All human studies and animal procedures conformed to the National Institutes of Health (NIH) guidelines, and were approved by the Ethics Committee of The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, China, and conducted in accordance with the Declaration of Helsinki.
Cell culture
The HEK293T cells were purchased from Zhongqiaoxinzhou Biotechnology (Shanghai, China) and cultivated in high-glucose DMEM enriched with 10% FBS and 100U/mL penicillin-streptomycin (P/S). Human cardiac microvascular endothelial cells (HCMECs) at passage 3, sourced from Lonza Bioscience (Basel, Switzerland), were cultivated in endothelial culture media (ECM; ScienCell, Carlsbad, CA, USA). Primary mouse CMECs (MCMECs) were obtained from the left ventricles of C57BL/6 mice using a previously established method [24, 25]. After the epicardial, endocardial, and coronary arteries were removed, the left ventricular tissue was chopped and digested with liberase. The cell suspension was incubated for 30 min at 4°C with microbeads conjugated to anti-CD31 antibody (ab7388, Abcam, UK; Thermo Fisher Scientific, USA) under gentle rotation. The first step was the isolation of primary MCMECs using a magnetic separator. These cells were subsequently cultivated on fibronectin-coated plates using a complete ECM (ScienCell, USA). Following the removal of serum, the cells were cultivated in hypoxic conditions consisting of 5% CO2, 1% O2, and the remaining balance of N2 for a period of 1–3 days, and the cells in the control group were kept in the normoxia mixture (5% CO2 and 21% O2), both the hypoxic cells and normoxic cells were derived from the same maternal generation.
Recombinant adenovirus and adeno-linked virus generation
In vitro experiments, recombinant adenovirus was used to infect endothelial cells to interfere with cirAXTN1, SLUG, and ALKBH5. Additionally, recombinant adeno-associated virus (AAV) type 9 systems, including vectors with control/circATXN1 driven by an endothelial-specific promoter (Tie promoter) or scramble shRNA/circATXN1 shRNA, were administrated into the mice tail veins to modify circATXN1 levels in the heart. The AAV2 vector genomes were administered to the mice via tail vein injection, with 100 µl of the virus containing 2 × 1011 VG.
Mice lower limb ischemia model
After anesthesia with inhaled 2% isoflurane, C57/BL6J mice (8–10 weeks, male) were subjected to a femoral artery surgery in the right hind limb. There was one ligation site at the opening of the profundal femoris artery and another near the opening of the saphenous artery. Then, the femoral artery between the two points was cut off, and finally, sew up the wound. For gain and loss of function experiments, the mice were injected with the AAV virus in the gastrocnemius muscle 1 week before HLI surgery. The blood flow was calculated by laser Doppler scanning system (Perimed, Sweden) on postoperative days 1, 3, 7, and 14. The perfusion ratio was quantified from the ischemic limb (right) and the control limb (left), which reflected the recovery of blood flow.
Mice myocardial infarction model
After anesthesia with inhaled 2% isoflurane, C57/BL6J mice (6–8 weeks, male) received a thoracotomy with the assistance of a small animal respirator. Under the microscope, the left anterior descending coronary artery was ligation with 10–0 nylon wire, and the color of the surrounding myocardial tissue turned immediately pale. Mice in the sham group received all the surgical procedures induced by MI except the ligation step. For gain and loss of function experiments, the mice were administered with AAV virus via the tail vein 1 week before MI surgery. Left ventricular ejection fraction was measured on pre- and postoperative days 7,14, and 28 by a small animal ultrasound instrument.
RT-qPCR
Aligning with manufacturer recommendations, TRIzol reagent (Takara, Japan) was implemented to obtain total RNA. The PrimeScriptTM RT reagent Kit (Takara, Japan) was implemented to convert 1 µg of total RNA into cDNA with reverse transcription, and random hexamer primers were used specifically for the reverse transcription of circRNAs. If RNase R pretreatment is required, 5 µg of total RNAs and 10 units of RNase R were incubated at 37°C for 15 min. The qPCR experiment was conducted with the SYBR Green master mix (Vazyme, China) and examined using the QuantStudio 6 Operating Software (Life Technologies, USA). Primer sequences are shown in the Supplementary table 3.
RNA sequencing for circRNA
Total RNA was obtained with the TRIZOL reagent (Takara, Japan) to find differentially expressed circRNAs in HCMECs under normoxic and hypoxic conditions. rRNA Depletion and VAHTS Universal V6 RNA-seq Library Prep Kits (Vazyme, China) were utilized to produce circRNA-sequencing libraries based on the manufacturer's recommendations. The CIRC2 algorithm was employed to identify circRNAs, which were then annotated by comparing sequences in circBase. circRNAs with a log fold change greater than 1 and a p-value less than 0.05 between the two conditions were considered to be significantly differentially expressed.
The mRNA half-life determination
The cells were treated with 5 µM actinomycin D (Sigma, USA) and afterward digested at specified time intervals for the purpose of RNA extraction. At each time point, the initial level (0 h) was implemented to adjust the remaining RNA levels. The decay kinetics of mRNA were analyzed by GraphPad Prism software, and the result was rendered with a one-phase exponential decay curve.
Cytosolic/Nuclear fractionation
Typically, 1×107 cells were gathered with trypsin-EDTA buffer. After washing and centrifugation 2–3 times, the cell pellets were operated according to the instructions of the NE-PER Nuclear and Cytoplasmic Extraction Reagents step by step. The obtained nuclear/cytosolic extracts were used for subsequent Western blot (WB) analysis. As references for the cytoplasmic and nuclear fractions, GAPDH and LMNB were chosen, respectively.
RNA fluorescence in situ hybridization (RNA-FISH)
A biotin-labeled oligonucleotide probe was developed in TSINGKE, either antisense (AS) or sense (S), that specifically targets the circ-ATXN1 junction. The biotin RNA Labeling Mix and T7 RNA polymerase (Roche, Germany) were employed to generate the probes for 18S and U6 through in vitro transcription of PCR fragments. The hybridization occurred inside a humidified chamber at 37°C for 16 h, either with or without treatment with RNase R at a concentration of 3 U/mg. The Fluorescent In Situ Hybridization kit (RiboBio, Guangzhou, China) was implemented for signal detection, whereas 4′,6-diamidino-2-phenylindole (DAPI, Sigma) was implemented for nuclei counterstaining. Primer sequences are shown in the Supplementary table 3.
Immunofluorescence assays
HCMECs were planted on the cell slide to a suitable density and treated with 4% paraformaldehyde for 15 min to fix them. The cell specimens were afterward rinsed three rounds with pre-chilled PBS and subjected to 0.1% Triton X-100 at ambient temperature for 15 min to allow for permeabilization. Following a 1–2 h blocking with a 1% BSA solution at ambient temperature, the slides were treated with primary antibodies and kept in a humidified container overnight at 4°C. After rinsing the cell slides with PBS three rounds for 5 min each. They were treated with fluorescent dye-labeled secondary antibodies for 1 h at ambient temperature in the darkness. Afterward, the slides were incubated with DAPI for 15 min at ambient temperature without exposure to light. For immunofluorescence staining (IFS) of tissues, the tissues need to be fixed, paraffin-embedded, and sectioned. Sections were successively deparaffinized, rehydrated, and treated in a pressure cooker with sodium citrate antigen repair solution. The subsequent steps, including blocking, primary and secondary antibody incubation, and DAPI staining, refer to the procedures described above. A fluorescence microscope (Zeiss, Germany) was used to visualize and acquire images, and ImageJ software was deployed to analyze images.
Immunohistochemistry
Immunohistochemical staining was conducted as earlier established [26]. Tissue samples were embedded in paraffin and sectioned, Vimentin immunohistochemical staining was performed. Photographs were taken using an optical microscope (Olympus, Japan).
Western Blot analysis
The cells were gathered and disrupted with RIPA buffer containing a protease inhibitor. Typically, a quantity of 20 µg of protein was isolated with a 10% SDS-PAGE method and then electro-transferred onto PVDF membranes (Millipore, USA). The membranes were obstructed with a 5% solution of skim milk and thereafter incubated overnight at 4°C with primary antibodies. Following the washing step with TBS-T solution, HRP-conjugated secondary antibodies were introduced and incubated at ambient temperature for 2 h. The blots were processed with Super ECL plus reagents (Vazyme, China) and were displayed using automated chemiluminescence image analysis equipment (Tanon 5200, China). Antibodies used are shown in the Supplementary table 2.
Migration assays
After 24h infection, HCMECs were enzymatically broken down and resuspended in serum-free ECM. A suspension containing 1×104 cells was placed into the upper chambers, while a full medium with 10%FBS was introduced to a 24-well plate lower chambers. The entire Transwell® device (Corning, USA) was incubated in the cell incubator for 24h, and the upper chamber was successively transferred to 4% paraformaldehyde for fixation and 0.1% crystal violet for staining. Next, a cotton swab was utilized to delicately cleanse the non-migratory cells located inside the top cavity, while the migrated cells were left intact on the underside of the polycarbonate membrane. The migrating cells were captured with microscopy and quantified.
FUCCI reporter cell line and Proliferation assays
The plasmid pBOB-EF1‐FastFUCCI‐Puro (RRID: Addgene 86849) was employed to create the cell cycle reporter (FastFUCCI reporter). This plasmid encodes two fluorescent probes (mKO2‐hCDT1 and mAG‐hGEM) that are used to distinguish different phases of the cell cycle. The FastFUCCI reporter was stably transduced into endothelial cells by lentivirus to form the FUCCI reporter cell line. The indicator cells exhibited red-, yellow-, and green-emitting populations, representing cells in the G1, G1/S-, and S- or G2-to-M phases, respectively. Through the gain- or loss-of-function experiments on the FUCCI reporter cell line, we could explore the effect of targeted gene(s) on cell cycle and cell proliferation.
Spheroid Sprouting Assay
HCMECs were seeded in a round bottom 96-well plate and cultured with a complete medium comprising 0.25% (w/v) methylcellulose for 24 h using hanging drops to create spheroids. The spheroids were embedded in a mixed collagen solution (formula as described previously), and the volume of the solution was calculated based on the number of spheroids, resulting in an approximate concentration of 50 spheroids/ml. The mixture was then introduced to the 24-well plate (1 ml/well) and polarized for 30min. After incubation with ECM containing 50 ng/ml VEGF for 24 h, the collagen with spheroids was fixed with 4% paraformaldehyde, photographed, and analyzed.
Dual-luciferase reporter assay
The genomic DNA was implemented to amplify the SLUG promoter region (–2089/ + 235) while the One Step Cloning Kit (C112-02, Vazyme Biotech Ltd., China) was deployed to insert the pGL3-Basic vector (Promega). The pRL-TK plasmid (Renilla luciferase reporter plasmid, Promega), which operates as an internal control, was co-transfected into HEK293T cells along with the Luciferase reporter structures. The Dual-Luciferase Reporter Assay Kit (Promega) was implemented to ascertain the luciferase activity, aligning with the manufacturer's recommendations.
Pulldown assay with Biotin-labeled RNA probes and mass spectrometry
Typically, 3 µg of biotin-labeled sense and antisense RNA probes were incubated at 4°C for 1 h with streptavidin magnetic beads. Subsequent to a first mild wash with the buffer solution (20 mM Tri-HCl, pH7.5, 1 mM EDTA, and 450 mM NaCl), the beads were then exposed to cell lysates at 4°C for an interval of 4 h. This was afterward followed by five further washes. Following that, the binding proteins were subjected to immunoblot analysis or mass spectrometry (Bioprofile Technology Co., Ltd, Shanghai, China). Probe sequences are shown in the Supplementary table 3.
RNA-IP assay
The cells were cross-linked using UV radiation at a dosage of 200 J/cm2 and a wavelength of 254 nm. Next, the cells were lysed with a solution comprising protease inhibitors and RNase inhibitors. The resulting mixture was then homogenized and centrifuged. The supernatant, was afterward incubated with anti-ALKBH5 and rabbit IgG for a duration of 4 h with gentle rotation. Next, Protein A/G Agarose beads (Millipore, USA) were introduced into the mixes and left to rotate for an additional 1 h. The immunoprecipitated RNA was purified using Trizol Reagent after being washed three times. It was then subjected to RT-qPCR analysis using primers. All processes were conducted on ice or at a temperature of 4°C.
MeRIP-qPCR
The MeRIP test was derived from a previously published methodology [27]. To summarize, the undamaged poly-A-purified RNA was denatured at 70°C for 10 min. Subsequently, the specimen was rapidly chilled with ice and mixed with m6A antibody in a 1ml solution comprising RNasin Plus RNase inhibitor 400 U (Promega, USA), 50 mM Tris-HCl, 750 mM NaCl, and 0.5% (vol/vol) Igepal CA-630 (Sigma Aldrich, USA). The combination was incubated for 2 h at 4°C. The mixture was incubated at 4°C with rotation for 2 h, following the Dynabeads Protein G (Invitrogen, USA) were purified and introduced to it. The m6A RNA was eluted two times using a solution containing 6.7 mM N6-methyladenosine 5′-monophosphate sodium salt. The rinses were conducted at 4°C for 1 h. Afterward, it was precipitated with 5 µg of glycogen, one-tenth of the amount of 3 M sodium acetate, and 2.5 times the volume of 100% ethanol. The precipitation procedure was conducted overnight at − 80°C. The m6A enrichment degree was evaluated using qPCR analysis. The fragmented mRNA was immediately incubated with a solution comprising m6A antibodies and subjected to comparable treatment. Primer sequences are shown in the Supplementary table 3.
Study population
Patients with acute myocardial infarction (AMI) and lower limb ischemia, comparing them with healthy volunteers. All participants were recruited from the First Affiliated Hospital of Zhengzhou University. The control group consisted of healthy volunteers. These volunteers had no significant systemic diseases, such as ischemic heart disease, cancer, pulmonary disease, or infectious diseases. Patients diagnosed with acute myocardial infarction (AMI) were included in the study. Blood samples were collected prior to any surgical intervention, primarily on the first day of admission. Patients with clinically diagnosed lower limb ischemia were also included. Blood samples were taken upon admission and prior to any surgical or interventional treatments. For all groups, blood samples were collected under standard clinical conditions. Clinical characteristics, vital signs, laboratory test results, and radiological reports were extracted from electronic medical records to complement the blood sample data. A total of 32 healthy volunteers, 32 acute myocardial infarction patients and 32 nondiabetic patients with a car accident were recruited in this study as well. Clinical characteristics, vital sign, laboratory tests, radiological reports were extracted from electronic medical records, between January 2023 and March 2024. The main clinical and laboratory data of human subjects were summarized in Supplementary Table 5.
Extraction of exosomes
Extraction of exosomes was performed as described previously with minor modifications [28]. Briefly, Cellular components in fresh blood samples were eliminated with centrifugation (two times 2,500×g, 4°C, 15 min). Supernatant (i.e., platelet-free plasma) was diluted to 2× with phosphate-buffered saline (PBS), and was filtered through a 0.8 µm filter (Merck, Darmstadt, Germany) by hydrostatic pressure to remove remaining platelets and apoptotic bodies. Then, the samples were centrifuged at 13,200 × g for 22 min at 4°C. The supernatant was filtered through 0.22 µm filters, and exosomes were pelleted by ultracentrifugation at 120,000×g for 1h, 3h, 6h, or 14h at 4°C. Additional conditions included ultracentrifugation at 37°C with protease inhibitors or using 5× diluted plasma. The pellets were washed with PBS and re-centrifuged at 120,000×g. Exosomes were used fresh to maintain integrity.
Statistical analysis
The data were presented as the mean value ± the standard error of the mean (s.e.m.). The cutoff values for gene expression were determined based on the median or mean levels. We used Shapiro-Wilk test to evaluate data normality. The differences were evaluated using Student's t-test, analysis of variance (ANOVA). For non-normally distributed data across multiple groups, we employed Kruskal-Wallis test. The expression correlation was assessed with the Pearson correlation coefficient analysis. The Log-rank test was deployed to evaluate the disparity in survival analysis. The two-sided Student’s t-test was implemented as to evaluate the expression difference, and the p-values below 0.05 were deemed significant.