vesicles are signatory noninvasive prognostic biomarkers against atherosclerosis in LDLr-/-mice. Oxidative Medicine and Cellular Longevity, 2022(8), 1–12.
  • Zhai, K., Duan, H., Wang, W., Zhao, S., Khan, G. J., Wang, M., Zhang, Y., Thakur, K., Fang, X., Wu, C., & Xiao, J. (2021). Ginsenoside Rg1 ameliorates blood-brain barrier disruption and traumatic brain injury via attenuating macrophages derived exosomes miR-21 release. Acta Pharmaceutica Sinica B, 11(11), 3493–3507. https://doi.org/10.1016/j.apsb.2021.03.032
  • Zhang, S., Chu, W. C., Lai, R. C., Lim, S. K., Hui, J. H. P., & Toh, W. S. (2016). Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthritis and Cartilage, 24(12), 2135–2140. https://doi.org/10.1016/j.joca.2016.06.022
  • Zhang, Y., Tu, B., Sha, Q., & Qian, J. (2022). Bone marrow mesenchymal stem cells-derived exosomes suppress miRNA-5189-3p to increase fibroblast-like synoviocyte apoptosis via the BATF2/JAK2/STAT3 signaling pathway. Bioengineered, 13(3), 6767–6780. https://doi.org/10.1080/21655979.2022.2045844
  • Zhao, J., Ouyang, Q., Hu, Z., Huang, Q., Wu, J., Wang, R., & Yang, M. A. (2016). A protocol for the culture and isolation of murine synovial fibroblasts. Biomedical Reports, 5(2), 171–175. https://doi.org/10.3892/br.2016.708