
[ad_1]
Antibodies and reagents
Primary antibodies against AP-2α (#3208, dilution: 1:1000), vWF (#65707, dilution: 1:1000), and GAPDH (#5173, dilution: 1:1000) were obtained from Cell Signaling Technology (Boston, MA, USA). Primary GTPCH1 antibody (#ab307507, dilution: 1:1000) was from Abcam. Primary phosphorylated AP-2α at serine 219 antibody was generated by Genscript Company (dilution: 1:1000) as we described previously29. HRP-conjugated Affinipure Goat Anti-Rabbit IgG (Hâ+âL) (SA00001-1, dilution: 1:5000) were from Proteintech. Lovastatin (#438186), pravastatin (#1554206), atorvastatin (#524403), angintensin II (#05-23-0101), dihydroethidium (#309800), diaminofluorescein (#D224), and oxidized low-density lipoprotein (#AB3230) were purchased from Sigma-Aldrich Company (USA). High fat diet was purchased from Research Diet (D12492). Commercial kits for determinations of glucose, cholesterol, and triglyceride, LDL-C, and HDL-C were purchased from Jian-Cheng Bioengineering Institute (Nanjing, China). All drug concentrations are expressed as working concentrations in the buffer.
Generation of AP-2α
flox/flox mice
Targeting vectors to generate AP-2αflox/flox mice were constructed using a bacterial artificial chromosome (BAC, B6Ng01-349O17) recombineering (see Supplementary Fig. 8A for details). Bruce4 ES cells41 were electroporated with the targeting vector and positive clones identified by PCR and southern blotting were injected into blastocysts from C57BL/6-Tyrc-2J mice. The resulting male chimaeric mice were bred to female C57BL/6-Tyrc-2J mice to obtain germline transmission. The FRT-Neo-FRT cassette was removed by crossing with Flp deleter mice, then the mice were backcrossed at least three additional times onto a C57BL/6 background.
BAC modification
A homologous recombination-proficient E. coli strain (DY380) was used for the BAC recombineering42. The 1st step of the process is the homologous recombination of the BAC using the modification cassette I. It occurs by crossing over between the homology arms and the genome in the BAC. In this case, recombination results in the incorporation of the modification cassette sequences into the genome to yield the modified BAC1 (mBAC1). The 2nd step of the process is the homologus recombination of the mBAC1 using the modification cassette II. It occurs by a second homologous recombination event that occurs within the mBAC1. Recombination yields the precisely modified BAC2 (mBAC2), with the modification cassette II inserted at the correct position in the BAC. This modified BAC carries the loxP-3â region for introduction to the HAC vector. In brief, overnight cultures containing the BAC were grown from single colonies, diluted 10-fold in LB medium, and grown to an optical density at 600ânm of 0.4â0.6 at 32â°C. Fifty milliliter cultures were then induced for the expression of recombineering factors by shifting the cells to 42â°C for 15âmin followed by chilling on ice for 10âmin. Cells were then centrifuged for 5âmin at 5500 X g at 4â°C and washed with 10âmL of ice-cold 1âmM HEPES 2 times. Cells were then resuspended in 100âμL of ice-cold 1âmM HEPES and electroporated. Cell transformation was performed by electroporation of 1âμg linear DNA into 100âμL of ice-cold competent cells in cuvettes (0.1âcm) using a Bio-Rad gene pulser set at 1.75âkV, 25âμF with a pulse controller set at 200âohms. One milliliter of SOC medium was added after electroporation. Cells were incubated at 32â°C for 1âh with shaking and spread on appropriate selective agar media.
Animals and induction of a âtwo-hitâ model of HFpEF
Male WT mice were obtained from Beijing Huafukang Company (Beijing, China). The CDH5-CreERT2 transgenic mouse exhibits tissue-specific expression of an inducible Cre-ERT2 fusion protein, enabling tamoxifen-induced Cre recombinase activity in vascular endothelial cells43, and were obtained from Taconic Biosciences, Inc (Model #13073). AP-2αflox/flox/CDH5-CreERT2 mouse was generated by crossing AP-2αflox/flox mice with CDH5-CreERT2 mice. Male CDH5-CreERT2/AP-2αflox/flox mouse was injected with tamoxifen (2âmg/mouse) for five consecutive days to induce endothelium-specific AP-2α gene knockout. Male CDH5-CREERT2 mouse injected with tamoxifen serves as a control because of endothelial CreERT2 toxicity44,45.
To induce HFpEF as the comorbidities of hypertension and obesity24, mice received HFD administration plus AngII infusion at a rate of 0.2âmg/kg per day for 12 weeks using a miniosmotic pump (Alzet), as reported previously with minor modifications46,47,48. This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The animal protocol was reviewed and approved by the Animal Care and Use Committee, Qilu Hospital of Shandong University.
Conventional echocardiography (ECG) and doppler imaging
Transthoracic ECG was performed using a VisualSonics Vevo 2100 system equipped with an MS400 transducer (Visual Sonics). LVEF and other indices of systolic function were obtained from short-axis M-mode scans at the midventricular level, as indicated by the presence of papillary muscles, in conscious, gently restrained mice. Anesthesia was induced by 5% isoflurane and confirmed by a lack of response to firm pressure on one of the hindpaws. During echocardiogram acquisition under body-temperature-controlled conditions, isofluorane was reduced to 1.0â1.5% and adjusted to maintain a heart rate in the range of 400â500Pâbeats per min.
To assess diastolic function, E, and Eâ²were measured. After measurement of the parasternal short-axis view, the sonographic probe was tilted 45 degrees to visualize the parasternal 4-chamber view. Mitral E waves were recorded with pulse-wave mode at the mitral valve opening. Mitral annulus movement, also known as Eâ² wave, was assessed from the medial mitral valve annulus with tissue velocity image mode.
Parameters collected include: A, peak Doppler blood inflow velocity across mitral valve during late diastole; mitral DT, mitral early filling deceleration time; E, peak Doppler blood inflow velocity across mitral valve during early diastole; Eâ², peak tissue Doppler of myocardial relaxation velocity at mitral valve annulus during early diastole; HR, heart rate; IVRT, isovolumic relaxation time; IVS,d, end-diastolic interventricular septal wall thickness; LVEF, left ventricular ejection fraction; LVID,d, left ventricular internal diastolic diameter; LVID,s, left ventricular internal systolic diameter; LVPW,d, left ventricular end-diastolic posterior wall; LVFS, left ventricular fractional shortening.
Blood pressure measurement
Blood pressure was determined by a left carotid catheter method at the end of animal experiments49. Mice were anesthetized with a ketamine and xylazine mixture and placed under warm light (37â°C). A catheter was inserted into the left common carotid artery, with the aid of a dissecting microscope, to measure arterial blood pressure. For catheter insertion, the left common carotid artery was carefully exposed via a 0.5â1.0âcm midline incision in the ventral neck region. The tip of the artery toward the head was ligated with a suture, and the tip toward the heart was occluded with a microclip. A small cut was then made in the vessel wall using microscissors. A 60â-cm catheter (PE10) containing a sterile 10% heparin-90% saline solution was inserted into the artery a distance of 0.65âcm toward the thorax. The arterial clip was removed, and the catheter was tied in place. Blood was directed to a pressure transducer through the catheter to obtain computerized blood pressure measurements (AD instruments). The mice were allowed to recover and the systolic and diastolic blood pressures and heart rate were monitored for at least 30âmin in conscious states.
Exercise exhaustion test
After three days of acclimatization to treadmill exercise, an exhaustion test was performed in the experimental groups of mice. Mice ran uphill (20°) on the treadmill (Columbus Instruments) starting at a warm-up speed of 5âm/min for 4âminutes after which speed was increased to 14âm/min for 2âminutes. Every subsequent 2âmin, the speed was increased by 2âm/min until the mouse was exhausted. Exhaustion was defined as the inability of the mouse to return to running within 10âs of direct contact with an electric-stimulus grid. Running time was measured and running distance was calculated.
Generations of AAV9 and infection to mice
The adeno-associated virus 9 (AAV9) construction compassing cDNA (AAV9-TIE-cDNA) was generated by according to the manufacturersâ recommendations from Shanghai Genechem Co., Ltd. (Shanghai, China). The endothelial cell specific promoter is âpAAV-TIEp-EGFP-MCS-3Flag-SV40 PolyAâ. Viruses were packaged and amplified in HEK293A cells and purified using CsCl2 banding followed by dialysis against 10âmM Tris-buffered saline with 10% glycerol. Titering was performed on HEK293 cells using the adeno-X Rapid Titer kit (BD Biosciences Clontech, PaloAlto, CA, USA) according to the manufacturerâs instructions.
Animal experimental protocols
In the first part of the animal study (Supplementary Fig. 5A), male WT mice received lovastatin administration (4âmg/kg/day) one week prior to HFD plus AngII treatments for another 12 weeks. In the second part of the animal study (Supplementary Fig. 8C), a male AP-2αflox/flox/CDH5-Cre-ERT2 mouse was injected with tamoxifen (2âmg/mouse) or vehicle for five consecutive days to induce endothelium-specific AP-2α gene knockout. Then mice received lovastatin administration (4âmg/kg/day) one week prior to HFD plus AngII treatments for another 12 weeks. In the third part of the animal study (Supplementary Fig. 10A), male WT mice were infected with AAV9 expressing negative control (NC) shRNA or circRNA-RBCK1 shRNA via tail vein injection. One week later, mice were treated with lovastatin (4âmg/kg/day) one week prior to HFD plus AngII treatments for another 12 weeks. An injection of AAV9 was repeated once in 4 weeks. For in vivo infection, AAV9 were injected via tail vein in 100âµl of PBS containing 1 Ã1011 IFUs of loaded virus per mouse. The concentration of DNA was 10âmg/kg. Before mice sacrificed, conventional ECG and Doppler imaging were assessed.
Hematoxylin-eosin staining
Slides with section were placed in a metal staining rack and immersed in the filtered Harris Hematoxylin for 10âs. Then, the sections were incubated in EOSIN stain for 30âs. Dehydration was performed in ascending alcohol solutions (50%, 70%, 80%, 95% X 2, 100% X 2) followed by clearance with xylene (3-4 X) in Columbia staining jars. The slides were mount using Permount (xylene based).
Massonâs trichrome staining
The heart was fixed with 4% paraformaldehyde overnight at room temperature, dehydrated sequentially through ethanol, butyl alcohol, and embedded in paraffin. Hearts were serially cut from the apex to the base perpendicular to the long axis. Transverse sections (4âμm) were stained with Massonâs trichrome. Images were acquired on Panoramic MIDI (3D HISTECH Inc., Hungary).
Quantitative PCR
Total RNA was isolated using a TRIzol-based (Invitrogen) RNA isolation protocol. RNA was quantified by Nanodrop (Agilent Technologies), and RNA and miRNA quality were verified using an Agilent 2100 Bioanalyzer (Agilent Technologies). Samples required 260/280 ratios of more than 1.8, and sample RNA integrity numbers of more than 9 for inclusion. RNA was reverse transcribed using the TaqMan microRNA Reverse Transcription Kit (Applied Biosystems) according to the manufacturerâs instructions. Quantification of circRNA and mRNA was performed using an ABI PRISM7500 system, and miRNA concentrations were determined using an ABI PRISM7900 system (Applied Biosystems, Carlsbad, CA, USA). Before calculation using the ÎÎCt method, the levels of GAPDH were used to normalize the relative expression levels of circRNA and mRNA, and the levels of small nuclear U6 were used to normalize the miRNA expression levels. Primers used for quantitative PCR were listed in Supplementary Table 7.
Mutagenesis of miR-133a binding sites in circRNA-RBCK1
These reporter constructs were generated in two steps. First, a coding-region fragment containing the miR-133a binding sites was generated by PCR and cloned into the pMIR luciferase vector (Ambion) using SpeI and MluI cloning sites. Next, site-directed mutagenesis was performed, introducing three mutations into the binding siteâs seed sequence in the 3â²-UTR of circRNA-RBCK1. Subsequently, a DNA fragment containing the 3â²-UTR with the mutant of miR-133 binding site (MT1, MT2, or MT3) was generated by PCR and cloned into pMIR vector, this time using MluI and HindIII sites. Again, site-directed mutagenesis was used to change all three binding sites within the seed sequence (MT1/2/3). All constructs were sequenced to confirm their identity.
Plasmid transfection into HEK293 and reporter assays
The sequences were synthesized and constructed into the pmirGLO luciferase plasmid vector, and promoter sequences were integrated and inserted into the GV272 vector by Genechem company (Shanghai, China). The plasmid constructs were co-transfected in HEK293 cells with the pCMV β-gal plasmid and 50ânM each of chemically synthesized miRNA oligonucleotides (Applied Biosystems) by using lipofectamine 2000 (Invitrogen). Cells were harvested 48âh after transfection, and luciferase and β-galactosidase activities were measured. The luciferase assay was conducted using a dual luciferase reporter assay (Vazyme, China) according to the manufacturerâs instructions.
Cell cultures and virus infection
Primary HUVECs, human EECs, human CAECs, human MCECs, and human AECs were obtained from Clonetics Inc. (Walkersville, MD, USA). Cells were grown in endothelial basal medium supplemented with 2% fetal bovine serum and penicillin (100âu/ml), and streptomycin (100âµg/ml). Cultured cells were used between passages 3 and 8. All cells were incubated in a humidified atmosphere of 5%CO2â+â95% air at 37â°C. When 70â80% confluent, the cells were treated with different agents. For HEK293 cells, cells were cultured in M200 medium supplemented with 2% fetal bovine serum and penicillin (100âu/ml), and streptomycin (100âµg/ml). For lentivirus infection, cells were infected with lentivirus expressing AP-2α shRNA or circRNA-RBCK1 shRNA from Shanghai Genechem Co., Ltd. (Shanghai, China) overnight in antibiotics-free medium supplemented with 2% FBS. The target sequence of CircRNA-RBCK1 shRNA is TCTTGCAGCAGTGGGTGATTG. The target sequence of AP-2α shRNA is TCCCAGATCAAACTGTAATTA. These targets were designed by VectorBuilder Inc. The cells were then washed and incubated in fresh medium for an additional 12âh before experiments.
Isolations of myocardial capillary endothelial cells, endocardium endothelial cells, coronary arterial endothelial cells, aortic endothelial cells from mice
For isolations of mouse aortic endothelial cells and mouse endocardium endothelial cells, heart and aorta were rapidly excised from mice (Supplementary Fig. 7A). Then, left ventricle and the aorta were injected with PBS containing 0.1% collagenase for 15âmin at 37â°C. After centrifugation at 250âg for 10âmin, the cell pellets were collected for RNA extraction immediately.
For isolation of mouse myocardial capillary endothelial cells, the distal left anterior descending coronary artery was cannulated, and the epicardial and endocardial surfaces were removed (Supplementary Fig. 7B). The remaining myocardial tissue was digested in PBS containing 0.1% collagenase at 37â°C for 30âmin, with gentle rotation. Digested tissue was pelleted at 250âg and resuspended in DMEM containing 20% BSA (Sigma) (w/v), then myelin fraction was separated by centrifugation at 1000âg for 10âmin. The cell pellet was resuspended and filtered through a 70âµM nylon mesh and collected following centrifugation at 250âg. The cell pellets were collected for RNA extraction immediately.
For isolation of mouse coronary arterial endothelial cells, the remained heart tissues by cutting the distal left anterior descending coronary artery were injected with PBS containing 0.1% collagenase for 15âmin at 37â°C through the aortic root (Supplementary Fig. 7B). After centrifugation at 250âg for 10âmin, the cell pellets were collected for RNA extraction immediately.
RNA extraction and sequencing data analysis
Total RNA from 6 samples was extracted. We have generally utilized 100âng of RNA for library construction for MeRIP-circRNA sequencing. Briefly, the mRNA with polyA in the total RNA was enriched by Oligo-dT magnetic beads. The intact mRNA was then fragmented using an ultrasound machine. The segmented RNA was divided into two parts. One part was added to an m6A-capturing antibody to enrich the mRNA fragments containing m6A methylation (MeRIP-seq), and the other part was used as an Input to directly construct a conventional transcriptome sequencing library (circRNA-seq). The conventional sequencing library was constructed according to the transcriptome library construction process. Illumina Hiseq X Ten was used for high-throughput sequencing of the library. The circBase database and Circ2Traits were used to annotate the identified circRNA. Then, DESeq2 software (v1.14.1) was used for data standardization and differentially expressed circRNA screening (log2FCââ¥â1.5, p-valueââ¤â0.05).
Biotinylated RNA pull-down assay
The biotinylated-circRNA-RBCK1 probe was incubated with C-1 magnetic beads (Life Technologies, Carlsbad, CA, USA) to generate probe-coated beads, then incubated with sonicated HEK293 cells at 4â°C overnight, followed by eluted and quantitative RT-PCR. For miR-133a pulled down circRNA-RBCK1, HEK293 cells with circRNA-RBCK1 overexpression were transfected with biotinylated miR-33a mimics or mutant using Lipofectamine 2000. The cells were harvested, lysed, sonicated, and incubated with C-1 magnetic beads (Life Technologies, Carlsbad, CA, USA), followed by quantitative RT-PCR.
Chromatin-immunoprecipitation assay for the binding of AP-2α and RBCK1 gene promoter
The binding between AP-2α and RBCK1 gene promoter was performed using a bioinformatic analysis (http://genexplain.com/transfac). According to the scores, ChIP assays were performed by using a ChIP-IT kit (Upstate, 17-295), according to the manufacturerâs protocol. 1âÃâ106 cells were seeded on a 10âcm dish. Proteins were cross-linked to DNA by adding formaldehyde directly to the culture medium at a final concentration of 1% and incubating for 10âmin at 37â°C. The cells were harvested in SDS lysis buffer and added protease inhibitors. Cell lysates were sonicated to shear DNA to lengths between 200 and 1000âbp. Sheared chromatin was precleared with protein G beads prior to incubation overnight at 4â°C with 4âµg of AP-2α antibody or IgG antibody. Purified, immunoprecipitated chromatin fragments from IP samples were subjected to PCR. PCR products were subjected to agarose gel electrophoresis and stained with ethidium bromide. The primer sequences were as follow: forward, 5â-attcatgtgcaaacggggc-3â, and reverse, 5â-aggcgacccggaggtagcatt-3â.
Electrophoretic mobility shift assay for AP-2α activity
Subcellular fractions were prepared using NE-PER Nuclear and Cytoplasmic Extract kit (Cat78833) from PIERCE. EMSA were performed following the commercial kits. AP-2α kit (AY1002) is from Panomics Company.
RNA fluorescence in situ hybridization
The RNA fluorescence in situ hybridization assay was performed by using a FISH kit (RiboBio, Guangzhou, China) according to the manufacturerâs guidelines. Cy3-labeled circRNA-RBCK1 probes and Dig-labeled locked nucleic acid miR-133a probes (Ribo-Bio, Guangzhou, China) were measured by the FISH kit, followed by visualized with a confocal microscopy.
Immunofluorescence analysis
Sections were deparaffinized, rehydrated, and blocked with 5% normal serum. Incubate tissues with primary antibody for 1âh at room temperature or overnight at 4âoC. After washing, incubate with fluorescence-conjugated secondary antibody for 45âminutes. Digital images were captured under a fluorescence microscopy. Quantitative analysis was performed by calculating fluorescence intensity using Alpha Ease FC software (version 4.0 Alpha Innotech).
Western blotting
Tissues were homogenized on ice in cell-lysis buffer (20âmM Tris-HCl, pH 7.5, 150âmM NaCl, 1âmM Na2EDTA, 1âmM EGTA, 1% Triton, 2.5âmM sodium pyrophosphate, 1âmM beta-glycerophosphate, 1âmM Na3VO4, 1âµg/ml leupeptin) and 1âmM PMSF. Cell was lysated with cell-lysis buffer. The protein content was assayed by BCA protein assay reagent (Pierce, USA). 20âµg proteins were loaded to SDS-PAGE and then transferred to membrane. Membrane was incubated with a 1:1000 dilution of primary antibody, followed by a 1:2000 dilution of horseradish peroxidase- conjugated secondary antibody. Protein bands were visualized by ECL (GE Healthcare). The intensity (area X density) of the individual bands on Western blots was measured by densitometry (model GS-700, Imaging Densitometer; Bio-Rad). The background was subtracted from the calculated area.
Detection of ROS
Cells were incubated with DHE (10âµM) for 30âmin, homogenized, and subjected to methanol extraction. HPLC was performed using a C-18 column (mobile phase: gradient of acetonitrile and 0.1% trifluoroacetic acid) to separate and quantify oxyethidium (product of DHE and O2–) and ethidium (a product of DHE auto-oxidation). ROS level was determined by conversion of DHE into oxyethidine. To measure ROS production in the artery in situ, fresh frozen sections of MCA were isolated from mice, and were stained with 10âμM DHE for 30âmin, rinsed, and observed by fluorescent microscopy. Results were quantified using BIOQUANT Image software.
Detection of intracellular NO
NO production in culture cells was detected using the fluorescent probe. Briefly, before the end of treatment, 10âµM DAF was added to the medium and incubated for 30âmin at 37â°C, then washing with PBS twice. The DAF fluorescent intensity was recorded by fluorescent reader at the wave of excitation (485ânm) and emission (545ânm).
Measurement of BH4
Homogenates of aorta or cell lysates were suspended in distilled water containing 5âmM dithioerythrol, centrifuged at 12,000âg at 4â°C for 10âmin, and then subjected to oxidation in acid or base. To 100âμl aliquot of supernatant, 20âμl of 0.5âM HCl and 0.05âM iodine were added for acidic oxidation, and 20âμl of 0.5âM NaOH plus 0.05âM iodine were added for basic oxidation. After incubation for 1âh in the dark at room temperature, 20âμl HCl was added to the basic oxidation only. All mixtures received 20âμl of 0.1âM ascorbic acid for the reduction of excess iodine. Samples were then centrifuged for 10âmin at 12000âg at 4â°C. Biopterin concentrations were determined by HPLC with a PR-C18 column. Elution was at a rate of 1.0âml/min of 50âmM potassium phosphate buffer, pH 3.0. Fluorescence was detected with an excitation at 350ânm and emission at 440ânm. BH4 concentrations were calculated as the difference in results from oxidation in acid and base.
Measurements of blood glucose, cholesterol, and triglyceride
Blood glucose, homocysteine, cholesterol, triglyceride, LDL-C, and HDL-C were assayed by using commercial kits as recommend by the protocol.
Statistical analysis
All quantitative results were expressed as meanâ±âSD. The normal distribution of data was tested by the Kolmogorov-Smirnov test before statistical comparisons, and the normality/equal variance was tested to determine whether ANOVA was appropriate. A one-way ANOVA followed by Tukeyâs HSD test, Scheffe test or Dunnett test were used to multiple comparisons between two groups. Statistical analysis was conducted using IBM SPSS statistics 20.0 (IBM Corp., Armonk, NY, USA). Pâ<â0.05 was considered significant. GraphPad Prism version 8 (GraphPad Software, San Diego, CA, USA, www.graphpad.com) was used to make figures.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
[ad_2]
Source link