Human samples
In this study, the psoriatic skin biopsies were obtained from the lesional skin of psoriasis patients, and normal skin biopsies were obtained from surgical discard specimens of healthy donors. Both patients and healthy donors were strictly screened and were free from medical treatment within 3 months before the recruitment. The study was approved by Ethics Committee of Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine (Shanghai, China) and was performed in accordance with the Declaration of Helsinki. All patients and donors signed consent forms.
Animal experiments
Male C57BL/6 mice (8-12 weeks old; shanghai laboratory animal center) were maintained under pathogen‐free conditions with ad libitum food and water. The mice in the treatment group were treated every day with a topical IMQ cream (62.5 mg in 5%; Sichuan Mingxin Pharmaceutical Co., Ltd) on the backs, whereas the mice in the control group were treated with Vaseline Lanette cream. All mice were observed for the following 7 consecutive days as previously described [32]. As for exosome treatment, 50 µg exosomes derived from different cell groups were subcutaneously injected into mice on Day 0, 2, 4, and 6. PASI score was calculated based on erythema, epidermal thickness, and scaling (score range, 0‐4), as previously described [33]. At day 8, mice were sacrificed, and tissues were obtained and analyzed as described below.
The animal experiments were conducted according to the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH publication no. 85 − 23, revised 1996) and were approved by the Animal Care and Use Committee of Shanghai Jiao Tong University School of Medicine. All studies involving animals are reported in accordance with the ARRIVE guidelines for reporting experiments involving animals.
Cell culture
Human monocytic cell line THP-1, human peripheral leukemia T cell line Jurkat and mouse lymphoma cell line EL4 were cultured and maintained in complete RPMI 1640 (Gibco, Life Technologies), supplemented with 10% (v/v) heat-inactivated fetal bovine serum (FBS) and 1% penicillin/streptomycin. THP-1 cells were differentiated into macrophages by treatment with 100 nM phorbol myristate acetate (PMA; Sigma–Aldrich, Poland) for 24 h, and then the adherent M0 type macrophages were polarized towards M2 macrophages by treatment with 20 ng/mL of IL-4 for 48h.
Mouse monocyte macrophage line RAW264.7 and human embryonic kidney epithelial cell line 293T were cultured in Dulbecco’s modified Eagle’s medium (DMEM, Gibco, Life Technologies) containing 10% heat-inactivated FBS and 1% penicillin/streptomycin. RAW264.7 macrophages were either treated (M2) or untreated (M0) with 20 ng/mL IL-4 for 48 h. For all experiments, cells were grown at 37°C in a humidified atmosphere containing 5% (v/v) CO2.
Lentivirus and cell infection
For overexpression assay, ANXA1 sequence was cloned into pHR lentiviral vector (pHR-ANXA1) with GFP gene. Empty pHR vector (pHR-NC) was used as negative control. pHR-ANXA1 or pHR-NC, delta8.9, and pMD2.G were co-transfected into 293T cells with PEI reagent. The supernatants containing viruses were harvested at post-transfection 24h, 48 h, and 72h. Before infection, T cells were expanded for 4 days in 48-well plate. Next, T cells were diluted into 1 million/well and infected with lentiviruses, in the presence of 8µg/mL polybrene (Sigma-Aldrich H9268) for 24h. 3 days post-infection, all the cells were washed with FACS buffer to sort the GFP-positive cells.
Isolation and identification of exosomes
For exosomes isolation, conditioned medium (CM) was prepared by incubating cells in media containing exosome-depleted FBS by ultracentrifugation at 100,000 × g at 4°C for at least 4 h, and then pre-cleared by centrifugation at 500 × g for 15 min and 10,000 × g for 20 min. Exosomes were isolated by ultracentrifugation at 100,000 × g for 210 min and washed in PBS using the same ultracentrifugation conditions. When indicated, DiI (1,1′-Dioctadecyl- 3,3,3′,3′-tetramethylindocarbocyanine perchlorate; Sigma) was added into the PBS at 1 µM. After incubation for 20 min, the excess dye was removed by washing spin. The pelleted exosomes were resuspended in ~ 100 µL of PBS and subjected to further treatments.
We named JA- EXO or JC-EXO as the exosomes isolated from Jurkat cells infected with ANXA1 or negative control lentivirus. We named EA- EXO or EC-EXO as the exosomes isolated from EL4 cells infected with ANXA1 or negative control lentivirus. At last, we named JAM or EAM as the engineered extracellular vesicles which were derived from JA-EXO or EA-EXO co-extruted with M2 macrophage membrane.
Fabrication of engineered extracellular vesicles
To fabricate M2 macrophage membrane vesicle, macrophages induced by IL-4 for 48h were collected in cell membrane extraction buffer (Beyotime, China). Then the cells was repeatedly freeze-thawed, centrifuged at 8000 ×g for 15 min and sonicated for 2 min at 42 kHz frequency with a power input of 100 W [34]. To investigate the contribution of M2 macrophage membrane input to the integrity and stability of engineered extracellular vesicles, engineered extracellular vesicles with different protein ratios of exosomes to M2 macrophage membrane vesicles (m:m, 1:1, 2:1, 3:1) were prepared and serially extruded through 100 nm polycarbonate porous membranes for 10 times each using an Avanti mini extruder (Avanti Polar Lipids, Alabaster, AL, USA) at room temperature. Then, the size distribution of engineered extracellular vesicles was tested to determine the final amount of M2 macrophage membranes to be used.
Characterization of nanoparticles/extracellular vesicles
Transmission electron microscopy (TEM) (JEOL JMPEG- PTMC-1230, Japan) and Nano tracking analysis (NTA) were performed using NanoSight (Malvern, Malvern, UK) to measure extracellular vesicles’ size and zeta potential. For the stability of the extracellular vesicles, samples were stored at − 80 ℃ for 6 months and then measured by NanoSight. Proteins extracted from extracellular vesicles or cells were determined by western blotting (WB). The BCA protein assay kit was used to quantify the extracellular vesicles.
Cellular uptake and endocytic mechanisms in vitro
Macrophages were seeded at a density of 2 × 104 cells/well in six-well plates, incubated for 12h, checked under the microscope for confluency and morphology. After being pre-incubated with Hank’s balanced salt solution (HBSS) for 15min, macrophages were incubated with DiI-labeled JAM at the final concentration from 0 to 150 µg/mL at 37°C for 0–48 h.
For cellular uptake mechanism assay, macrophages were seeded in six-well plates. After checking the confluency and morphology, inhibitory agents including sucrose (300mM), 5-(N, N-dimethyl) amiloride hydrochloride (DMA, 100µM) and Genistein (200µM) were added into each well and incubated for 30min, respectively. Then the compounds were withdrawn from the wells, and DiI-labeled JAM was added at the final concentration of 120µg/mL. After incubation, the cells were visualized under fluorescent microscope (Leica, Germany).
Real-time q-PCR
Total RNA was extracted from cells by using TRIzol reagent (Thermo, Shanghai, China). The purity of the isolated RNA was determined by the optical density 260/280 ratio using the NanoDrop ND-2000 (Thermo Scientific). The isolated RNA was reverse transcribed by using the Hiscript™ III 1st Strand cDNA Synthesis kit (Vazyme, China). qPCR was performed using the SYBR qPCR Master Mix Kit (Vazyme, China) following the manufacturer’s instructions. The relative expression levels of the genes were normalized to that of GAPDH by using 2 − ΔΔCt.
Macrophage polarization identification
The polarization state of macrophages in vitro was further confirmed by immunofluorescence assay. The details of the experimental procedures are the same as the previous studies. Briefly, after THP-1 cells were treated with 100nM PMA (Sigma– Aldrich, Poland) for 24h, the adherent macrophages were washed with PBS and co-cultured with PBS, JC-EXO, JA-EXO or JAM, respectively. 48h later, cells were washed three times with PBS and then fixed with 4% paraformaldehyde for 20 min. Then, the cells were blocked with 3% BSA and incubated with rabbit-anti-iNOS or rabbit-anti-Arg-1 antibody overnight at 4°C. After washing three times with PBS, the samples were stained with the secondary antibody. Fluorescence signals were detected by a confocal laser scanning microscope (Leica, Germany).
Cell Counting Kit-8
Cell Counting Kit − 8 assay was adopted to test the influence of different exosomes on the number of macrophages. The cells were seeded onto 96-well flat-bottomed plates with a density of 2500 cells/well and then were incubated in 5% CO2 atmosphere at 37°C, followed by samples treatment for different duration. After incubation, the medium was added with 10µL of CCK8 solution for each well. The absorbance value was measured at 450 nm using microplate reader (Thermo, USA).
Western blotting
Isolated exosome pellet or cultured cells were lysed in radio-immunoprecipitation assay (RIPA) buffer supplemented with complete protease inhibitor cocktail tablets (Roche, Basel, Swiss) and incubation at 95◦C for 5 min. Lysates of cells or extracellular vesicles were separated by 8–12% SDS-polyacrylamide gels, transferred to PVDF membranes. The membranes were blocked for 2 h in 5% no-fat milk buffer, then incubated with primary antibodies at 4°C overnight. Protein expression levels were semi-quantitatively analyzed using densitometry analysis.
Anti-CD63 (ab59479), anti-Hsp70 (ab2787), and anti-Alix (ab117600) were purchased from Abcam (Cambridge, MA). Anti-ANXA1 (D5V2T) and HRP linked anti-rabbit IgG secondary antibody were obtained from CST (Beverly, MA, USA). Anti-IL-1R2 (60262-1-Ig) and anti-GAPDH (60004-1-Ig) was purchased from Proteintech (Rosemont, IL 60018, USA). Secondary antibody (HRP conjugated anti-mouse IgG was purchased from Promega (Beijing, China).
Histological analysis
For histological analysis, patient biopsies and mouse tissue samples were fixed in paraformaldehyde at room temperature for 24 h and embedded in paraffin. Hematoxylin and eosin (H&E) and standard immunohistochemical staining were performed, according to the standard protocols. Six sections taken from the middle portion of tissue were examined. Epidermal thickness of skin was calculated by ImageJ. Cells positive for CD3, CD19, CD68 and IL-17A were quantified as the mean number of positive cells in five fields (original magnification, ×400).
Anti-CD3 (GB111337), anti-CD19 (GB11061-1), anti-TNF-𝛼 (GB13452), anti-IL-6 (GB11117), anti-IL-1𝛽 (GB11113), anti-CD68 (GB113109), anti-iNOS (GB11119), and anti-Arg-1 (GB11285), anti-IL-17Awere obtained from Servicebio (Wuhan, China).
Statistical analysis
All the results reported here are representative of at least three independent experiments, and the data are presented as the mean ± SEM (standard error of mean). All data were evaluated for normal distribution using the Kolmogorov–Smirnov test. When data were normally distributed, Student’s t-test was used for comparisons between two groups, and One-way analysis of variance (ANOVA) or Two-way ANOVA was used for multiple comparisons followed by Tukey’s post-hoc test; otherwise, Kruskal–Wallis test followed by the Dunn post-hoc test was used. P values < 0.05 were considered statistically significant. All statistical analyses were performed using GraphPad Prism 5.0 (Graph Pad Prism Software Inc, San Diego, CA, USA) and SPSS software (version 17.0, SPSS Inc., Chicago, IL, USA) for Windows.
GraphPad Prism 5.0 (Graph Pad Prism Software Inc, San Diego, CA, USA) was used to analyze data by ANOVA with Tukey’s post-hoc test, Student’s t-test, or Kruskal–Wallis test followed by Dunn post-hoc test when appropriate. Data were first analyzed for normality and equal variance to determine whether applied parametric tests were appropriate using Kolmogorov–Smirnov test with SPSS software (version 17.0, SPSS Inc., Chicago, IL, USA) for Windows. Data were plotted as means ± SD, and individual data points were included as dot points.