Patient Samples and Human IgG Purification
Blood samples were collected from four healthy controls (HCs) and four patients diagnosed with Neuromyelitis Optica Spectrum Disorder (NMOSD) at the Ophthalmology Department of the Chinese People’s Liberation Army (PLA) General Hospital. The selected patients were seropositive for AQP4-IgG, as confirmed by a cell-based indirect immunofluorescence assay, and were diagnosed according to the 2015 International Consensus Diagnostic Criteria. Exclusion criteria included the presence of other immune disorders, infections, or cancer. Patient demographics, including age, sex, and disease duration, are summarized in Table 1. None of the healthy controls had a history of disease or infection, and none had received any treatment in the preceding two months. Informed consent was obtained from all participants.
Table 1
Demographic and clinical characteristics of NMOSD patients and healthy controls.
| Age (years) | Sex (M/F) | AQP4-IgG titer | Disease duration (Months) |
CSF | Plasma |
Patient 1 | 52 | F | 1:3.2 | 1:1000 | 11 |
Patient 2 | 31 | F | 1:1 | 1:1000 | 20 |
Patient 3 | 50 | F | - | 1:1000 | 3 |
Patient 4 | 45 | F | 1:3.2 | 1:1000 | 9 |
Control 1 | 42 | M | - | - | - |
Control 2 | 42 | F | - | - | - |
Control 3 | 31 | F | - | - | - |
Control 4 | 40 | F | - | - | - |
Serum samples were heat-inactivated at 56°C for 30 minutes to deactivate complement proteins. Total IgG was purified from 4 mL of pooled serum from NMOSD patients and healthy controls using a protein-A column (GE Healthcare Bio-sciences, USA) and concentrated with Amicon Ultra-4 centrifugal filters (Merck Millipore, Germany). The purified IgG was diluted to 4 mL with phosphate-buffered saline (PBS) and sterilized using a 0.2 µm filter.
Animals and Intravitreal Injection
Female C57BL/6 mice aged 6–8 weeks and weighing 20–25 g were obtained from SPF Biotechnology (Beijing, China) and housed under controlled conditions with a 12-hour dark/light cycle, with free access to food and water. All experimental protocols adhered to the Statement for the Use of Animals in Ophthalmic and Vision Research and were approved by the Institutional Animal Care and Research Advisory Committee of the PLA General Hospital (Approval number: 2021-X17-08).
Mice were anesthetized via intraperitoneal injection of sodium pentobarbital. Intravitreal injections of 2 µL purified NMO-IgG were administered to the right eyes of the mice, while the left eyes served as controls and received equal volumes of purified HC-IgG. To prevent leakage, the needle was kept in place for an additional minute post-injection. Retinal circulation was confirmed by assessing the color of the retinal vessels, and antibiotic ointment was applied to prevent infection.
Pharmacological Ablation of Microglia
To deplete retinal microglia, mice were fed a diet containing the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX3397 (600 mg/kg, Moldiets, China) starting 7 days before the intravitreal injection and continuing until sacrifice. Control groups were fed an AIN76A chow diet, which had the same composition as the experimental diet but lacked PLX3397. The effectiveness of microglial ablation was evaluated by immunostaining retinal tissue sections.
Evans Blue Assay
A 2% (w/v) solution of Evans Blue dye (Sigma-Aldrich, St. Louis, Missouri, USA) was injected via the tail vein, followed by a 2-hour circulation period. After circulation, eyeballs were collected and immersed in 4% paraformaldehyde (PFA) for 2 hours. The retinas were carefully dissected and shaped into a four-leaf clover configuration. Blood-retinal barrier (BRB) integrity was examined using a microscope (Olympus, Japan) with an excitation wavelength of 594 nm.
Immunofluorescence Staining
After anesthesia, mice were perfused with saline and 4% PFA. Eyeballs were then fixed with 4% PFA for 1 hour at room temperature (RT). For flat mounts, retinas were dissected and rinsed with PBS containing 0.1% Tween (PBST). For frozen sections, eyeballs were dehydrated in sucrose at increasing concentrations (10%, 20%, and 30%) after fixation. Following the removal of corneas and lenses, the eyeball cups were embedded in O.C.T. and frozen at − 80°C. Cross-sections (12 µm) were obtained using a Thermo cryostat (CRYOSTAR NX50, Thermo, USA) and mounted onto microscope slides.
Tissues were blocked with PBS containing 5% bovine serum albumin and 0.1% Triton X-100 (blocking buffer) and incubated with primary antibodies overnight at 4°C. After washing with PBST, tissues were incubated with secondary antibodies for 1 hour at RT, followed by counterstaining with DAPI. Images were captured using the Operetta CLS High-Content Analysis System (PerkinElmer, Waltham, MA, USA) or a confocal imaging system (Olympus FV100, Olympus, Japan).
The antibodies used included mouse anti-AQP4 (ab9512, Abcam), rabbit anti-AQP4 (A5971, Sigma), mouse anti-GFAP (MAB360, Sigma), rabbit anti-IBA-1 (ab178846, Abcam), rat anti-IBA-1 (ab283346, Abcam), rabbit anti-GS (PA5-28940, Thermo Scientific), rat anti-C3 (ab11862, Abcam), rabbit anti-RBPMS (PA5-31231, Invitrogen), mouse anti-C1q (ab182452, Abcam), rat anti-C1q (ab11861, Abcam), mouse anti-iNOS (sc-7271, Santa Cruz), rat anti-CD31 (ab56299, Abcam), rabbit anti-MBP (ab40390, Abcam), and goat anti-human IgG Alexa Fluor 488 (A11013, Invitrogen).
Images were analyzed using Fiji software (National Institutes of Health, Bethesda, MD, USA). The average retinal ganglion cell (RGC) count was calculated using two fields from each retinal quadrant in flat mounts. AQP4 signals were quantified by measuring immunofluorescence intensity at vascular walls, and signal intensity along lines was determined using the ImageJ plot profile. Colocalization was quantified using Pearson’s correlation coefficient via the JACoP plugin in ImageJ.
Hematoxylin and Eosin (H&E) Staining
Mice were euthanized by cervical dislocation, and retinas were promptly isolated, fixed in 4% PFA, embedded in paraffin, and sectioned at a thickness of 4 µm. The paraffin-embedded eye sections were stained with hematoxylin and eosin (H&E) for histological examination. Images were captured using a light microscope (Olympus, Japan).
RT-qPCR Assessment
Total RNA was extracted from retinas and cultured cells using TRIzol reagent (Vazyme, Nanjing, China) following the manufacturer’s protocol. Complementary DNA (cDNA) was synthesized using the PrimeScript RT Reagent Kit (RR047A, Takara, Tokyo, Japan) for reverse transcription of mRNA. Quantitative real-time PCR (RT-qPCR) was performed with UltraSYBR Mixture (CW0957M, CW Biotech Co., Beijing, China) on the CFX96 RT-PCR system (CFX96 Optics Module; Bio-Rad, USA).
The sequences of mouse primers used were as follows:
-
GAPDH: Forward: 5′-AGGTCGGTGTGAACGGATTTG-3′; Reverse: 5′-TGTAGACCATGTAGTTGAGGTCA-3′
-
C3: Forward: 5′-CCAGCTCCCCATTAGCTCTG-3′; Reverse: 5′-GCACTTGCCTCTTTAGGAAGTC-3′
-
C1q: Forward: 5′-AAAGGCAATCCAGGCAATATCA-3′; Reverse: 5′-TGGTTCTGGTATGGACTCTCC-3′
-
iNOS: Forward: 5′-GTTCTCAGCCCAACAATACAAGA-3′; Reverse: 5′-GTGGACGGGTCGATGTCAC-3′
-
Arg-1: Forward: 5′-CCTGAAGGAACTGAAAGGAAAG-3′; Reverse: 5′-TTGGCAGATATGCAGGGAGT-3′
The sequences of human primers used were as follows:
-
GAPDH: Forward: 5′-GGAGCGAGATCCCTCCAAAAT-3′; Reverse: 5′-GGCTGTTGTCATACTTCTCATGG-3′
-
AQP4: Forward: 5′-TCAGCATCGCCAAGTCTGTC-3′; Reverse: 5′-CTGGGAGGTGTGACCAGATAG-3′
-
C3: Forward: 5′-GGCTGTCTTCTCTCAAGCA-3′; Reverse: 5′-GGGAATCTCACACATCACTCT-3′
GAPDH was used as a reference gene for normalization. Gene expression levels were calculated using the 2−△△CT method.
Optical Coherence Tomography (OCT)
Retinal thickness was assessed using a retinal imaging system (isOCT 4D-ISOCT, Optoprobe, Burnaby, Canada). Mice were anesthetized, and their eyes were dilated with 0.5% tropicamide eye drops. A single rectangular scan consisting of 800 A-scans by 800 B-scans over a 2.5 × 2.5 mm area centered on the optic nerve head (ONH) was performed on each eye. Retinal thickness was measured at 0.30 mm from the ONH. Images were acquired and retinal thickness measurements were calculated using software (version 2.0) from OptoProbe Research Ltd.
Electroretinogram (ERG) and Flash Visual Evoked Potential (f-VEP)
Mouse flash visual evoked potentials (f-VEP) and electroretinograms (ERG) were recorded using an OPTO-III visual electrophysiology instrument (Optoprobe Science, Burnaby, BC, Canada) to assess visual function. Mice were kept in darkness for over 12 hours before the procedure, which was conducted under dim red light.
For ERG recording, a loop electrode was placed on the corneal surface of the eye being tested, while needle electrodes were inserted under the skin of the cheek and tail. For detecting flash VEPs, a needle electrode was inserted under the skin between the ears, replacing the loop electrode. Three stable waveforms were recorded for each eye (normal and injured) in each animal.
Cell Culture
MIO-M1 cells, an immortalized retinal Müller glial cell line, were obtained from Biopike Technology Company Ltd (Minnesota, USA) and cultured in Dulbecco's Modified Eagle Medium (DMEM) containing 4.5 g/L glucose, supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin/streptomycin, and 1.0 mM glutamine.
BV2 microglia were obtained from Procell Life Science & Technology Co. Ltd (Wuhan, China) and cultured in DMEM supplemented with 10% FBS and 100 U/mL penicillin/streptomycin. Both cell lines were maintained in a humidified incubator with 5% CO2 at 37°C. The culture medium was refreshed every 2 to 3 days until the cells reached confluency.
Small-Interfering RNA (siRNA) Knockdown of AQP4
Three siRNA sequences targeting human AQP4 were designed and synthesized by Hanheng Biological Co. Ltd (Shanghai, China). Non-targeting siRNA was used as a control (NC). The siRNA sequence with the highest silencing efficiency was selected for subsequent experiments (siAQP4 sequence: GGUUGAGUUGAUAAUCACATT).
Transfection was carried out using the RNAiMAX transfection reagent (No.13778075, Invitrogen) according to the manufacturer's instructions. MIO-M1 cells were seeded into a 6-well plate, and RNAiMAX reagent, along with 5 pmol siRNA, was diluted separately in Opti-MEM and then added to each well. Treatments were conducted 24 hours post-transfection.
Transwell Co-culture of Retinal Müller Cells and Microglia
To investigate the effects of activated Müller cells on retinal microglia, MIO-M1 cells (retinal Müller glial cells) and BV2 cells (microglia) were co-cultured using a Transwell system. MIO-M1 cells were seeded onto the wells of a 24-well plate, while BV2 cells were seeded onto the Transwell permeable support membrane inserts with an 8 µm pore size (Corning, USA), which were placed above the MIO-M1 cells.
MIO-M1 cells were exposed to either control IgG or patients' purified human-IgG (diluted 1/50 in culture media). After treatment, these cells were co-cultured with BV2 microglia for 24 hours. Following the co-culture period, the cells were washed with PBS. MIO-M1 and BV2 cells were then harvested for further mRNA analysis and immunofluorescent staining.
To assess cell migration, the BV2 cells on the lower surface of the Transwell filters were stained with 5% crystal violet, allowing visualization and analysis of the BV2 cells that had migrated through the membrane pores towards the MIO-M1 cells.
Cell Viability Assay (CCK-8)
The primary retinal ganglion cells were acquired from Procella Co., Ltd (Catalog No. CP-M122) for the C1q neurotoxicity assay. The cell viability was assessed using the Cell Counting Kit-8 (CCK-8, Dojindo Molecular Technologies, Japan) according to the manufacturer's instructions. Briefly, cells were seeded in 96-well plates at a density of 106 cells per well and allowed to adhere overnight at 37°C in a humidified containing C3 treated BV2 culture medium (without or with 100µg/ml C1q neutralizing antibody ANX005) for 3 hours 10 µL of CCK-8 solution was added to each well, and the plates were incubated for 4 hours at 37°C. The absorbance at 450 nm was then measured using a SpectraMax® i3x Multi-Mode Microplate Reader (Molecular Devices). The absorbance values were directly proportional to the number of living cells, and cell viability was calculated as a percentage of the control group.
Statistical Analysis
Quantitative data were normalized to the control group and presented as a percentage of the control (%). The statistical significance of differences between groups was evaluated using an independent sample t-test when comparing two groups. For comparisons involving more than two groups, one-way ANOVA was employed. If the data were not normally distributed, the Mann-Whitney rank-sum test was used instead. Statistical analyses and the generation of graphs were performed using GraphPad Prism 8.0 software (GraphPad Software Inc., San Diego, CA, USA). Statistical significance was indicated as follows: *P < 0.05; **P < 0.01; ***P < 0.001.