Human subjects research
All patients included in this study signed a written informed consent form for the use of abandoned specimens (knee meniscus, cartilage, synovium, and synovial fluid) and publication of any potentially identifiable images or data, as approved by the Ethics Committee of XXX Hospital (2009-06, 2020 − 126). We included patients with RA who met the 1987 American Rheumatism Association criteria28 and the 2010 ACR/EULAR criteria29.
For patients with RA who underwent systemic treatment without remission of severe knee pain or knee deformity (knee varus or knee valgus), we performed knee arthroplasty and obtained meniscus, cartilage, synovium, and synovial fluid samples. For patients with OA who met Kellgren-Lawrence Ⅲ or Ⅳ grading30 and had severe knee pain which could not be relieved by NASIDs, we performed knee arthroplasty and obtained knee tissue samples. For the control knee synovium samples, we obtained healthy knees by thigh amputation or hemipelvectomy. Radiological data from patients with OA and RA were used for radiological analysis, and samples from normal, OA, and RA knees were used for histological analysis, single-cell RNA sequencing, cell cytometry, cell culture, in vitro and in vivo experiments, and transcriptomic RNA sequencing.
The cohort was established in 2013 for long-term follow-up in RA cohort studies. All patients with RA underwent knee Parker-Pearson fine needle biopsy at the onset of knee symptoms (knee pain and swelling) to obtain knee synovium tissue samples. After the biopsy, all patients underwent systematic antirheumatoid treatment. By the end of May 2024, 17 patients completed the follow-up period. We categorised the patients into non-TKA and TKA groups based on whether they underwent TKA. For the TKA group, the follow-up endpoint was set at the time of surgery; for the non-TKA group, the follow-up endpoint was set at May 2024. Knee joint function was assessed using the KSS score23 for both groups.
Radiological analysis of human knee data
An X-ray in the anteroposterior view in the standing posture was used to measure the medial joint space and the lateral joint space by selecting the minimum segment between the medial and lateral compartments 31. All measurements were based on a real-world plotting scale. MRI with a T2-weighted sequence was used for meniscal measurement. In the coronal sequence, the image plan in which the medial tibial spine volume was maximal was selected for measuring the thickness and width of the meniscal body segment, whereas in the sagittal sequence, the image plan on the midpoint of the medial/lateral compartment was selected for measuring the thickness and width of the medial/lateral meniscal anterior and posterior horn32.
Histological morphology analysis and scoring of human synovium and meniscus
For histological sections, fresh meniscus, cartilage, and synovial tissues obtained from knee arthroplasty surgery (for RA and OA subjects) were fixed in 4% paraformaldehyde and embedded in paraffin. Human knee synovial fluid was centrifuged at 2000 rpm for 5 min, and the deposit was fixed in 4% paraformaldehyde and embedded in paraffin. All paraffin-embedded specimens were dissected into 5 µm sections and stored at room temperature. Sections of the human synovium and meniscus were stained with haematoxylin and eosin (HE), and Safranin-O-Fast Green. The lining (cell) layer of the synovium was identified on HE staining as the membrane facing the joint cavity, characterised by ellipsoidal cells arranged in layers33. Synovial sections were scored using the total Krenn synovitis score (ranging from 0–9 points) and the enlargement of the synovial lining cell layer component of the Krenn synovitis score (ranging from 0–3 points)19.
Meniscal sections were scored using the Pauli degeneration score17 (ranging from 0–18 points). Synovium invasion area on the meniscus was defined as the area containing limited clustered or widely infiltrated cells, usually accompanied by new angiogenesis. The area was calculated as the ratio of the invasion area to the total meniscal area using ImageJ software (version 1.54).
Immunohistochemistry and mIHC
For immunohistochemistry (IHC), antigen retrieval was performed at pH 9.0 using Tris-EDTA for 20 min by microwave heating. Sections were incubated with primary antibodies of CD142 (Bioss, 1:200), PRG4 (Abcam, 1:200), THY1 (Proteintech, 1:200), and ABCC4 (Proteintech, 1:200) at 4℃ overnight, followed by incubation with anti-rabbit horseradish peroxidase (HRP) secondary antibody (Servicebio, 1:200) at room temperature for 50 min. HRP staining was performed using DAB Peroxidase HRP Substrate (Servicebio).
For mIHC, antigen retrieval was performed at pH 9.0 using Tris-EDTA for 20 min by microwave heating. The main steps of mIHC are as follows: sections were incubated with primary antibody at 4℃ overnight, and then with anti-rabbit HRP secondary antibody at room temperature for 50 min, followed by tyramide signal amplification (TSA) staining at room temperature for 10 min. Antigen retrieval was performed again for the next primary antibody incubation. Primary antibodies against PRG4 (Abcam, 1:1000), CD146 (Proteintech, 1:1000), CD142 (Cell Signaling Technology, 1:600), and THY1 (Abcam, 1:600) were incubated. IF440-TSA (anti-CD146, Servicebio, 1:500), iF488-TSA (anti-CD142, Servicebio, 1:500), iF555-TSA (anti-PRG4, Servicebio, 1:500) and iF647-TSA (anti-THY1, Servicebio, 1:500) were used for TSA staining. DAPI staining (Servicebio) was performed last to label nuclei.
Quantitative and spatial analysis of mIHC
Human synovial sections stained with mIHC were scanned using Akoya whole-slide multispectral imaging in fluorophore Spectral DAPI (excitation: 368; emission: 461), Opal Polaris 480 (excitation: 450; emission: 500), Opal 520 (excitation: 494; emission: 525), Opal 570 (excitation: 550; emission: 570), and Opal 690 (excitation: 6676; emission: 694). Three fields (923 µm × 692 µm) in each section were chosen for analysis, and their data were summed as one sample. Akoya InForm (version 1.6) loaded the chosen fields to output phenoptic data, followed by tissue segmentation, cell segmentation, and phenotyping. For tissue segmentation, the lining and SLs were segmented using haematoxylin and corresponding HE staining. For cell segmentation, the minimum nuclear size was set at 10, the cytoplasm thickness was set at 5, and the membrane search distance was set at 10. For cell phenotyping, 20 cells for each fluorophore were manually chosen for classifier training, and the classifier was used for field phenotyping to label each segmented cell. The R package PhenoptrReports (version 0.3.3) processed the phenoptics data and calculated all phenotyping cells in the LL and SL of each section and the nearest distance between each pair of phenotyping cells. The results from three fields were summed and averaged for each sample.
Human meniscus, cartilage, and synovium tissue processing for cell suspensions and cell culturing
Fresh meniscus, cartilage, and synovial tissues were obtained from knee arthroplasty (for RA and OA subjects) and thigh amputation or hemipelvectomy (for control subjects). For flow cytometry, cell culturing and RNA sequencing, fresh synovium tissues were dissected in 2 mm pieces and then digested in collagenaseⅠsolution (2 mg/mL, Gibco; +10% FBS, Gibco; +1% penicillin-streptomycin, Gibco) for 2 h in 37℃, after which solution was filtered by 70 nm cell strainer (BIOFIL) and centrifuged at 1000 rpm for 5 minutes to obtain cells and made into single cell suspensions. Meniscal and cartilage tissues were dissected in 2 mm pieces and then digested in collagenase P solution (2 mg/mL for meniscus and 0.25 mg/mL for cartilage, Roche; +10% FBS, Gibco; +1% penicillin-streptomycin, Gibco) for 6–8 h in 37℃ for cell suspensions.
For human primary meniscal cells and SF, we cultured them in Dulbecco’s modified Eagle’s medium/Hams F12 (DMEM/F-12, Gibco) with 10% FBS and 1% penicillin-streptomycin in an incubator at 37℃ and 5% CO2.
Flow cytometry and synovial fibroblast collection
For the flow cytometric analysis of human synovial cells, red blood cell lysis buffer (Solarbio) was added to the cell suspensions for 10 min at room temperature, and the suspensions were centrifuged, washed, and resuspended. After removing red blood cells, synovial cells were stained with antibodies against CD31 (BV421, BioLegend, CA, USA), CD45 (APC, BioLegend), CD142 (PE, BioLegend), and 1% BSA in PBS for 20 min at room temperature. 7-AminoactinomycinD (7-AAD, BioLegend) was added to cell suspensions and cells were passed through a 100 µm filter. Data were acquired using BD FACSverse and analysed using FlowJo (version 10.8). Based on the above protocol, we sorted CD31-/CD45- living fibroblasts from primary human synovial cells and collected CD142 + and CD142- fibroblasts using BDJAzz for further in vivo and in vitro experiments.
Generation and analysis of single-cell RNA sequencing data
Fresh meniscus, cartilage, and synovial tissues obtained from the same RA knee by TKA were made into cell suspensions, in which meniscal cells, chondrocytes, and synovial cells were captured with 10x Genomics based on the NovaSeq platform. Fastp was applied with a default parameter for filtering the adaptor sequence and removing low-quality reads34. Umi tools were used for single-cell transcriptome analysis to identify the cell barcode whitelist, extract the cell barcode UMIs, and calculate the cell expression counts based on the filtered clean FASTQ data35. Downstream analysis was performed using the Seurat R package (version 4.0.2) as follows: cells with > 20% mitochondrial reads, < 200 genes, or < 200 UMI were excluded from the analysis. We merged cells from the meniscus, cartilage, and synovium, and the per-cell counts were normalised and scaled. The fastMNN function (k = 5, d = 50, approximate = TRUE) in the R package scran (version 1.12.1) was used to apply the mutual nearest-neighbour method to correct for batch effects among samples. The first ten principal components were retained for the UMAP projection. GraphCluster and K-means were used for cell clustering (resolution set at 0.8 for the full analysis), and the Wilcoxon rank-sum test was used for marker gene analysis (min.pct = 0.1, logfc. threshold = 0.25). We identified the meniscal cells as COL1A1+/CEMIP+, chondrocytes as ACAN+/COL2A1+, SF as PRG4+/COL1A1+, macrophages as CD14+/LYZ+, smooth muscle cells as MYL9+/NOTCH3+, T cells as CD3D+/CD8A+, endothelial cells as MCAM+(CD146+)/PECAM1+, and mast cells as CD79A+/ TPSB2+. SF were re-analysed to identify subclusters in the fastMNN-based UMAP zoom with a resolution set at 1.8, and marker genes were calculated (min.pct = 0.1, logfc. threshold = 0.25).
Cell-to-cell interaction analysis
After identifying the three subclusters of SF, we performed a cell-to-cell interaction analysis using the R package CellChat (version 1.6.1). We created a CellChat object with clusters of meniscal cells and chondrocytes and three subclusters of SF. CellChat database for humans (CellChatDB.human) was loaded, and the “Secreted Signaling” category was chosen for analysis. Overexpressed genes and interactions were identified and projected onto the protein-protein interactions (PPI) of the CellChat object. We then computed the communication probability based on the algorithm’s truncated mean (cutoff = 20%) and filtered out the cell-to-cell communication if there were < 10 cells in certain cell groups, which were aggregated into the final cell-to-cell communication network. Three subclusters of SF were set as communication senders, while meniscal cells and chondrocytes were set as communication receivers, and the interaction numbers were output for a heatmap and bubble plot.
Transwell assays for SF
CD142 + and CD142- fibroblasts were used for in vitro experiments. For transwell assays, we used 8.0 µm PET membrane in a 24-well format (Corning) for migration assays and 8.0 µm Matrigel coated PET membrane in a 24-well format (Corning) for invasion assays. Note that MK571 (Selleck) was dissolved in DMSO, and when added to the experiment system of DMEM/F-12, the concentration of MK571 was 5 µM, and DMSO was lower than 0.1%. We added 1×105 fibroblasts and 200 µL DMEM/F-12 (0% FBS, 1% penicillin-streptomycin, and 5µM MK571 for the specified group) in the upper chamber, and 600 µL DMEM/F-12 (10% FBS, 1% penicillin-streptomycin) in the nether chamber. Twenty-four well plates were placed in a 37℃ incubator for 24 h for migration assay and 48 h for invasion assay.
Co-culture of SF-meniscal cells
Primary human RA MCs were used in this study. We used 0.4µm PET membrane in 6-well format (Corning). We added 1×105 CD142 + or CD142- fibroblasts, 1 ml DMEM/F-12 (10% FBS, 1% penicillin-streptomycin, and 5 µM MK571 for the certain group) in the upper chamber, 1×105 meniscal cells, and 2 mL DMEM/F-12 (10% FBS, 1% penicillin-streptomycin) in the nether chamber. Six well plates were placed in a 37℃ incubator for 72 h; then, meniscal cells were washed with PBS and extracted to total RNA (details written below) for further analysis.
Mouse OA and RA models establishment
The animal study design was reviewed and approved by the Ethics Committee of XXX Hospital (2020-B0270). We created OA models by surgically destabilizing the medial meniscus (DMM) in C57BL/6 male mice at 8 weeks of age14. After 8 weeks (16 weeks of age), DMM mice were sacrificed to obtain knee samples.
We created an RA model with CIA in DBA/1 male mice36. Mice were immunised with 200 µg chick typeⅡcollagen (CⅡ, Chondrex) emulsified 1:1 in complete Freund’s adjuvant (CFA, Sigma) at 8 weeks of age, and were boosted 3 weeks later with 200 µg CⅡemulsified 1:1 in Freund’s incomplete adjuvant (IFA, Chondrex). At 16 weeks of age, CIA mice were sacrificed to obtain knee samples.
Mouse invasive SF arthritis model establishment
Human RA CD142 + and CD142- SF were used to create an invasive synovial fibroblast arthritis model. Living CD142 + and CD142- fibroblasts were first labelled with autofluorescence using CellTrace CFSE dye (Thermo Fisher Scientific), which diffused into cells and bound covalently to intracellular amines, resulting in fluorescent staining (excitation: 488 nm; Ex/Em: 592/571 nm). Resuspended fibroblasts in PBS were then added to the CFSE dye at a working concentration of 5 µM and incubated for 20 min at room temperature, protected from light. Five times the original staining volume of the culture medium was added to the fibroblasts, incubated for 5 min, centrifuged, and resuspended in DMEM/F-12. For intra-articular injection, each knee joint was injected with 4µl cell suspensions (5×104 fibroblasts in DMEM/F-12, without FBS and penicillin-streptomycin), while in MK571 group, each knee joint was injected 2µl cell suspensions (5×104 fibroblasts in DMEM/F-12, without FBS and penicillin-streptomycin) plus 2 µL MK571 (10 µM).
DBA/1 male mice were anaesthetised using isoflurane. We made a 1 cm vertical incision above the ligamentum patellae to fully expose it, then used a microinjector at a range of 0–10 µL to fetch cell suspensions. The needle tip was parallel to the platform and pointed at the midpoint of the medial or lateral side of the ligamentum patellae. The knee joint was then penetrated, and cell suspensions were injected until visible swelling was observed on the joint without spillage. The tip of the needle was withdrawn, the penetration point was pressed for 2 min using a cotton ball, and the skin was sutured. Intra-articular injection was administered once a week at 8, 9, 10, and 11 weeks of age. Mice were sacrificed at the age of 12 weeks for paraffin-embedded slices and at 16 weeks for both frozen OCT and paraffin-embedded slices. OCT embedded slices were stained with DAPI and directly observed under a fluorescence microscope to detect the invasive fibroblasts. OCT and paraffin slices were prepared according to the protocol described above.
Histological analysis of mouse knee slices
The knee joints of mice were decalcified for 3 weeks with 10% EDTA solution (pH 7.2, Solarbio) and embedded in paraffin. Consecutive slices were made (paraffin in 5 µm thick, OCT in 10 µm thick) at the knee lateral compartment in the sagittal view. We used the degeneration score and invasion area to assess the destruction of the mouse meniscus, as discussed above. The Krenn score was calculated to assess synovitis in the mouse knees. To evaluate cartilage damage, the OARSI score was calculated, with a maximum of six scores37. The scores of three slices (one slice after an interval of nine slices) were summed and averaged as one joint, whereas both the left and right joints were summed and averaged as an independent sample.
Quantitative reverse transcription PCR
RNA was isolated from the cell suspensions using an RNA isolation kit (EZ Bioscience) according to the manufacturer’s instructions. cDNA synthesis was performed for all samples (500 ng of RNA was transcribed) using the RT Premix cDNA synthesis kit (Accuracy Biology). Reverse transcription quantitative PCR (RT-PCR) was performed using the SYBR GreenPro Taq qPCR kit (Accuracy Biology) on a real-time PCR detection system (Roche LightCycler480Ⅱ). The gene expression levels were quantified using the 2 − ΔΔCt method. Each primer (forward, F; reverse, R) was designed based on the sequences available in the NCBI database (Supplementary Data Table 2).
Transcriptomic RNA sequencing data
Human RA primary CD142 + and CD142- fibroblasts with three independent samples sorted by flow cytometry from the RA knee synovium obtained by TKA surgery were used to extract RNA. We applied fastp with the default parameters to filter the adaptor sequence and remove low-quality reads. The clean reads were aligned to 9606 (Taxonomy ID) genomes (version: GRCh38) using Hisat2. HTseq was used to calculate the gene counts. Reads/fragments per kilobase million reads (RPKM/FPKM) were used to standardise expression data. We applied the DEseq2 algorithm to filter the differentially expressed genes and then filtered the fold change and FDR under the following criteria: i) log2FC > 0.585 or < -0.585; ii) FDR < 0.05. Volcano plots and heat maps were drawn using R based on the analysis of differentially expressed genes, and the colour was determined using the filtering criteria. GO analysis was performed to elucidate the biological implications of unique genes in the significant or representative profiles of the differentially expressed gene in the experiment38. We downloaded from the National Center for Biotechnology Information (NCBI; http://www.ncbi.nlm.nih.gov/), UniProt (http://www.uniprot.org/), and GO (http://www.geneontology.org/) databases. Fisher's exact test was used to identify significant GO categories, and FDR was used to correct p-values.
Statistical analysis
Statistical analyses were performed as described in each section using GraphPad Prism 9 software. Data are presented as mean ± s.d. from at least three independent experiments. Spearman’s correlation analysis was used to test the correlations between ordinal variables. Differences were considered significant at P < 0.05. Multiple testing corrections were applied where appropriate.