Materials
CuO NPs was purchased from Nanjing XFNANO Materials Tech Co., Ltd. (XF130, Nanjing, China); Cell Counting Kit- 8(CCK-8) was purchased from Dojindo Laboratories, (Dojindo, Japan); Calcein-AM/PI Live Cell/Dead Cell Double Staining Kit was obtained from Solarbio (Beijing, China). Tublin, 4′,6-diamidino-2-phenylindole (DAPI) were obtained from Thermo Fisher Scientific (Shanghai, China). Cell-LightTM EdU Apollo567 Kit was purchased from Ribobio (Guangzhou, China); FITC-conjugated Isolectin B4 (IB4), Methyl cellulose and Triton-X-100 were purchased from Sigma-Aldrich (Shanghai, China). Matrigel Matrix was obtained from BD Biosciences (Shanghai, China); Antibodies to FDX1, DLAT, SDHB and β-Actin were obtained from Proteintech Group (Wuhan, China); The Pyruvate assay kit was acquired from Nanjing Jiancheng Bioengineering Institute in Nanjing, China. Additionally, all cell culture plates and dishes were obtained from Corning Life Sciences.
Cell culture
Human umbilical vein endothelial cell lines (HUVEC) were purchased from ScienCell (SanDiego, USA) and cultured in ndothelial Cell Medium (ECM, Cell Research, Shanghai, China) supplemented with 5% fetal bovine serum (FBS), 1% penicillin/streptomycin and 1% endothelial cells growth supplement (ECGS). The cells were maintained at 37°C in humidified 5% CO2, and the medium was changed every 2 days. HUVEC cells tested negative for mycoplasma.
Cell Proliferation
Cell proliferation rate was measured using EdU assays. Briefly, HUVECs were cultured in 96-well plates overnight and allowed to grow for 24 hours in the presence of CuO NPs. Subsequently, the culture medium was replaced with 100 uL of fresh medium containing EdU. Following a two-hour incubation period, the cells were fixed with 4% paraformaldehyde for 30 minutes, washed with glycine for 5 minutes and twice with 100uL 0.5% Trion X-100. They were then stained with 100uL of Apollo Dye Solution in darkness for 30 minutes, rinsed three times with 100 µL of 0.5% Trion X-100, and subsequently dyed with Hoechst for 30 minutes. The cells were subsequently rinsed thrice with PBS, followed by acquisition of images using a fluorescence microscope (IX81, Olympus). Statistical analysis was conducted using the ImageJ software.
Cell Migration
Cell migration was assessed utilizing a wound healing technique. Briefly, a total of 1×105 cells were seeded in a 2-well ibidi chamber plate to establish a confluent monolayer. Subsequently, the cells were subjected to a 24-hour pretreatment with CuO NPs. Following this, the insert was lifted, and the cells were washed thrice with PBS to eliminate any suspended cells, after which they were cultured with the initial medium. Microscopic images were captured at 0 hours and 16 hours using an Olympus IX81 microscope. All images were processed using ImageJ software.
Tube Formation
The Matrigel tube formation assay was conducted to evaluate the impact of CuO NPs on angiogenesis. Initially, 100 µL of Matrigel (12 mg/mL) was dispensed onto a pre-cooled 96-well plate and allowed to solidify at 37°C for a duration of 1.5 hours. Subsequently, HUVECs that had been exposed to either a control or CUO-NPs for a period of 24 hours were enzymatically detached using trypsin, and 1 × 104 cells were seeded into the aforementioned 96-well plate. Following an incubation period of 2 hours at 37°C, microscopic images were captured using an Olympus IX81 microscope, and the tube length was determined utilizing ImageJ software.
Sprouting Assay
Spheroid sprouting assay was conducted to evaluate the effects of CuO NPs on angiogenesis
in vitro. Initially, HUVECs were trypsinized and resuspended into control medium or medium containing CuO NPs at a density of 2 × 104 cells/mL. The cells were combined with 1 mL of methocel stock solution and subsequently transferred to a sterile multichannel pipette reservoir. A volume of 25 µL of the solution was then dispensed onto a 10 cm square petri dish using a pipette. Subsequently, the dish was inverted and placed in a cell culture incubator for a duration of 24 hours. The resulting spheroids were delicately rinsed with 10 mL of PBS and subsequently transferred into a 15 mL conical tube. The spheroids were subjected to centrifugation at a force of 200 g for a duration of 5 minutes, followed by resuspension in a 2 mL solution of methanol containing 20% FBS. A mixture of 4 mL collagen solution and 0.5 mL 10x Medium 199 was prepared, and the pH was adjusted by the addition of sterile ice-cold NaOH. Subsequently, 2 mL of the collagen/Medium 199 solution was combined with the methanol solution containing the spheroids. For each well of a 24-well plate, 1 mL of the spheroid-collagen solution was added and allowed to incubate at a temperature of 37°C for a duration of 30 minutes to facilitate collagen polymerization. Spheroids were subjected to simulation by adding 200 µL of DMEM containing CuO NPs for a duration of 24 hours within a humidified incubator set at 37°C and 5% CO2. Subsequently, images were captured utilizing an Olympus microscope model IX81, and the quantification of sprouting numbers or vascular length was performed using ImageJ software.
Live/dead stain
The cytotoxicity of CuO NPs on HUVECs was assessed in vitro using the Live/Dead fixed cell staining method. HUVECs were seeded in 24-well plates at a density of 1 × 104 cells/well and cultured for 12 hours. Subsequently, CuO NPs were suspended in DMEM and co-incubated with the HUVECs for 24 hours. Following this, the cells were stained with calcein-AM to identify living cells and propidium iodide to identify dead or late apoptotic cells for a duration of 20 minutes. The cellular fluorescence was observed and recorded using an Olympus IX81 microscope.
Oxygen Induced Retinopathy
Oxygen-induced retinopathy (OIR) mouse model was established as we previously reported [20].
From postnatal day 7 (P7) to postnatal day 12 (P12), pups with their nursing mother were subjected to an oxygen concentration of 75%. Subsequently, they were exposed to room air for an additional 5 days. On postnatal day 13 (P13), the mice were randomly divided into two groups. One group received a single dose of CuO NPs (0.5µg) injected into the vitreous cavity, while the other group received a PBS injection (3µL). On postnatal day 17 (P17), the pups were euthanized, and their eyes were removed for subsequent immunofluorescence assays. Throughout the experiment, the oxygen levels were continuously monitored using an oxygen analyzer (XBS-03S, HangZhou Aipu Instrument Equipment, Hangzhou, China).
Retinal Vascular Development
To assess the effect of CuO NPs on the postnatal development of retinal angiogenesis, control pups and pups received intraperitoneal injection of 0.5 µg CuO NPs at P5. The pups were euthanized, and the eyes were harvested at P7. Whole-mount retinas were stained with IB4 and ERG as described in the in vivo endothelial cell proliferation sections. The ratio of the vascular area to the retinal area was used to quantify superficial vascular layer development.
In Vivo Endothelial Cell Proliferation
To investigate the in vivo proliferation of endothelial cells, we conducted a proliferation analysis using the Cell-LightTM EdU Apollo567 Kit. The staining procedure for EdU was carried out in accordance with the instructions provided by the manufacturer. P17 mice were administered with EdU (5 mg/kg)and euthanized after a 6-hour interval. The eyes were then removed and fixed in a 4% PFA solution for a duration of 30 minutes at RT. Subsequently, the eyes were isolated under a microscope and subjected to blocking using a 1% BSA and 5% FBS buffer containing 0.5% Triton- X-100 for 30 minutes at RT. Following three washes in PBS, the retinas were subjected to overnight incubation at 4°C with FITC-conjugated Isolectin B4 and ERG primary antibodies(diluted at a ratio of 1:250 in a solution containing 1% BSA, 5% FBS, and 0.05% Triton-X-100 in PBS). Subsequently, the retinas underwent three additional washes with PBS. Next, the retinas were incubated with secondary antibodies for a duration of 2 hours at RT. Following another three washes with PBS, the retinas were incubated with EdU in accordance with the manufacturer's instructions. Finally, the retinas were mounted onto glass slides and sealed using a fluorescent mounting medium. A confocal microscope system (Leica TCS SP5-II) was used to capture the images, and Image J was used to quantify the avascular and neovascular areas. The Leica TCS SP5-II confocal microscope system was employed to acquire the images, while Image J software was utilized to quantify the avascular and neovascular areas.
RNA Sequencing
Cells in culture dishes were treated with CuO NPs for 48 h and total RNA prepared for RNA-seq analyses were extracted from cells using Trizol reagent as described by the manufacturer (Invitrogen). RNA sequencing and analysis were conducted at TIANGEN Biotech (Beijing) Co., Ltd. The pathway enrichment analysis was carried out using the KEGG database and GO database. A robust algorithm was employed to identify genes that were differentially expressed between two samples, following the principles described in "The significance of digital gene expression profiles". Multiple tests and analyses were performed, using a P value as the threshold for determining significance. Pathways with a P value < 0.05 were considered to have a significant enrichment of differentially expressed or modified genes.
Targeted metabolomic analysis
Cell supernatant was prepared for metabolomic analysis. Briefly, HUVECs treated with CuO NPs for 24 h were suspended in 400 µL of an extraction solution (methanol: acetonitrile: water (v/v/v, 4:4:1)). Afterwards, cells were submitted to Grinding treatment for 4 min, and were sonicated in an ultrasonic water bath for 20 min and incubated at -40°C for 1 h. Next, samples were centrifuged at 12000 g for 15 min at 4°C and the supernatant was spin dry under vacuum. The film was redissolved by the addition of pure water, followed by passing and collecting the supernatant. Subsequently, Shanghai iProteome Biotechnology Co., Ltd. conducted further analysis. HPIC-MS/MS analysis was carried out using a Thermo Scientific Dionex ICS-6000 HPIC high performance ion chromatograph with AS11-HC RFIC separation column. Mass spectrometry in multiple response monitoring (MRM) mode was performed using a 6500 QTrap + mass spectrometer. AB SCIEX Analyst Work Station Software, MultiQuant software, and Chromeleon7 were utilized for the acquisition of mass spectrum data and quantitative analysis of the target compounds.
Western Blotting
The extraction of total protein from cells or exosomes was performed using RIPA lysis buffer supplemented with 2% protease and protease inhibitor. The protein concentrations were determined using a standard trace bisphenolic acid assay (BCA, Beyotime, Shanghai, China) following the manufacturer's instructions. The samples were prepared in 5 × SDS-PAGE loading buffer and subjected to heating at 95°C for 10 minutes. Subsequently, the protein samples and protein markers were separated by 12.5% SDS-PAGE at 120 volts for 60 minutes and transferred to PVDF membranes at 100 V for 120 minutes. The membranes were blocked using a 5% nonfat dry milk solution in TBST (Tris-buffered saline with 0.1% Tween-20) for a duration of 2 hours at room temperature (RT). Subsequently, the membrane was briefly rinsed in TBST and incubated overnight at 4°C with primary antibodies diluted in a 5% BSA solution in TBST. The primary antibodies used in this study were summarized in Table S1. Following three washes in TBST, the membrane was incubated with a horseradish peroxidase-conjugated secondary IgG antibody (in a 5% non-fat dry milk solution) for 1.5 hours at RT. Following an additional three washes in TBST, chemiluminescence was observed using the ECL Western blotting detection reagent (Merck-Millipore, Darmstadt, Germany). The Gelview 6000Plus Image Capture System (Guangzhou Biolight Biotechnology Co., Ltd, Guangzhou, China) was employed to capture images, which were subsequently quantified using the ImageJ software.
Pyruvate Detection
The extracellular Pyruvate Levels were measured using the Pyruvate Assay Kit (Nanjing Jiancheng Bioengineering Institute, A081-1-1) according to the manufacturer’s instructions. In summary, a volume of 100 µL of cell lysate supernatant from HUVECs was added to a previously prepared 0.5 mL working solution and thoroughly mixed. The reaction was halted by the addition of 2.5 mL of a terminator solution after incubation at 37 ℃ for 10 minutes. Subsequently, 100 µL of the reaction mixtures were dispensed into a 96-well plate and the absorption peak at a wavelength of 505nm was quantified.
Blood routine examination and blood biochemistry analysis
In order to assess the blood compatibility of CuO NPs, an analysis of blood routine and biochemical indexes was conducted. C57BL/6 mice were utilized for the blood routine and biochemical evaluations. The mice were randomly allocated into twpo groups and received intravenous infusions of phosphate-buffered saline (PBS) and CuO NPs. Following a 14-day period, blood samples (100 µL) were obtained from the heart. The blood biochemical analysis was carried out using an LW C400 Mindray automatic biochemical analyzer (Shenzhen Lanyun Medical Equipment Co., Ltd., Shenzhen, China), while the routine examination analysis was performed using an automatic hematology analyzer (TEK8500 VET, Tecom Science, Jiangxi, China).
Statistical analysis
Statistical analysis was performed using ImageJ software (ImageJ software, 1.52a National Institutes of Health, Bethesda, MD, USA) and GraphPad Prism (GraphPad software 9.0, GraphPad, Bethesda, MD, USA) and. The measurement data of the two groups were subjected to analysis using an unpaired Student's t-test, while the comparison among multiple groups was conducted through one-way analysis of variance (ANOVA). Each experiment was replicated three times, and statistical significance was determined at a threshold of P < 0.05 (* P < 0.05; ** P < 0.01; *** P < 0.001; **** P < 0.0001).