Cell culture & antibodies
The immortalized RA-FLS (MH7A) was purchased from ATCC. Reagents for cell culture, such as Dulbecco’s modified Eagle’s medium (DMEM), foetal bovine serum (FBS), trypsin-EDTA (0.05%), phosphate-buffered saline (PBS), and 100× penicillin/streptomycin (PS), were obtained from Gibco (Oklahoma, USA). RA fibroblast-like synoviocytes (FLSs) were used between passages three and eight, and the cell type was identified as described in our previously published work. RA-FLSs were cultured in 6-well plates (1×105 cells/well to ensure that the confluence reached 60–80%) in 2 ml of Dulbecco's modified Eagle’s medium (DMEM) supplemented with 10% FBS and 1% PSG at 37 ℃ in a 5% CO2 humidified environment. The medium was removed and washed once with PBS for subsequent experiments. The anti-TREX1 antibody (diluted 1:1000 for WB and 1:200 for IHC were purchased from Novus Biologicals, USA; the anti-cGAS antibody (1:1000 for WB) was obtained from Taiclone, China Taiwan; the anti-β-actin antibody (1:1000), anti-rabbit secondary antibody (1:2500), and anti-mouse secondary antibody (1:2500) were purchased from Santa Cruz, USA; and flow cytometry antibodies such as APC/Cyanine7 anti-rat CD45 (1:1000), FITC anti-rat CD3 (1:1000), PerCP/Cyanine5.5 anti-rat CD4 (1:1000), APC anti-rat CD8a (1:1000) and PE anti-mouse/rat/human FOXP3 (1:1000) were purchased from Biolegend, USA.
Extraction and assay kits
Total RNA was extracted using a FavorPrepTM Blood/Cultured Cell Total RNA Mini Kit (Favorgen Biotech, China). Genomic DNA was prepared by using a FavorPrep Tissue Genomic DNA Extraction Mini Kit (Favorgen Biotech, China). For Western blotting, signals were visualized by using a SuperSignal West Femto Maximum Sensitivity Substrate Kit (Thermo, USA). Lymphocyte isolation was performed with a Ficoll density gradient centrifugation kit (GE Healthcare, USA). Cell-free DNA was isolated and quantified by using a Dynabeads® SILANE Viral NA Kit (Thermo, USA) and Quant-iT™ PicoGreen ® dsDNA Reagent and Kits (Thermo, USA).
Reagents and services: Lipofectamine™ 3000 (Thermo, USA); TREX1 siRNA (Santa Cruz, USA); cGAS siRNA (Santa Cruz, USA); siRNA control plasmid (Sigma, USA); Mycobacterium tuberculosis (M. tuberculosis; Difco, USA); mineral oil (Sigma, USA); methotrexate (MTX) (LC labs, USA); Cre adeno-associated virus construction (Ubigene, China); TREX1 adeno-associated virus construction (Ubigene, China); dimethyl sulfoxide (DMSO; ACROS, USA); DAPI (Invitrogen, USA); RIPA buffer (10×, Cell Signaling Technology, USA); EDTA-free protease inhibitors, PhosSTOP inhibitor (Roche, Basel, Switzerland); TRIzol (Invitrogen, USA); FastStart Universal SYBR Green Master Rox (Roche Diagnostics, USA); Maxima™ H Minus cDNA Synthesis Master Mix (Thermo, USA); PVDF membranes (Bio-Rad, USA).
Clinical samples
Blood samples were collected from patients with RA, patients with osteoarthritis (OA), and healthy volunteers upon the provision of written informed and voluntary consent at Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou (China), and approval by the research ethics committee (GDREC 2015391H). All of the epidemiological investigations and classification of the volunteers were carried out according to the American College of Rheumatology criteria. The clinically relevant information and data are provided in Table 1.
Table 1
RA, OA, and healthy volunteers’ baseline data
| RA (N = 39) | OA (N = 25) | Healthy (N = 25) |
Age (years (mean (range))) | 53 (30–73) | 57 (30–72) | 45 (24–74) |
Sex (N (F/M)) | 24/15 | 19/6 | 20/5 |
Disease duration (years (mean (range)) | 5 (1 ~ 20) | 8 (1 ~ 23) | NA |
CRP (mg/L (mean (range))) | 20.2 (1.4 ~ 93) | 14.5 (3.13 ~ 39.5) | NA |
ESR (mm/h (mean (range))) | 49.5 (8 ~ 120) | 22.3 (8 ~ 47) | NA |
RF (> 20 IU/mL) | 31/38 | NA | NA |
CRP, C-reactive protein; ESR, erythrocyte sedimentation rate; RF, rheumatoid factor; NA, not assessed. |
DNA fragment preparation & DNA damage model
RA-FLSs were cultured in 6 cm dishes to a confluence of 60–80%. The cells were then exposed to UV light for 5 min, 10 min, and 15 min. The medium was removed and washed twice with PBS for the following experiment. RA-FLSs were harvested for DNA extraction using a DNA extraction kit (Favorgen). DNA fragmentation was performed by ultrasonication for 30 min to shear the DNA into fragments of 500 bp as determined by DNA gel electrophoresis. RA-FLSs cultured in 6-well plates were transfected with 5 µg of DNA fragments for 24–72 h in the absence or presence of Lipofectamine 3000. Furthermore, another group of RA-FLSs was also transfected with different concentrations of DNA fragments (1 µg, 5 µg, and 10 µg) with Lipofectamine 3000 for 24 h. All the medium was removed, and the cells were washed once with PBS before the next step of analysis.
siRNA transfection
The selected RA-FLSs were transfected using a Lipofectamine 3000 kit (Thermo, USA) according to the manufacturer’s protocol. Briefly, after the cells were cultured in a plate overnight to a confluence of 60–80%, the medium was aspirated and washed once with PBS. The TREX1 or cGAS siRNA plasmid for knockdown of TREX1 or cGAS was mixed with 250 µl of Lipofectamine 3000 and incubated for 15 min. Then, this mixture was mixed with Opti-MEM in a total volume of 1 ml and added to a 6-well plate for incubation for 12 h. After incubation for 4 h, another 1 ml of medium was added to the wells and incubated for 24 h. After 48 h, the cells were transfected with DNA fragments for 24 h, 48 h and 72 h. Real-time PCR was used to verify the mRNA expression of TREX1, cGAS, and inflammatory cytokines.
Gene expression analysis by real-time PCR
Total RNA from blood and cultured cells was isolated using the FavorPrep™ Blood/Cultured Cell Total RNA Mini Kit (Favorgen Biotech) according to the manufacturer’s protocol. Total RNA from frozen animal synovium and tissues was extracted with TRIzol (Invitrogen, USA). Approximately 20–40 mg of frozen tissue was mixed with 1 ml of TRIzol reagent and homogenized by at least 3 cycles in a homogenizer. Then, 100 µl of chloroform was added and shaken vigorously for 5 min, and the mixture was centrifuged at 4°C and 12,000 × g for 15 min. The supernatant from the phenol/chloroform extraction step was collected (approximately 400–500 µl) and precipitated with isopropanol (1:1.2 supernatant:isopropanol) for 30 min on ice; this mixture was then centrifuged at 12,000 × g for 15 min. The precipitate was washed with 70–80% cold ethanol. The white pellet was completely dissolved in 50 µl of RNase-free water. A UV spectrophotometer (NanoDrop Technologies, USA) was used to measure the quality and concentration of RNA.
For real-time PCR analysis, 1 µg of RNA was reverse transcribed with Maxima™ H Minus cDNA Synthesis Master Mix (Thermo, USA). The mixture was incubated at 25°C for 10 minutes and at 50°C for 15 minutes, and the reaction was then terminated by heating at 85°C for 5 minutes. The PCR mixture (total 20 µl) comprised 0.5 µl of template DNA, 0.5 µl each of the forward and reverse primers, 0.5 µl of template, 10 µl of SYBR Master Mix (Roche Diagnostics, USA), and 8.5 µl of ddH2O to bring the total volume up to 20 µl. Quantification of gene expression was performed with a ViiA 7 Real-Time PCR System (Applied Biosystems). All mRNA expression data were normalized to the reference gene β-actin using the ΔΔCT method for relative quantification.
Western blotting
RA-FLSs incubated with DNA fragments in the absence or presence of Lipofectamine 3000 were added to RIPA lysis buffer for protein lysate preparation. Proteins in the centrifuged supernatant were then separated by 10% SDS–PAGE. The proteins on the PAGE were transferred to a PVDF membrane, which was probed first with antibodies against TREX1 (1:1000), cGAS (1:1000), and β-actin (1:1000) and then with HRP-conjugated goat anti-rabbit IgG as the secondary antibody (1:5000). The immunoreactive bands were then visualized by ECL with a FluorChem R system (ProteinSimple, America). Quantitative analysis of Western blot signals was performed using ImageJ software.
SD rats used for establishment of the experimental arthritis model (AIA) and treatment
Wild-type male Sprague–Dawley (SD) rats weighing 110–120 g were purchased from SPF Biotechnology Co., Ltd. Conditional knockout (Cko) TREX1+/- SD rats weighing 80–120 g were generated using CRISPR/Cas9 by Cyagen (USA). Rats were bred and maintained at the State Key Laboratory of Quality Research in Chinese Medicines of Macau University of Science and Technology. The genotypes of TREX1 transgenic rats were identified by PCR of tail genomic DNA using two pairs of primers. The animal study was conducted according to the guidelines of the Institutional Animal Care and Use Committee of Macau University of Science and Technology and approved by the Ethics Committee of Macau University of Science and Technology (protocol code MUSTARE – 003–2020) on 23 Feb 2020. During the experiment, the animals were housed for 1 week on a 12:12 h light-dark cycle at room temperature (22 ± 1°C).
Model 1 (Inflammatory potency of DNA fragment injection in the AIA model)
SD rats were divided into 6 groups (n = 6 ~ 8 rats/group) as follows: (1) healthy control (n = 7), (2) AIA model (n = 8), (3) AIA + DNA (200 µg; n = 7), (4) AIA + DNA (50 µg; n = 7), (5) healthy control + DNA (200 µg; n = 7), and (6) healthy control + DNA (50 µg; n = 7). Mineral oil (Sigma) containing 2.5 mg/ml M. tuberculosis cells (Difco, USA) was ground and rolled intensively until the mixture turned white. DNA fragments were injected into the knee joints of rats, and 0.1 ml of the above mixture was then injected subcutaneously at the base of the tail on Day 0. Arthritis scores and hind paw volumes were evaluated and recorded every 3 days until Day 30.
Model 2 (monitoring proinflammatory cytokine levels in AIA rats injected with DNA fragments)
SD rats were injected with 100 µg of DNA fragments via the tail vein and were then subjected to AIA model establishment. Blood samples were collected at 0 min, 30 min, 2 h, 4 h, 24 h, 48 h, and 72 h for proinflammatory cytokine detection using RT–PCR.
Model 3 (monitoring proinflammatory cytokine levels in AIA rats injected with DNA fragments after the first appearance of hind paw swelling)
SD rats were divided into 4 groups (n = 6 rats/group) as follows: (1) healthy control, (2) AIA model, (3) AIA + DNA (100 µg), and (4) AIA + DNA (50 µg). Mineral oil (Sigma) containing 2.5 mg/ml M. tuberculosis cells (Difco, USA) was ground completely until the mixture turned white, indicating that it was ready for AIA model establishment. The SD rats were then injected with complete adjuvant for arthritis induction (on Day 0). In addition, the AIA rats were t injected with 50 or 100 µg of DNA fragments (sonicated DNA from rat-derived muscle tissue) via the tail vein beginning on Day 0 and continuing every 2 days until the day (Day *) when hind paw swelling first became visible. Blood samples were then collected on day (*) for proinflammatory cytokine detection using RT–PCR.
Model 4 (evaluating the anti-inflammatory effect of TREX1 overexpression in AIA rats)
SD rats were divided into 6 groups (n = 8 rats/group) as follows: (1) healthy control, (2) AIA (adeno-associated virus, AAV-EGFP), (3) AIA + methotrexate (MTX; 7.6 mg/kg/week), (4) AIA + AAV-TREX1 (tail vein injection with 1×1011 PFU), (5) AIA + AAV-TREX1 (tail vein injection with 1×109 PFU), (6) AIA + AAV-TREX1 (joint injection with 2.5×1010 PFU) and (7) AIA + AAV-TREX1 (joint injection with 2.5×108 PFU). Tail vein injection of AAV-TREX1 (1×1011 PFU) and AAV-TREX1 (1×109 PFU) was conducted 10 days before AIA induction, and joint injection of AAV-TREX1 (2.5×1010 PFU) and AAV-TREX1 (2.5×108 PFU) was performed 10 days before AIA induction. The SD rats that received AAV injection were subsequently injected subcutaneously with 0.1 ml of the mixture at the base of the tail on Day 0. Arthritis scores and hind paw volumes were evaluated and recorded every 3 days until Day 30.
Joint swelling measurement and clinical scoring
Joint swelling and clinical scores were evaluated daily from the first injection (Day 0 for rats) until the animals were sacrificed. The investigator was blinded to the group allocation when measuring joint swelling. In the AIA model, joint swelling in the rats was evaluated by measuring the volume of the hind paws with a plethysmometer (Ugo Basile, Italy; or Kent Scientific Corp, Connecticut, USA) and calculating the average volume every 3 days. Animals that died during the experimental period were excluded from the analysis. Clinical scores for each hind paw and forepaw of the rats were determined following the standard evaluation process for clinical scoring: the severity of arthritis can be objectively inspected in four paws and is scored for each paw on a scale of 0 ~ 4 according to the arthritis index described in Table 2.
Table 2
Score | Symptom |
0 | No evidence of swelling and erythema of the joints, including the small joints of the forepaw and the phalangeal joint and the large joints such as the wrist and ankle. |
1 | Mild swelling and erythema of the ankle joint. |
2 | Mild swelling and erythema extending to the small joints. |
3 | Severe swelling and erythema of large joints. |
4 | Severe swelling and erythema encompassing the small and large joints. |
Micro-CT analysis
At the end of the treatment period, the rats were humanly sacrificed, and the left hind paw of each rat was amputated, fixed with 4% PFA, and then scanned using an in vivo Micro-CT scanner (SkyScan 1176, Bruker, Belgium). The following scanning parameters were used to obtain high-quality images of the joints of the rats: 35 µm resolution, 85 kV tube voltage, 385 µA tube current, 65 ms exposure time, rotation step over 360° 0.7, and a 1 mm Al filter. The images were reconstructed using NRecon software (Bruker Micro CT, Belgium). The Micro-CT score was obtained from five disease-related indices for Micro-CT analysis of the calcaneus: bone mineral density, bone volume fraction, cortical mineral density, trabecular number, and total porosity.
Cell-free DNA (cfDNA) extraction and measurement
For extraction of cfDNA in the plasma of rats, peripheral blood samples of rats were collected on Day 30 in EDTA-containing tubes. Then, the blood samples were centrifuged at 400×g for 10 min at 4°C, and the plasma fraction was recentrifuged at 12,000×g for 10 min at 4°C to remove cell debris and was then stored at − 80°C for analysis. cfDNA was extracted from 100 µL of plasma using a Dynabeads® SILANE Viral NA Kit. The concentration of cfDNA was determined with Quant-iT™ PicoGreen ® dsDNA Reagent and Kits.
Haematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) of rat tissue samples
Experimental AIA rats were sacrificed on Day 30, and the synovium and organs, including the brain, heart, kidneys, liver, spleen, thymus, and joint tissues, were collected for weighing and H&E staining. Synovial samples were immediately frozen in liquid nitrogen and stored at -80°C. Organs and joint tissues were fixed with 4% paraformaldehyde for 24 h and were then dehydrated and embedded in paraffin blocks at 60°C. Six-micrometre-thick sections were dehydrated, deparaffinized, and rehydrated according to standard protocols. Synovial, joint, and organ sections were incubated with the anti-TREX1 primary antibody (1:200, Novus Biologicals, USA) at 4°C overnight. After washing, the slides were incubated with streptavidin-conjugated horseradish peroxidase (BioSite Histo Plus (HRP) polymer anti-rabbit kit; Nordic BioSite, Sweden) for 1 h. They were then mounted with FluorSave™ Reagent (Millipore, USA). Images were acquired by light microscopy (Leica DM2500, Germany). Joint and organ sections from AIA rats were subjected to H&E staining. Images were acquired by light microscopy (Leica DM2500, Germany).
Generation of TREX1 conditional knockout (Cko TREX1-/-) SD rats
LoxP-E2-loxP-mediated knockout of Trex1 in SD rats was accomplished by CRISPR/Cas-mediated genome editing. The rat TREX1 gene (GenBank accession number: NM_001024989.1, Gene ID: 100049583) is located on chromosome 8. Two exons have been identified, with the ATG start codon and TAA stop codon in exon 2. Exon 2 was thus selected as the region for conditional knockout. Deletion of exon 2 was expected to result in loss of function of the rat Trex1 gene by inducing a frameshift of downstream exons. The loxP sequence was inserted into the donor vector upstream and downstream of exon 2 by homology-directed repair. gRNA targeting vectors were constructed and confirmed by sequencing, and a donor vector with flanking of homologous arms was also constructed. Cas9 mRNA and gRNA generated by in vitro transcription were co-injected with the donor vector into fertilized eggs to obtain KI rats. The pups were genotyped by PCR, and sequence analysis was then performed.
Genotyping of TREX1-deficient rats
Genomic DNA was isolated from rat tails. Genotyping of TREX1 was performed by polymerase chain reaction (PCR). PCR amplification was performed using primers. Primer sequences: F-AGAGACTCACGGGCTTGTTTGAATC, R- GAGAGTAAGGCAGGCCAGTGGC. Each 25 µL PCR mixture consisted of 4 µL of genomic DNA (~ XX µg), 1 µL of each primer (10 µmol/L), 12.5 µL of 2 × Taq PCR Master Mix (constituents: 0.1 U Taq polymerase/µL, 500 µM each dNTP and PCR buffer), and 6.5 µL of ddH2O (DNase/RNase-free). PCR was performed with an initial denaturation step at 95°C for 5 min, followed by 35 cycles of denaturation at 95°C for 30 s, annealing at 60°C for 30 s and elongation at 72°C for 35 s. The amplified PCR products were subjected to electrophoresis on a 1.0% agarose gel with 0.5 µg/mL ethidium bromide (EB) prior to visualization by ultraviolet light.
Establishment of the AIA model using Cko TREX1-/- rats
Model 1 (Hind paw joint injection of Cre adeno associated virus into Cko TREX1-/- rats)
TREX1 conditional knockout (Cko TREX1−/−) rats were divided into 6 groups (n = 6 ~ 7 rats/group) as follows: (1) Cko TREX1−/− (healthy control, n = 7), (2) Cko TREX1−/− (AIA, n = 6), (3) Cko TREX1−/− (AIA + high concentration (2.5×1010 PFU) of Cre adeno associated virus (CreH), n = 6), (4) Cko TREX1−/− (AIA + low concentration (2.5×108 PFU) of Cre adeno associated virus (CreL), n = 6), (5) Cko TREX1−/− (healthy control + CreH, n = 6), and (6) Cko TREX1−/− (healthy control + CreL, n = 6). Rats in the three healthy control Cko TREX1−/− groups and three AIA Cko TREX1−/− groups were injected in the hind limb joints with nucleic acid-free water or DNA fragments (200 µg), respectively, on Day 0. Approximately 10 days before the Cko TREX1−/− rats were injected with complete adjuvant for arthritis induction (on Day 0), they were injected in the hind limb joints with Cre adeno associated virus s (2.5 × 108 PFU (CreL) or 2.5 × 1010 PFU (CreH)) to facilitate knockout of the TREX1 gene.
Model 2 (tail vein injection of Cre adeno associated virus into Cko TREX1-/- rats)
Cko TREX1−/− rats were divided into 6 groups (n = 6 ~ 12 rats/group) as follows: (1) Cko TREX1−/− (healthy control, n = 7), (2) Cko TREX1−/− (AIA, n = 12), (3) Cko TREX1−/− (AIA + CreH, n = 7), (4) Cko TREX1−/− (AIA + CreL, n = 8), (5) Cko TREX1−/− (healthy control + CreH, n = 8), and (6) Cko TREX1−/− (healthy control + CreL, n = 6). Rats in the three healthy control Cko TREX1−/− groups and the three AIA Cko TREX1−/− groups were injected via the tail vein with nucleic acid-free water or DNA fragments (100 µg), respectively, on Day 0. Approximately 10 days before the Cko TREX1−/− rats were injected with complete adjuvant for arthritis induction (on Day 0), they were injected via the tail vein with Cre adeno associated virus (1 x 109 PFU (CreL) or -1 x 1011 PFU (CreH)) to facilitate knockout of the TREX1 gene.
On Day 30, all rats were sacrificed, and blood, organs and joint tissue were collected for biochemical assays.
Lymphocyte isolation, intracellular cytokine staining, and flow cytometric analysis
Peripheral blood mononuclear cells (PBMCs) were isolated from AIA rats using a standard Ficoll density gradient centrifugation kit (GE Healthcare). T cells were stained as indicated with the following anti-rat CD monoclonal antibodies (mAbs): APC/Cyanine7 CD45, FITC CD3, PerCP/Cyanine5.5 CD4, and APC CD8a. For Treg cell staining, cells were surface stained for CD3, CD4, and CD45; washed, fixed, and permeabilized using a Foxp3 Staining Buffer Set; and then immunostained with PE anti-mouse/rat/human FOXP3 antibodies (all reagents were obtained from BioLegend, Beijing). Cells were acquired using a FACSAria flow cytometer (BD Bioscience) and further analysed using FlowJo v10 software (TreeStar, Inc.).
Statistical analysis
For analysis of two groups, Student's t test was used. One-way analysis of variance was used to analyse differences among 3 or more groups. All results are expressed as the means ± SEMs. GraphPad Prism 9.0 software (GraphPad Software, USA) was used for statistical evaluation of the data. p < 0.05 was considered significant.