Chemical materials. Dopamine hydrochloride, sodium hydroxide (NaOH), sodium nitrite (NaNO2), doxorubicin hydrochloride (DOX), 1,1-Diphenyl-2-picrylhydrazyl radical (DPPH), DETA NONOate (NOC18), 5,5-dimethyl-1-pyrrolineN-oxide (DMPO), 4,5-Dihydro-4,4,5,5-tetramethyl-2-phenyl-1H-imidazol-1-yloxy-1-oxide (PTIO), X-Gal (5-bromo-4-chloro-3-indolyl-β-D-galactoside) and dihydroethidium (DHE) were purchased from Meryer (Shanghai) Chemical Technology. Phosphate-buffered saline (PBS, pH 7.4, Na2HPO4-NaH2PO4, 10 mM) solution was prepared in the laboratory. All chemicals and reagents were of analytical grade and used as received without further purification. Ultrapure water (18.2 MΩ cm− 1 at 25°C) purified by a Milli-Q system was used throughout the experiment.
TEM imaging and spectroscopic analysis. TEM imaging was conducted on a JEM-1400Flash HC (JEOL, Japan) at 120 kV. The sample was prepared by dispersing a small amount of freeze-dried powder in PBS. Then, the suspension was dropped on 230 mesh copper TEM grids covered with thin amorphous carbon films. FT-IR spectra were measured by a VERTEX70 spectrometer (Bruker, Germany) in the range of 4000–400 cm− 1. UV-vis spectra were obtained using a VNANODROP 8000 spectrometer (Thermo, Germany). A DLS particle size analyzer (Malvern 2000, U.S.A) was used to determine the hydrophilic diameters of the particles. XPS was performed with an ESCALAB 250Xi (thermofisher scientific, China) X-ray source. The crystal structure and oxidation state of UPDA NPs were analyzed using a X-ray diffractometer (XRD, Smartlab, China). The ESR spectroscopy signal was obtained on a Bruker A300 (X-band) spectrometer (Bruker, Germany). All the measurements were performed at room temperature if not specified otherwise.
Preparation of UPDA NPs. 180 mg of dopamine hydrochloride was dissolved in 90 mL of deionized water. 840 µL of 1 mol L− 1 (M) NaOH solution was added to the dopamine hydrochloride solution at 60°C and the mixture underwent vigorous stir. The reaction lasted for 5 h. The solution color turned to pale yellow as soon as NaOH was added and gradually changed to dark brown. The product was collected by centrifugation at 16000 rpm for 20 min and was then washed with deionized water three times. The aqueous solvent was removed by freeze-drying to obtain black solids of PDA. Under vigorous stir, 20 mg polydopamine nanoparticles was dissolved in 10 mL of 0.1 M NaOH. Then we swiftly dropped 0.1 M HCl into the obtained solution to adjust the pH to 7.0 under sonication with an output power of 600 W for 2 min. We obtained a bright black polydopamine solution. The particles were retrieved by centrifugation with a centrifugal-filter (centrifugal filter device, MWCO = 30kDa) at 8000 rpm for 8 min and was washed several times with deionized water to remove the byproduct NaCl, followed by freeze-drying to obtain black solids of UPDA NPs.
Determination of RONS-scavenging capability. The evaluation of ABTS radical scavenging activity was based on a previously reported method16. Briefly, the ABTS radicals were generated by incubating 7 mM ABTS stock solution with 2.45 mM potassium persulfate in dark for 16 h. Then, the ABTS radical solution was diluted with PBS to reach an absorbance at 405 nm. 2 mL UPDA NPs solutions (0, 20, 40, 60 µg mL− 1, respectively) were mixed with 2 mL ABTS solution and were placed in dark for 10 min. Then the absorbance at 405 nm was monitored with a UV-vis spectrophotometer. The ABTS radical scavenging abilities were calculated as follows:
ABTS scavenging ratio (%) = ((Acontrol-Asample)/Acontrol) ×100;
where Acontrol is the absorbance of a standard solution without any radical scavengers, and Asample is the absorbance after the reaction with the radical scavengers, respectively.
The SOD-like activity was determined using an SOD assay kit (WST-1 method) (Nanjing Jiancheng Bioengineering Institute, Nanjing, China). Briefly, O2− was generated through the oxidation of xanthine by xanthine oxidase (XO), which can convert WST-1 into formazan with a characteristic absorption at 450 nm. The formazan concentration was measured using a multiple plate reader.
The CAT-like activity was determined based on the H2O2 decomposition reaction. A tube containing 10 mM H2O2 and 0.1 mM CTPO in PBS was pumped with N2 for 10 min. Then, 50 µg mL− 1 UPDA NPs was added and incubated for 5 min. ESR spectra were monitored at different time points. H2O2 of different concentrations (5–60 µM) was added to 100 µg mL− 1 UPDA NPs. The final volume was adjusted to 3 mL, the absorbance value was measured at 640 nm and the dynamics curve was registered on a UV spectrophotometer. The steady-state kinetic assay was performed to confirm the CAT-like catalytic mechanism of UPDA NPs. The Michaelis-Menten constant (Km) of UPDA NPs-catalyzed reaction was obtained by plotting the initial velocity versus H2O2 concentration. The maximum velocity Vmax was calculated from the Lineweaver-Burk plot, where 1/V = 1/Vmax + Km (VmaxC).
The amount of •OH was measured with the Fenton reaction, in which •OH causes MB to oxidize and its color fading degree is proportional to the amount of •OH. Different concentrations of UPDA NPs (0, 45, 90 µg mL− 1) were added to the •OH-MB reaction solution, and the absorbance at 664 nm was measured. The •OH-scavenging rate was obtained with the following equation:
Inhibition (%) = (Acontrol-Asample)/Asample,
where Acontrol is the absorbance of the control group, and Asample is the absorbance of the UPDA NPs-treated group.
DPPH was used to evaluate the RNS scavenging activity of UPDA NPs. Different concentrations of UPDA (0, 5, 25, 100 µg mL− 1) were mixed with 40 µM DPPH for 12 h and the absorbance spectra at 532 nm were recorded.
The ·NO-scavenging ability of UPDA NPs was tested by ESR using carboxy-PTIO as the trapper and the detection dye for ·NO. NOC18 was used as the source of ·NO. Carboxy-PTIO was dissolved in phosphate buffer (250 mM, pH 7.4), and NOC18 was dissolved in NaOH (1 mM). In a test tube, 0.5% methylcellulose was mixed with NOC18 (5 µM) for 30 min at room temperature, and the mixture was then added into the carboxy-PTIO solution (5 µM) in the absence or presence of UPDA NPs. The ESR spectra were then recorded. The ESR signal of carboxy-PTIO is characterized by five peaks. ·NO generated from NOC18 can reduce carboxy-PTIO into carboxy-PTI, which shows seven peaks of ESR signals. The reduction of carboxy-PTIO by ·NO will lead to a color change from purple to yellow. In addition, the ONOO- scavenging ability of UPDA NPs was evaluated by UV-vis spectroscopy using pyrogallol red as the indicator.
Animals. The animal experiments were performed in accordance with the guidelines of the Administrative Committee of Laboratory Animals. All the procedures involving animals were approved by Shanghai University Administrative Committee of Laboratory Animals (ECSHU 2021-039). A maximum of four mice per cage were kept on a 12-h light/dark cycle at a constant temperature (22°C) with food and water ad libitum. C57BL/6J mice of 26 weeks were used for DOX-induced senescent model. Mice were given two intraperitoneal injections of DOX at a dose of 10 mg/kg. All mice were kept in group housing until the start of the experiment and were then randomly assigned to control and experimental groups.
Biocompatibility and biosafety evaluation. C57BL/6J mice (2 months old, 20–25 g) were intravenously administrated with UPDA NPs at a single dose of 200 µL (100 µg mL− 1). The mice injected with PBS were used as the control group. At day 30 post injection, the mice were anesthetized with intraperitoneal injection of chloral hydrate and sacrificed to harvest major organs (including heart, liver, spleen, lung, and kidney) for hematoxylin and eosin (H&E) staining and histological analysis. Mouse blood was collected through heart puncture. Routine blood parameters, including WBC, LYM, MON, GRA, MPV, RBC, HGB, MCH, HCT, RDWSD, MCV, MCHC, PLT, PDW and RDWCV, were analyzed on a Sysmex XS-800i automated hematology analyzer (Sysmex, Japan).
Transcriptomics analysis. DOX-induced senescent mice were randomly divided into two groups: the control group (n = 5) and the group treated with 100 µg mL− 1 UPDA NPs at a dosage of 200 µL (experimental group, n = 5). After 24 h post injection, mice were sacrificed to collect the kidneys. Total RNA was extracted using TRIzol® Reagent (Invitrogen) according to the manufacturer’s instructions and genomic DNA was removed using DNase I (TaKara). Then RNA quality was determined by a 2100 Bioanalyser (Agilent) and quantified using an ND-2000 instrument (NanoDrop Technologies). Only high-quality RNA samples (OD260/280 = 1.8 ~ 2.2, OD260/230 ≥ 2.0, RIN ≥ 6.5, 28S:18S ≥ 1.0, > 1µg) were used to construct sequencing library. RNA-seq transcriptome library was prepared with a TruSeqTM RNA sample preparation Kit (Illumina, San Diego, U.S.A) using 1µg of total RNA. Briefly, messenger RNA with polyA tails was captured by oligo (dT) beads and was then fragmented in fragmentation buffer. Next, double-stranded cDNA was synthesized using a SuperScript double-stranded cDNA synthesis kit (Invitrogen, U.S.A) with random hexamer primers (Illumina). The synthesized cDNA was subjected to end-repair, phosphorylation and adenylation on the 3’ end. Libraries were enriched for cDNA fragments of 300 bp on 2% Low Range Ultra Agarose followed by PCR amplification using Phusion DNA polymerase (NEB) for 15 PCR cycles. After the quantification with a TBS380 fluorometer, paired-end RNA-seq sequencing library was sequenced with a HiSeq xten/NovaSeq 6000 sequencer (2 × 150bp read length, Illumina). The raw paired end reads were trimmed and subjected to quality control by SeqPrep (https://github.com/jstjohn/SeqPrep) and Sickle (https://github.com/najoshi/sickle) with default parameters. Then clean reads were separately aligned to reference genome with orientation mode using the HISAT2 software (http://ccb.jhu.edu/software/hisat2/index.shtml). The mapped reads of each sample were assembled by StringTie (https://ccb.jhu.edu/software/stringtie/index.shtml? t = example) in a reference-based approach. To identify DEGs between two different samples, the expression level of each transcript was calculated according to the transcripts per million reads (TPM) method. RSEM (http://deweylab.biostat.wisc.edu/rsem/) was used to quantify gene abundances. Differential expression analysis was performed using the DESeq2 package with |log2FC|>1 and Q value < = 0.05 as the criteria of DEGs. GO functional enrichment and KEGG pathway analyses were carried out using Goatools (https://github.com/tanghaibao/Goatools) and KOBAS (http://kobas.cbi.pku.edu.cn/home.do) with Bonferroni-corrected p-value ≤ 0.05 considered to be the threshold of enrichment. The PPI network of selected DEGs was constructed using the STRING database [PMID: 30476243] and was mapped with Cytoscape software (v 3.8.2).
Cell culture. The human embryonic renal epithelial 293T cell line was acquired from ATCC, and cultured in Dulbecco’s modified Eagle’s medium (Gibco, U.S.A) containing 10% fetal bovine serum (Gibco, U.S.A) and 1% s7 penicillin/streptomycin (Gibco, U.S.A) at 37°C with 5% CO2. For DOX-induced senescence, 293T cells were treated twice with 0.2 mM DOX with a 2-day interval and were analyzed 7 days later. The expression of SA-β-GAL was detected by the cleavage of X-Gal. The levels of IL-6 and IL-1β were determined using commercially available kits and quantified from 3 replicates (n = 3).
Measurement of ROS scavenging activities in vitro. 293 T cells were seeded into 96-well and 24-well plates at a density of 1 × 104 cells per well, and 1 × 105 cells per well, respectively. After incubation for 24 h, cells were treated with 2×10− 7 M DOX, 8g/L D-gal, or 250 µM H2O2 and 100 ug mL− 1 UPDA NPs, and were further incubated at 37°C for a 2-day interval and analyzed 7 days later. For determining cell viability, cells seeded in 96-well plates were evaluated with cell counting kit-8 (CCK-8, Beyotime, China). Cells without the addition of UPDA NPs were regarded as the control. At least 50,000 cells were analyzed in each sample. For determining the ROS-scavenging effect of UPDA NPs (20 ug mL− 1), DCF, an oxidation sensitive fluorescent dye, was used to detect the intracellular ROS level 60. Briefly, DCFH-DA is a non-fluorescent chemical compound which can diffuse through the cell membrane freely and can be hydrolyzed by an intracellular esterase to generate DCFH. The non-fluorescent DCFH can be oxidized by the intracellular ROS to form fluorescent DCF. Therefore, the quantity of intracellular ROS is correlated with the fluorescent intensity of DCF. After the aforementioned incubation, cells were gently rinsed three times with serum-free medium to remove the free UPDA NPs. Then, a final concentration of 10 µM of DCFH-DA in serum-free medium was added to the cells and incubated in dark at 37°C for 30 min. Afterwards, the cells were washed with serum-free medium three times to remove unloaded DCFH-DA probe, imaged using a laser confocal microscope (Zeiss, Germany), and subjected to flow cytometry analysis.
Immunocytochemistry. Normal and DOX-induced senescent 293T cells were fixed with 4% paraformaldehyde (PFA) for 15 min, permeabilized with 0.2% Triton X-100 for 3 minutes and was then blocked with 10% (v/v) BSA in PBS for 2 h at room temperature or at 4℃overnight. Subsequently, the cells were washed with PBS three times and were then incubated with primary antibodies mouse anti-FOXO4 (1:500, Abcam, UK), rabbit anti-p16ink4a (1:500, Abcam, UK), or rabbit anti-lamin B1 rabbit (1:500, Abcam, UK) for 2 h. The cells were then washed three times with PBS and incubated with secondary antibodies Goat anti-Mouse IgG (H + L) Alexa Fluor TM Plus 555 or goat anti-rabbit IgG (H + L) Alexa Fluor®488 at a dilution of 1:200 for 2 h. After washing three times with PBS, the cells were imaged on an inverted fluorescence microscope (Zeiss, Germany). The fluorescence intensity was obtained from 3 randomly selected regions of the cell culture to determine the expression levels of FOXO4, p16ink4a and lamin B1 in each group.
Western blotting. 293T cells (1.0 × 106) were collected and total protein was extracted with RIPA lysis buffer IV (Beyotime, China). The protein lysates were then separated by SDS-PAGE and transferred to polyvinylidene difluoride (PVDF) membranes. The membranes were then incubated with primary antibodies (diluted at 1:1000) against p21, lamin B1 or DcR2, and then with a horseradish-peroxidase conjugated secondary antibody (1:500). β-Actin (1:1000) was used as a loading control. Membranes were subsequently exposed according to manufacturer’s instructions (Western blot kit, Sangon Biotech Co., Ltd.). The band densities were quantified in ImageJ and normalized against GAPDH, representing the protein expression levels of p21, lamin B1 and DcR2.
Immunohistochemistry. The mice were anesthetized and perfused with PBS. The kidneys were collected, fixed with 4% PFA, and cut to obtain 30 µm sections using a vibratome (Invitrogen, U.S.A). After three rinses in PBS (5 min each), the sections were blocked in PBST (PBS + 0.3% Triton X-100) with 3% bovine serum albumin for 2 h, and were then incubated in primary antibodies (1:1000) at 4°C for 24 h, followed by three rinses in PBS (5 min each). Then, the sections were incubated with secondary antibodies (1:500) for 2 h at room temperature. After the incubation, the sections were washed with PBS for 5 min, and the nuclei were counterstained with DAPI for 10 min. Finally, the sections were washed in PBST and mounted in mounting medium. Fluorescence images were acquired with a 20× objective on a confocal microscope.
Flies were anesthetized with CO2, dissected in PBS and fixed with 4% PFA at room temperature. Samples were incubated with anti-cleaved caspase-3 polyclonal antibody (1:500, Beyotime, China) and Alexa fluor 594 conjugated secondary antibody (1:500, Invitrogen), followed by DAPI staining and fluorescence imaging.
Determination of renal and hepatic function parameters. Blood samples were acquired from normal mice and DOX-induced senescent mice with or without the treatment of UPDA NPs for plasma separation. These samples were then centrifuged for 10 min at 1200 g. The supernatants were transferred into 1.5 mL tubes and recentrifuged for 5min at 1200 g. The supernatants were transferred again into 1.5 mL tubes for determining the levels CRE, BUN, ALT and AST.
Assessment of swimming performance. We monitored the swimming behavior of different mouse groups by continuously measuring their swimming speed. Swimming activity was recorded for 30 days.
Drosophila culture. Drosophila melanogaster (w1118) stocks were maintained and crossed according to standard laboratory procedures. Flies were raised under a 12/12 h, light/dark cycle at 25 ℃ with 60% humidity.
Measurement of ROS level in Drosophila. Different groups of Drosophila were sacrificed and washed with PBS. 50 mg of tissue was suspended in homogenate buffer A and was thoroughly homogenized with a glass homogenizer. The tissue homogenates were centrifuged at 100 g and 4°C for 3 min. The supernatant was collected for further experiments. For ROS level determination, 200 µL supernatant was added to a 96-well plate, and mixed with 2 µL DHE detection solution with a pipettor. The mixture was incubated at 37°C in dark for 30 min. The fluorescence intensity was measured in a fluorescence plate reader with an excitation wavelength of 488–535 nm and an emission wavelength of 610 nm. For determining the protein concentration, 50 µL supernatant was diluted 30 times with PBS and 100 µL diluent was subjected to Bradford protein assay. The ROS level is expressed as fluorescence intensity per mg protein.
Determination of lifespan and climbing ability of Drosophila. Newly eclosed wild-type female Drosophila were used for lifespan assay. 100 flies were used for each group. Flies were transferred for fresh food and the death number was recorded every day. Data were presented as survival curves and statistical analysis was performed with log-rank test. For climbing assay, eight male and eight female flies were placed in a plastic vial. The flies were gently knocked to the bottom of the vial before timing. Climbing distances within 4 s and 20 s were measured.
Statistical analysis. All quantitative results are shown as mean ± standard deviation. Statistical significance was determined using the likelihood ratio test for comparing the transcriptional levels of DEGs, the log-rank test for the lifespan assay of Drosophila and one-way ANOVA with Tukey’s post hoc test for all other experiments. The significance levels were set at *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001.
Data availability
The data that support the findings of this study are available within the paper and its supplementary information file or from the corresponding authors upon reasonable request.