Abstract The transition of healthy contractile vascular smooth muscle cells to an inflammatory and senescent phenotype is a key driver of abdominal aortic aneurysm (AAA). Although CD147 is highly expressed in VSMCs and upregulated in aneurysmal tissue, the precise role of VSMC-derived CD147 in phenotypic switching and AAA pathogenesis remains elusive. Here, we identified a previously unrecognized nuclear localization of CD147 in VSMCs, and pathological stimuli upregulated the nuclear CD147 expression through reactive oxygen species-dependent mechanisms. Multi-omics analysis integrating RNA sequencing, CUT&Tag, and protein interactome profiling revealed that nuclear CD147 directly interacts with the STAT1/STAT2 complex to activate the IRF7-IFNα/β axis under oxidative stress (H[2]O[2] exposure), thereby driving VSMC senescence and inflammatory reprogramming. Functionally, CD147 deletion in VSMCs significantly mitigated Angiotensin II- and CaPO[4]-induced AAA formation, accompanied by improved VSMC phenotype, reduced vascular inflammation and extracellular matrix degradation in vivo. Pharmacological inhibition of CD147 using Myricetin, a food-derived natural small-molecule compound, effectively discouraged oxidative stress-induced VSMC fate transition in vitro, and suppressed AAA progression and improved vascular integrity in two murine AAA models, underscoring its therapeutic potential. Collectively, these findings identify CD147 as a key driver of interferon-mediated VSMC fate transition, providing mechanistic insights into AAA progression and a promising therapeutic target for vascular diseases. Keywords: CD147, Type I interferon, Vascular smooth muscle cell, Abdominal aortic aneurysm, Myricetin, Oxidative stress Graphical abstract [41]Image 1 [42]Open in a new tab Highlights * • Nuclear CD147 drives VSMC fate transition via activating type I interferon signaling axis under oxidative stress. * • VSMC-specific deletion of CD147 significantly attenuates AngII- and CaPO[4]-induced AAA formation in vivo. * • Myricetin, a natural CD147 inhibitor, alleviates oxidative stress-induced VSMC senescence and inflammatory reprogramming. Myricetin treatment reduces AAA progression in vivo. 1. Introduction Abdominal aortic aneurysm (AAA) is the second most prevalent aortic disease following atherosclerosis characterized by the abnormal and permanent dilation of the abdominal aorta. Although an abdominal aortic aneurysm can remain asymptomatic for a long time, sudden rupture leads to the life-threatening hemorrhage with a high mortality rate [[43]1]. Despite advancements in surgical techniques, including open repair and endovascular aneurysm repair, the lack of effective pharmacological interventions to prevent AAA progression remains a significant challenge [[44]1]. Understanding the molecular mechanisms underlying AAA formation is crucial for identifying new therapeutic strategies. As the principal cellular component of healthy blood vessels, vascular smooth muscle cells (VSMCs) exhibit a quiescent, contractile phenotype under physiological conditions to maintain the structural integrity of the vasculature; however, pathological stimuli drive their transition to a senescent, inflammatory, and extracellular matrix-secreting phenotype, thus contributing to AAA development [[45]2,[46]3]. On one hand, as a contributor to vascular inflammation, inflammatory VSMCs exhibit high expression of innate immune pattern recognition receptors such as Toll-like receptors and Nod-like receptors [[47]4,[48]5]. Upon activation, VSMCs secrete a repertoire of pro-inflammatory cytokines, thereby recruiting macrophages, neutrophils, and other immune cells to the vascular wall, exacerbating inflammatory responses. On the other hand, inflammatory cytokines and reactive oxygen species (ROS) within the vascular microenvironment downregulate contractile gene expression in VSMCs and promote cellular dysfunction and senescence [[49][6], [50][7], [51][8]]. Moreover, the cGAS-STING signaling pathway, a critical component of innate immune cascades, is highly activated in damaged VSMCs. It senses cytosolic leakage of mitochondrial and nuclear DNA, triggering an autocrine/paracrine type I interferon (IFN–I) response, primarily involving IFNα and IFNβ, that drives VSMC senescence and phenotypic transition [[52]3,[53]9,[54]10]. Despite being the first line of defense against vascular injury and pathological stimuli, the precise mechanisms underlying VSMC-mediated vascular inflammation remain incompletely understood. Further refinement of these pathways may provide novel targets for blocking VSMC phenotypic alteration and preventing aortic aneurysm formation and progression. CD147 (cluster of differentiation 147, also known as EMMPRIN or basigin), encoded by the BSG gene of the immunoglobulin superfamily, is a multifaceted molecule with diverse regulatory roles due to its interactions with various molecular partners [[55][11], [56][12], [57][13]]. Targeting CD147 has a protective role in inflammation, various cancers, and COVID-19 [[58][13], [59][14], [60][15]]. Previous studies from our group and others have identified CD147 as a critical therapeutic target in cardiovascular diseases [[61][16], [62][17], [63][18]]. For instance, targeting pressure overload-induced CD147 overexpression effectively attenuates heart failure and pathological cardiac hypertrophy [[64]18,[65]19]. Similarly, inhibiting myeloid-derived CD147 prevents atherosclerotic plaque progression by reducing inflammation and enhancing efferocytosis [[66]20,[67]21]. Interestingly, CD147 upregulation has been observed in angiotensin II (AngII)-stimulated VSMCs [[68]22], elastase- or AngII-induced murine aneurysmal aortas [[69]23,[70]24], and human aneurysmal specimens [[71]22,[72]25,[73]26]. Moreover, systemic inhibition of CD147 has been shown to attenuate AAA formation in mice [[74]24]. However, the direct role of CD147 in VSMC biology and its mechanistic contribution to AAA pathogenesis remain largely unexplored. Moreover, exploration of therapeutic agents targeting CD147 remains urgent. In the present study, we found that CD147 is obviously expressed in the nucleus of VSMCs and promotes oxidative stress-induced VSMC inflammation and senescence by transcriptionally activating the IRF7-IFNα/β signaling pathway through binding to STAT1/2. VSMC-specific knockout of CD147 or treatment with its natural inhibitor Myricetin (3,3,4,5,5,7−hexahydroxyflavone, Myr) significantly alleviated AAA in vivo. 2. Material and methods 2.1. Animal The animal protocols for this study were approved by the Ethics Committee of Renji Hospital, School of Medicine, Shanghai Jiao Tong University and all procedures were conducted in accordance with the National Institutes of Health Guidelines for the Care and Use of Laboratory Animals (8th Edition, 2011). VSMC-specific CD147-knockout mice (CD147^ΔSMC) were generated by crossing Tagln-Cre transgenic mice with CD147-floxed mice (on a C57BL/6J background), in which exons 2–8 of the CD147 gene were flanked by two loxP target sites (Cyagen Biosciences, Guangzhou, China). Then, CD147^ΔSMC mice were bred on an ApoE^−/− background to establish ApoE^−/−/CD147^ΔSMC mice. Littermate controls were homozygous for the floxed CD147 allele (ApoE^−/−/CD147^flox/flox). All mice were maintained in a controlled environment at 24 ± 2 °C and 40–60 % humidity with a 12-h light/dark cycle and ad libitum access to food and water. Animal experiments were performed by two independent investigators in a blinded manner. Randomization was carried out using the RAND function in Excel, generating random group assignments based on body weight, with group allocations concealed from the experimenter. Sample sizes for the mouse aortic aneurysm studies were calculated using a priori power analysis via PASS software, based on data from similar AAA models in our research group [[75][27], [76][28], [77][29]]. A sample size of more than 8 animals per group was determined to achieve 90 % power (β = 0.1) to detect significant changes in maximal aortic diameters at an α value of 0.05. Each mouse served as an independent experimental unit. AngII- or calcium phosphate (CaPO[4])-induced AAA models were established in 8- to 10-week-old male CD147^flox/flox and CD147^ΔSMC mice (18–22g), with or without ApoE knockout background, as previously reported [[78][27], [79][28], [80][29]]. To evaluate the effects of Myricetin (Myr) (S2326, Selleck, TX, USA) on AAA, AngII-infused ApoE^−/− mice and CaPO[4]-treated AAA mice were treated with Myr (10 mg/kg/day) or vehicle via intraperitoneal injection for 28 days. Myricetin was dissolved in a solution containing 5 % DMSO, 40 % PEG300, 5 % Tween 80, and 50 % sterile water at a concentration of 2.5 mg/ml. At the end of study, all mice were euthanized by cervical dislocation under deep anesthesia, and aortas were harvested at 28 days post-implantation. Aneurysmal aortas were defined as those exhibiting a diameter increase of 50 % or more. Mice that died before the study endpoint were subjected to autopsy, and aneurysm rupture was identified by the presence of blood clots outside the adventitia of the dilated aortic wall. 2.2. In vivo ultrasound imaging analysis Mice were positioned in a supine orientation on the operation platform and anesthetized with isoflurane. Abdominal hair was removed using depilatory cream. Aortic imaging was performed using a Vevo 2100 high-resolution ultrasound system (Fujifilm VisualSonics, Toronto, Canada). Initially, the probe was applied along the long axis to visualize and capture morphological images of the abdominal aorta. Subsequently, the probe was rotated to the short axis for precise measurement of the maximal aortic diameter (B-mode). 2.3. Blood pressure measurements Noninvasive measurements of blood pressure were performed in conscious and trained mice using tail-cuff plethysmography (BP-2010A, Softron Biotechnology), as previously described [[81][17], [82][28], [83][29]]. 2.4. Isolation, culture, and treatment of primary cells Primary VSMCs were isolated from the aortas of Sprague-Dawley rats (150–180 g), CD147^flox/flox or CD147^ΔSMC mice via collagenase digestion, following established protocols. Briefly, aortic tissues were dissected, rinsed in cold phosphate-buffered saline (PBS), and stripped of endothelium and adventitia. The tissues were then minced into approximately 1 mm^3 fragments and enzymatically digested with 1 % type II collagenase (B204; Worthington, USA) in a cell culture incubator for 6–8 h. Isolated VSMCs were cultured in Dulbecco’s Modified Eagle Medium/F12 (DMEM/F12) supplemented with 10 % fetal bovine serum and 1 % penicillin-streptomycin, and their identity was confirmed by immunofluorescence staining for α-smooth muscle actin (α-SMA). Cells from passages 2–4 were utilized for subsequent experiments unless otherwise specified. Human aortic smooth muscle cells (HASMCs) were provided by ScienCell (Cat. No. 6110, USA). All cells were maintained at 37 °C in a humidified atmosphere of 5 % CO[2]. VSMCs at passages 2–4 and approximately 80 % confluence were used to establish in vitro models. For phenotypic switching, VSMCs were treated with 1 μM angiotensin II (AngII) for 24 h or 100 μM H[2]O[2] for 12 h. To induce cellular senescence, VSMCs were exposed to 100 μM H[2]O[2] for 12 h, followed by two washes with serum-free DMEM/F12 medium to remove residual H[2]O[2]. Cells were then cultured in fresh complete growth medium for an additional 72 h prior to analysis. 2.5. Histological and immunostaining analysis Aortic Sections and VSMC Crawls were used for immunofluorescent staining and immunohistochemical analyses. Double immunofluorescent staining of aortic tissue was performed to visualize protein expression and localization. Nuclei were visualized using 4′,6-diamidino-2-phenylindol (DAPI, [84]P36931; Invitrogen, USA) staining. Fluorescence intensity was calculated by Image-Pro Plus software (Media Cybernetics, Rockville, MA, USA). Immunohistochemical staining was performed to visualize protein expression in the indicated groups. Images were captured and analyzed using PANNORAMIC MIDI Scanner (3DHISTECH, Budapest, Hungary) with CaseViewer software (3DHISTECH). The relative positive area was analyzed using Image-Pro Plus software. Hematoxylin and eosin (H&E), Masson’s trichrome, and elastic Van Gieson (EVG) staining were used to visualize morphological characteristics, collagen content, and elastic fiber, respectively. Qualitative evaluation of elastin integrity was performed by semiquantitative grading as described [[85]30,[86]31]. The severity of elastin fragmentation was graded as follows: grade 1, no degradation; grade 2, mild elastin degradation; grade 3, severe elastin degradation; and grade 4, aortic rupture. 2.6. RNA sequencing Total RNA was extracted from VSMCs using TRIzol reagent (Invitrogen, USA) and converted into a cDNA library. RNA sequencing was subsequently performed through the high-throughput commercial sequencing service provided by Metware Co., Ltd. ([87]www.metware.cn). P-value adjustments were made using the Benjamini-Hochberg method (R package DESeq2) to control for the false discovery rate. Genes with an adjusted P-value <0.05 and |log2(fold change)| ≥ 0.5 were considered differentially expressed. Heatmaps and volcano plots were generated using the pheatmap and ggplot2 R packages, respectively. Gene Ontology (GO) pathway enrichment analysis was carried out using DAVID Tools ([88]https://david.ncifcrf.gov/tools.jsp). To explore the pathway and biological changes induced by CD147 overexpression or knockout, Gene Set Enrichment Analysis (GSEA) was performed using the complete gene expression dataset, and normalized enrichment score for each gene set was calculated. 2.7. Analysis of microarray datasets The [89]GSE233625 single-cell RNA sequencing dataset was reanalyzed using the R programming language. The raw data were downloaded from the Gene Expression Omnibus (GEO) repository. Initial data processing involved quality control to exclude low-quality cells and genes with minimal expression. Subsequently, the dataset was normalized to correct for technical variations and batch effects. Principal component analysis (PCA) was applied for dimensionality reduction to identify major sources of variation. Cells were clustered based on their gene expression profiles using unsupervised clustering methods. Differentially expressed genes (DEGs) between different clusters were identified and GO enrichment analyses were conducted to explore the biological functions associated with the DEGs. Data visualization was performed using R packages, including Seurat for clustering and ggplot2 for generating heatmaps, volcano plots, and other graphical representations. All statistical analyses were performed in R (version 4.3.2), ensuring reproducibility and consistency throughout the study. 2.8. Cleavage under targets and tagmentation–sequencing (CUT&Tag) and cleavage under targets & release using nuclease (CUT&Run) CUT&Tag and CUT&Run assays were performed using the Hyperactive Universal CUT&Tag Assay Kit for Illumina Pro (TD904, Vazyme Biotech) and Hyperactive pG-MNase CUT&Run Assay Kit for PCR/qPCR (HD101, Vazyme Biotech), respectively, following the manufacturer’s instructions. Briefly, cells were mixed with 10 μl Concanavalin A-coated magnetic beads and incubated at room temperature for 10 min. Bead-bound cells were resuspended in 100 μL of buffer containing primary antibody CD147 (1:50, CY6711, Abways, Shanghai, China) or IgG Isotype Control (1:50, CY5125, Abways). The mixture was incubated overnight at 4 °C. After removing the primary antibody buffer using a magnetic rack, cells were incubated with the secondary antibody goat anti-rabbit IgG antibody (1:100, Ab207, Vazyme) for 60 min at room temperature. The Hyperactive pA-Tn5 transposase (CUT&Tag) or pG-MNase Enzyme (CUT&Run) was prepared and incubated with cells for 1h at room temperature. Next, the cells were resuspended in the Tagmentation buffer and incubated at 37 °C for 1 h. DNA was purified using phenol-chloroform-isoamyl alcohol extraction and ethanol precipitation. The DNA product was used directly for qPCR in Cut&Run assay. For Cut&Tag, DNA library preparation and sequencing were performed by Romics Biotech (Shanghai, China). The libraries were amplified by PCR with a universal N5 and a uniquely barcoded N7 primer. Library size analysis was performed using the Agilent 4200 TapeStation system (Agilent Technologies, Santa Clara, CA, USA). Sequencing was carried out on the Illumina NovaSeq 6000 platform, generating 150 bp paired-end reads for subsequent analysis. Raw reads were processed using Trimmomatic to remove adapter sequences and low-quality bases. Clean reads were aligned to the rat reference genome (Rat6_ensembl_v112) using the BWA algorithm. MACS2 was used for peak calling (q-value <0.05), and peaks were annotated using the ChIPseeker R package. GO and KEGG enrichment analysis was conducted with clusterProfiler, and p-values were corrected using the false discovery rate (FDR). 2.9. Dual luciferase reporter assay Rat IRF7 promoter DNA (−2000 bp to +100 bp from the transcription start site) was synthesized by OBiO Technology (Shanghai, China) and cloned into the pGL3-Basic luciferase reporter vector. Rat VSMCs were co-transfected with the pGL3-IRF7-promoter firefly luciferase construct, a Renilla luciferase control vector, and either a CD147 expression plasmid or empty vector using Lipofectamine 3000 (Invitrogen). The pGL3-Basic vector (promoterless) and pGL3-SV40-promoter construct were used as negative and positive controls, respectively. Luciferase activity was measured using the Dual-Luciferase Reporter Assay System (Beyotime, Shanghai, China), and firefly signals were normalized to Renilla luciferase activity. 2.10. Alphafold3 To predict the potential structure of the rat CD147-STAT1-STAT2 complex, we utilized AlphaFold 3, a deep learning-based algorithm for protein structure prediction [[90]32]. The complex model was generated based on the known sequences of CD147, STAT1, and STAT2. After obtaining the predicted structure, visualization and further analysis were conducted using PyMOL software to examine the binding interactions and spatial configuration of the complex. 2.11. Molecular docking Autodock Vina (1.2.2) was employed to analyze the binding affinities and modes of interaction between Myricetin and CD147. The molecular structure of Myricetin was retrieved from PubChem Compound ([91]https://pubchem.ncbi.nlm.nih.gov/). The 3D coordinates of the monomer full-length CD147 were predicted by Alphafold3 [[92]32]. For docking analysis, all protein and molecular files were converted into PDBQT format with all water molecules excluded and polar hydrogen atoms were added. The grid box was centered to cover the domain of each protein and to accommodate free molecular movement. The grid box was set to 30 Å × 30 Å × 30 Å, and grid point distance was 0.05 nm. The docking scores were automatically generated by the software. The PyMOL software was used to analyze and visualize the binding patterns and hydrogen bonds. 2.12. RNA isolation and quantitative RT-PCR Total RNA was extracted using TRIzol reagent and subsequently used to synthesize cDNA with the HiScript ® III RT SuperMix for qPCR (Vazyme, China). mRNA levels were quantified through real-time PCR using ChamQ Universal SYBR qPCR Master Mix (Vazyme, China) on a LightCycler® 480 Real-Time PCR System (Roche Applied Science). Gapdh served as the internal reference genes for normalization. Detailed primer sequences are listed in [93]Table S1. 2.13. Subcellular fractionation analyses VSMCs were collected and washed three times with ice-cold PBS. Nuclear and cytoplasmic fractions were isolated using the Nuclear Extract Kit (SB-PR013HZ, Share-bio, Shanghai, China) according to the manufacturer’s protocol. The nuclear pellets underwent three additional washes with a specialized nuclear washing buffer to thoroughly eliminate any cytoplasmic and mitochondrial contamination. The purity of the nuclear extracts was verified via Western blot analysis using anti-GAPDH (cytoplasmic marker) and anti-H3 (nuclear marker) antibodies. 2.14. Immunoelectron microscopy To investigate the subcellular localization of CD147 in VSMCs, immunoelectron microscopy was performed on both freshly isolated rat aortic tissue and primary cultured rat VSMCs. Small tissue segments (∼1 mm^3) were immediately fixed in immunoelectron microscopy fixation buffer (Servicebio, Wuhan, China). Cultured VSMCs were gently scraped, pelleted, and fixed using the same protocol. Samples were dehydrated and embedded in pure resin using a low-temperature UV polymerization method at −20 °C for at least 48 h. Ultrathin sections were obtained using an ultramicrotome and collected onto nickel grids. After three washes with TBS, the sections were blocked with 1 % BSA for 30 min at room temperature and then incubated overnight at 4 °C with a rabbit anti-CD147 primary antibody (1:200, Abways, Shanghai, China). Following three additional washes, sections were incubated with a 1:50 dilution of gold-conjugated anti-rabbit IgG secondary antibody (Servicebio) at room temperature for 20 min and then at 37 °C for 1 h. After thorough washing with TBS and ultrapure water, the grids were counterstained with 2 % uranyl acetate in ethanol for 15 min in the dark, air-dried, and examined using a transmission electron microscope (JEM-1400, JEOL, Japan). 2.15. Assay of MMP activity In situ zymography was performed on cryosections (8–10 μm thick) of freshly harvested abdominal aortas to assess matrix metalloproteinase (MMP) activity. Briefly, tissue sections were incubated with a fluorogenic gelatin substrate (DQ™ gelatin, E−12055; Molecular Probes, USA) according to the manufacturer’s instructions. MMP activity was visualized as green fluorescence, and cell nuclei were counterstained with DAPI. Gelatin zymography was also conducted to evaluate MMP activity in VSMCs. Protein samples were separated by SDS-PAGE on gels containing 0.8 mg/mL gelatin. Following electrophoresis, gels were washed with 2.5 % Triton X-100 to remove SDS and then incubated in zymography buffer [50 mmol/L Tris-HCl (pH 8.0), 10 mmol/L CaCl[2], and 0.05 % Brij-35] at 37 °C for 48 h. After incubation, gels were stained with Coomassie Brilliant Blue to detect zones of gelatinolytic activity. 2.16. Western blotting analysis and co-immunoprecipitation Proteins were extracted from abdominal aortas, whole cells, and nuclear fractions. Equal amounts of total protein from murine tissues or VSMCs were resolved by 7.5 %–12.5 % SDS‒PAGE and transferred onto polyvinylidene fluoride membranes, blocked with 5 % bovine serum albumin, and incubated with the respective primary antibodies. Protein bands were identified using enhanced chemiluminescence (Millipore) after incubation with the corresponding secondary antibodies. The quantification of individual Western blot bands was performed with ImageJ software (National Institutes of Health, Bethesda, MD, USA) and the relative expression levels of the target proteins were expressed as a fold-change against GAPDH expression. After the total VSMC proteins were extracted and quantified, 500-1000 μg of protein was incubated with the indicated antibodies for co-immunoprecipitation assays. Following incubation at 4 °C overnight, the protein–antibody complexes were immunoprecipitated with 40 μL PureProteome™ Protein A/G Mix Magnetic Beads (LSKMAGAG02) for 10–30 min at room temperature with continuous mixing to capture the immune complex. Finally, the precipitated proteins were collected and separated by SDS-PAGE, and specific proteins were detected using western blotting. 2.17. Mass spectrometry analysis of CD147-interacted proteins Protein interactome analysis was conducted through the commercial services provided by Metware Co., Ltd. ([94]www.metware.cn). The samples underwent reductive alkylation, trypsin digestion, and peptide extraction. The resulting peptides were subsequently analyzed using a Vanquish Neo UHPLC system coupled with LC-MS/MS (Thermo Fisher Scientific, Waltham, MA). 2.18. RNA interference, plasmid construction, and adenovirus generation Small interfering RNAs (siRNAs) against rat STAT2 and a scrambled siRNA were designed and synthesized by OBiO Technology (Shanghai, China). Transfection of rat VSMCs with siRNAs (50 nM) in vitro was performed using Lipofectamine™ 3000 Reagent (L3000015; Thermo-Fisher, USA) according to the manufacturer’s protocol. To validate the predicted nuclear localization sequence (NLS) of CD147, we constructed two expression vectors. The full-length CD147 and a truncated version of CD147, where the predicted nuclear localization sequence (NLS, amino acids 350–353) was deleted, were both fused with FLAG and EGFP tags. These constructs were then cloned into the pcDNA3.1(+) vector. The plasmids were subsequently transfected into rat VSMCs using Lipofectamine™ 3000 Reagent, according to the manufacturer’s instructions. The transfected cells were cultured for further analysis to investigate the localization of CD147. To overexpress CD147, four recombinant replication-defective adenoviral vectors were constructed, each encoding FLAG-tagged rat CD147, FLAG-tagged mouse CD147, FLAG-tagged NLS-deleted mouse CD147, or a negative control, all under the control of the cytomegalovirus (CMV) promoter. The viruses were packaged and titrated by HanBio Technology (Shanghai, China). 2.19. CCK8 To assess the cytotoxicity of Myr, VSMCs were treated with Myr at concentrations of 5, 10, and 20 μM for 48 h. After the treatment period, cell viability was evaluated using the CCK-8 assay according to the manufacturer’s instructions. The absorbance was measured at 450 nm to determine the extent of cell proliferation and cytotoxicity induced by Myr treatment at the specified concentrations. 2.20. Statistical analysis All statistical analyses and graphing were conducted using GraphPad Prism 8 (GraphPad Software, San Diego, CA, USA). Data normality was assessed using the Shapiro–Wilk test. For comparisons between two groups, the Levene test was applied to detect homogeneity of variances. Comparisons were performed by the Student’s t-test (for equal variances) or Welch’s t-test (for unequal variances) for normally distributed variables or the Mann-Whitney U test for non-normally distributed variables. For comparisons among three or more groups, the Brown-Forsythe test was used to evaluate the homogeneity of variance, followed by a one-way ANOVA and Tukey’s post-hoc test when variances were equal, or Welch’s ANOVA with Tamhane’s T2 post-hoc test when variances were unequal. Two-way ANOVA with Tukey’s post-hoc analysis was used when more than two groups and variables were compared. The Chi-squared test was employed for bivariate comparisons of categorical variables. Ordinal variables were expressed as median (interquartile range, IQR), and statistical comparisons between groups were performed using the Mann–Whitney U test. Kaplan–Meier survival curves were generated, and survival differences were evaluated using the log-rank test. Unless stated otherwise, parametric data are expressed as mean ± SE, and non-parametric data are presented as median (IQR). A P-value of less than 0.05 was considered statistically significant. 3. Results 3.1. CD147 is highly expressed in the nucleus of VSMCs To investigate the role of CD147 in VSMCs, we first examined its expression and subcellular localization in primary rat aortic VSMCs. Immunofluorescence co-localization staining revealed that CD147 is obviously expressed in the nucleus of primary VSMCs, in addition to its cytoplasmic localization ([95]Fig. 1A). This nuclear distribution is distinct from its traditionally cytoplasmic and membrane expression observed in the breast cancer 4T1 cell line, rat cardiomyocytes (H9C2), and neonatal rat cardiomyocytes ([96]Supplementary Fig. 1A–1C), as reported in previous studies [[97]33,[98]34]. Cellular Immunoelectron microscopy and nuclear-cytoplasmic fractionation experiments further confirmed the obvious nuclear expression of CD147 in primary VSMCs ([99]Fig. 1B and C). We obtained further, more persuasive evidence for the nuclear localization of CD147 by performing nuclear-cytoplasmic fractionation on primary VSMCs overexpressing FLAG-CD147 via adenoviral transduction. Western blot analysis showed that exogenously expressed FLAG-CD147 was also highly abundant in the nucleus ([100]Fig. 1D). Furthermore, both immunofluorescence and immunoelectron microscopy analyses of rat aortic tissue confirmed significant nuclear expression of CD147 in VSMCs in vivo ([101]Fig. 1E and F). Notably, pharmacological stimulation of VSMCs with PDGF-BB or AngII induced a significant nuclear translocation of CD147 ([102]Fig. 1G-J), suggesting that its nuclear localization may play a crucial role in the cellular response to pathological stimuli. Similar results were observed in primary human and mouse aortic VSMCs with another two commercially available specific antibodies from different manufacturers (240134, Zenbio; ab188190, Abcam; [103]Supplementary Fig. 2A–2G). Fig. 1. [104]Fig. 1 [105]Open in a new tab CD147 is obviously localized in the nucleus of primary rat aortic VSMCs. (A) Representative immunofluorescence images showing CD147 expression and subcellular localization in rat VSMCs. The slides were co-stained with the SMC marker α-SMA. Nuclear localization of CD147 was determined by colocalization with DAPI. Different focal planes from Z-stack imaging are presented. Scale bar, 10 μm. (B) Representative immunogold electron microscopy images of CD147 protein expression in rat VSMCs. Scale bar, 200 nm. (C) Western blot analysis of nuclear and cytoplasmic protein fractions isolated from rat VSMCs. GAPDH was used as a cytoplasmic marker, and H3 (histone 3) was used as a nuclear marker. (D) Representative Western blot images of FLAG-tagged CD147 in nuclear and cytoplasmic protein fractions of rat VSMCs. (E) Representative immunofluorescence images of CD147 expression in rat aortic tissue. The slides were co-stained with α-SMA. CD147 nuclear localization was verified by colocalization with DAPI. Scale bar, 10 μm. (F) Representative immunogold electron microscopy images of CD147 protein expression in rat aorta tissue. Scale bar, 200 nm. (G) Left: Western blot analysis of CD147 protein levels in nuclear and cytoplasmic fractions of rat VSMCs treated with PBS or PDGF-BB (10 ng/mL). Right: Quantification of nuclear CD147 protein expression (n = 4). ∗∗∗P < 0.001. (H) Representative immunofluorescence images showing CD147 expression and subcellular localization in rat VSMCs treated with PBS or PDGF-BB (10 ng/mL). The slides were co-stained with the SMC marker α-SMA. Scale bar, 10 μm. (I) Left: Western blot analysis of CD147 protein levels in nuclear and cytoplasmic fractions of rat VSMCs treated with PBS or AngII (1 μM). Right: Quantification of nuclear CD147 protein expression (n = 3). ∗∗∗∗P < 0.0001. (J) Representative immunofluorescence images showing CD147 expression and subcellular localization in rat VSMCs treated with PBS or AngII (1 μM). The slides were co-stained with the SMC marker α-SMA. Scale bar, 10 μm. Recent studies have suggested that the nuclear translocation of certain proteins can be regulated by redox-dependent mechanisms [[106]35,[107]36]. To determine whether ROS mediate AngII- or PDGF-BB-induced nuclear localization of CD147, VSMCs were pretreated with the ROS scavenger N-acetyl cysteine (NAC) for 1 h prior to stimulation. NAC treatment markedly inhibited AngII- or PDGF-BB-induced nuclear accumulation of CD147 ([108]Supplementary Fig. 3A–3D), indicating that ROS may play a critical role in regulating CD147 nuclear expression. Furthermore, we identified a putative and highly conserved nuclear localization sequence (NLS) at positions 349 to 354 of rat CD147 employing three bioinformatics online tools, matching the classical K (K/R) X (K/R) NLS motif [[109]37] ([110]Supplementary Fig. 4A and 4B). To study the possible role of the NLS motif in regulating CD147 nuclear localization, VSMCs were transiently transfected with the pCD147-EGFP-FLAG or pCD147(NLS deletion)-EGFP-FLAG plasmid. Subsequent subcellular fractionation analysis showed that NLS deletion eliminated CD147 nuclear translocation ([111]Supplementary Fig. 4C), a finding that was also obtained by direct EGFP fluorescence ([112]Supplementary Fig. 4D). These results indicate that the nuclear localization of CD147 in VSMCs depends on the conserved NLS sequence ‘KRRK’. 3.2. CD147 binds to the promoter region of Irf7 and transcriptionally regulates its expression in VSMCs Considering the prominent nuclear expression of CD147 and its enhanced nuclear translocation in response to stressed stimuli, we hypothesize that CD147 may play a role in regulating the transcriptional functions of VSMCs. To investigate this further, we performed RNA sequencing of VSMCs transduced with either Vector control (Vector) or CD147-overexpressing (CD147-OE) adenovirus following PDGF-BB stimulation ([113]Supplementary Fig. 5A–C). Bioinformatics analysis revealed that DEGs were predominantly enriched in pathways related to the innate immune response and type I interferon (IFN–I) signaling ([114]Fig. 2A–B). To further assess whether CD147 interacts with specific DNA regions (such as promoters and enhancers) and regulates transcription, we performed a CUT&Tag assay using a recombinant monoclonal antibody against CD147 ([115]Supplementary Fig. 5D–E). Annotation of sequencing data showed that CD147 predominantly binds to transcriptionally relevant genomic regions ([116]Fig. 2C), such as promoters (31.37 %) and intergenic regions (32.7 %), further supporting the notion that CD147 may play a crucial role in transcriptional regulation. Subsequent KEGG functional enrichment analysis indicated that CD147 binding targets are primarily involved in regulating innate immune response-related pathways, including JAK-STAT, chemokine signaling, and Toll-like receptor pathways ([117]Fig. 2D). Fig. 2. [118]Fig. 2 [119]Open in a new tab CD147 binds to the Irf7 promoter region and regulates its transcription in VSMCs. (A) Top-ranked categories identified through Gene Ontology (GO) analysis of differentially expressed genes (DEGs) in CD147-overexpressing (CD147-OE) VSMCs compared to Vector controls treated with PDGF-BB (10 ng/mL). (B) Gene Set Enrichment Analysis (GSEA) of RNA-seq data highlighting the top-enriched gene sets based on normalized enrichment scores and p-values in CD147-OE VSMCs versus Vector controls. (C) Genome-wide distribution of CD147-binding sites in isolated VSMCs as determined by CUT&Tag analysis. (D) KEGG pathway enrichment analysis of target genes identified from CUT&Tag analysis. (E) Overlap of putative CD147 targets identified from CUT&Tag data with DEGs from RNA-seq analysis. Heatmap showing expression levels of overlapping genes across samples from the RNA-seq data. (F) IGV visualization of CUT&Tag analysis demonstrating CD147 enrichment at the Irf7 promoter. (G) CUT&Run was performed with a monoclonal anti-CD147 antibody, and the target promoter region of Irf7 was quantified by qPCR. (n = 5). ∗∗P < 0.01. (H) VSMCs were co-transfected with either CD147 overexpression plasmid or control vector along with the pGL3-IRF7-promoter luciferase construct for 48 h. Luciferase activity was measured using a dual-luciferase reporter assay system (n = 6). ∗∗∗P < 0.001. (I) Quantitative PCR analysis of mRNA expression levels of key genes in the type I interferon signaling pathway in VSMCs treated with PDGF-BB (10 ng/mL) in the indicated groups. ∗P < 0.05; ∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. (J) Western blot analyses showing significant upregulation of IRF7 protein levels in CD147-OE VSMCs compared to Vector group following PDGF-BB treatment (10 ng/mL, n = 6). ∗∗∗∗P < 0.0001. To explore the relationship between CD147 occupancy and mRNA expression, we compared the CUT&Tag and RNA sequencing datasets. A Venn diagram revealed 20 overlapping genes identified from both CUT&Tag peaks and DEGs of RNA-seq ([120]Fig. 2E). Among these potential targets, IRF7 ([121]Fig. 2F), a member of the interferon regulatory factors family, along with its associated IFN-I response pathway [[122]38], was selected for further investigation based on three considerations: (1) The RNA-seq data revealed a significant differential expression of IRF7, and Gene Set Enrichment Analysis (GSEA) suggested that CD147 overexpression significantly enhanced the IFN-I response in VSMCs ([123]Fig. 2B); (2) VSMCs are both IFN-I-responsive and IFN-I-productive cells. IFN-I response induces VSMC phenotypic switching and innate immune responses in an autocrine/paracrine manner [[124]6,[125]9,[126]39]; (3) IRF7-mediated IFN-I signaling, including the upstream cGAS-STING-TBK1 axis and downstream JAK-STAT pathway, plays a pivotal role in vascular smooth muscle pathological responses [[127]10,[128]40,[129]41]. The putative binding site of CD147 in the promoter region of the Irf7 locus was confirmed by Cut&Run followed by qPCR analysis ([130]Fig. 2G). Moreover, dual luciferase reporter assays showed that CD147 overexpression enhanced Irf7 promoter activity ([131]Fig. 2H; [132]Supplementary Fig. 5F). Consequently, CD147 overexpression significantly increased IRF7 mRNA and protein levels in PDGF-BB-treated VSMCs ([133]Fig. 2I and J). These results suggest that CD147 functions as a novel transcriptional regulator, involved in the transcriptional control of IRF7 in VSMCs. 3.3. CD147 promotes VSMC phenotypic transition, IFN-I response and cellular senescence under H[2]O[2] exposure PDGF-BB primarily drives the phenotypic transition of VSMCs from a contractile to a proliferative and osteogenic state, its influence on IFN-I responses is limited ([134]Supplementary Fig. 6A and 6B). In multiple vascular pathologies, excessive ROS leads to mitochondrial and nuclear DNA leakage into the cytoplasm, significantly activating the cGAS-STING and JAK-STAT pathways to promote IFN-I responses and the phenotypic shift of contractile VSMCs to an inflammatory state [[135]9,[136]42,[137]43]. Thus, we used H[2]O[2], a potent inducer of ROS and cellular dysfunction, to trigger IFN-I signaling in VSMCs ([138]Supplementary Fig. 6C and 6D). Excessive ROS stimulation induced by H[2]O[2] significantly upregulates CD147 protein expression and promotes its translocation to the nucleus in VSMCs ([139]Fig. 3A-C). As anticipated, CD147 overexpression significantly enhanced the expression of key components in the IFN-I signaling pathway, including IRF7, IFNα, and IFNβ, in response to H[2]O[2] ([140]Fig. 3D and E; [141]Supplementary Fig. 7A–7C). Previous studies documented that enhanced IFN-I response induces cellular premature senescence and VSMC phenotypic switching in an autocrine/paracrine manner [[142]9,[143]44,[144]45]. Thus, we further investigated the role of CD147 in H[2]O[2]-induced VSMC senescence. Results demonstrated that CD147 overexpression markedly promoted H[2]O[2]-induced VSMC senescence, as demonstrated by significantly enhanced expression of senescence markers (P21, P16) and activated senescence-associated secretory phenotype (SASP) (IL-6, CXCL1) ([145]Fig. 3D-E; [146]Supplementary Fig. 7A–7C). Similarly, the area of senescence-associated β-galactosidase (SA-β-gal) positive cells per field was significantly increased in H[2]O[2]-induced VSMCs with CD147 overexpression ([147]Fig. 3E). Additionally, H[2]O[2] induced a significant decrease in the expression of contractile phenotype-related proteins (CNN1, MYH11, α-SMA, and SM22), which were enhanced by CD147 overexpression ([148]Fig. 3D; [149]Supplementary Fig. 7A–7C). Fig. 3. [150]Fig. 3 [151]Open in a new tab CD147 overexpression promotes H[2]O[2]-induced VSMC phenotypic transition, IFN-I response, and cellular senescence. (A) Left: Western blot analysis of CD147 protein levels in VSMCs treated with PBS or H[2]O[2] (100 μM). Right: Quantification of CD147 protein expression (n = 6). ∗∗∗P < 0.001. (B) Top: Western blot analysis of CD147 protein levels in nuclear and cytoplasmic fractions of rat VSMCs treated with PBS or H[2]O[2] (100 μM). Bottom: Quantification of nuclear CD147 protein expression (n = 4). ∗∗P < 0.01. (C) Representative immunofluorescence images illustrating CD147 expression and subcellular localization in VSMCs treated with PBS or H[2]O[2] (100 μM). The slides were co-stained with SMC marker α-SMA and DAPI. (D) Western blot analysis of IRF7, IFNα, IFNβ, P21, α-SMA, SM22, CNN1 and MYH11 protein levels in VSMCs transfected with CD147-OE or Vector adenovirus, followed by exposure to PBS or H[2]O[2] (100 μM). (E) Left: Representative immunofluorescence images showing IRF7, P16, P21 protein expression, and SA-β-gal staining in VSMCs transfected with CD147-OE or Vector adenovirus, followed by exposure to PBS or H[2]O[2] (100 μM). The slides were co-stained with the SMC marker α-SMA and nuclear stain DAPI. Right: Quantification of IRF7, P16, P21 protein levels, and SA-β-gal positive areas in the indicated groups (n = 10–15). ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. To further explore the effects of CD147 inhibition, we generated VSMC-specific CD147 deletion mice (CD147^ΔSMC) by intercrossing Tagln-Cre transgenic mice on a C57BL/6J background with CD147-floxed mice (CD147^flox/flox) ([152]Supplementary Fig. 8A). Genotyping was performed and successful ablation of CD147 was demonstrated by CD147 detection via quantitative PCR assays and western blotting in isolated aortas ([153]Supplementary Fig. 8B–8E). Subsequently, mouse aortic VSMCs were isolated from CD147^ΔSMC and CD147^flox/flox mice and subjected to H[2]O[2] exposure. Remarkably, CD147 knockout effectively suppressed H[2]O[2]-induced IFN-I responses, cellular senescence, and VSMC phenotypic transition ([154]Supplementary Fig. 9A–9E). To investigate the role of nuclear CD147 in VSMC fate transition, we overexpressed FLAG-tagged wild-type CD147 (FLAG-CD147-WT) or FLAG-tagged NLS-deficient CD147 (FLAG-CD147-ΔNLS) in VSMCs isolated from CD147^ΔSMC mice via adenoviral vectors, followed by H[2]O[2] exposure. The results showed that overexpression of wild-type CD147 significantly enhanced H[2]O[2]-induced IFN-I responses, cellular senescence, and VSMC phenotypic transition. In contrast, these effects were significantly abolished in cells overexpressing NLS-deficient CD147 ([155]Supplementary Fig. 9F–9J). These data hinted that nuclear CD147 may transcriptionally activate the IRF7-IFNα/IFNβ signaling pathway, thereby promoting H[2]O[2]-induced phenotypic transition and senescence in VSMCs. 3.4. CD147 regulates the IRF7-IFNα/IFNβ pathway via binding to STAT1/STAT2 in VSMCs To further investigate the transcriptional regulatory mechanism of CD147 on IRF7, we performed immunoprecipitation using FLAG antibodies in lysates from senescent VSMCs overexpressing FLAG-CD147, followed by mass spectrometry to identify potential interacting proteins ([156]Fig. 4A). Among the identified proteins, the STAT1/STAT2 complex, a key transcription factor for IRF7 and essential regulator for IFN-I induction, was of particular interest ([157]Fig. 4B) [[158]46,[159]47]. Immunoprecipitation experiments confirmed that CD147 interacts with the STAT1/STAT2 complex ([160]Fig. 4C; [161]Supplementary Fig. 10A), and this interaction occurs within the nucleus ([162]Supplementary Fig. 10B). Phosphorylation of STATs is critical for their nuclear translocation and transcriptional activity. Therefore, we performed co-localization staining of CD147 and phosphorylated STAT2 (p-STAT2), which revealed that CD147 co-localizes with p-STAT2 in the nucleus of VSMCs ([163]Supplementary Fig. 10C). Using AlphaFold 3 [[164]32], we predicted the structural features between CD147 and the STAT1/STAT2 complex, revealing that CD147 primarily binds directly to STAT2 ([165]Fig. 4D). Knockdown of STAT2 using small interfering RNA significantly disrupted the interaction between CD147 and the STAT1/STAT2 complex ([166]Fig. 4E). Further phenotypic analyses demonstrated that STAT2 knockdown effectively reversed the promoting effects of CD147 overexpression on the IRF7-IFNα/IFNβ signaling pathway, cellular senescence, and phenotypic transition in VSMCs ([167]Fig. 4F and G; [168]Supplementary Fig. 10D–10F). These results suggest that CD147 may transcriptionally regulate IRF7-IFNα/IFNβ signaling and promote VSMC fate transition by forming a complex with STAT1/STAT2. Fig. 4. [169]Fig. 4 [170]Open in a new tab CD147 interacts with STAT1/STAT2 complex to activate IFN-I signaling in VSMCs. (A) Schematic workflow of the CD147 protein interactome analysis. Senescent VSMCs were infected with adenovirus expressing FLAG-CD147. Cell lysates were immunoprecipitated using anti-FLAG beads and subjected to mass spectrometry for protein identification. (B) Five unique peptides from the STAT1/STAT2 complex were identified (left), and a representative LC-MS/MS spectrum is shown (right). (C) Western blot analysis showing the association of FLAG-CD147 with the STAT1/STAT2 complex in VSMCs. FLAG-CD147 was immunoprecipitated with anti-FLAG antibody from VSMC lysates, followed by Western blotting. (D) Predicted structural model of the CD147/STAT1/STAT2 complex generated by AlphaFold3. (E) VSMCs were infected with adenovirus expressing FLAG-CD147 and either STAT2-targeting siRNA or a negative control (si-NC, scrambled siRNA). Lysates were immunoprecipitated with anti-FLAG antibody, followed by Western blot analysis. (F) Western blot analysis of STAT2, IRF7, IFNα, IFNβ, P21, α-SMA, SM22, CNN1 and MYH11 protein levels in VSMCs transfected with CD147-OE adenovirus and either STAT2-targeting siRNA or negative control (si-NC, scrambled siRNA), followed by exposure to PBS or H[2]O[2] (100 μM). (G) Left: Representative immunofluorescence images showing IRF7, P16, P21 expression, and SA-β-gal staining in VSMCs transfected with CD147-OE adenovirus and either STAT2-targeting siRNA or negative control, followed by exposure to PBS or H[2]O[2](100 μM). The slides were co-stained with SMC marker α-SMA and DAPI. Right: Quantification of IRF7, P16, P21 protein expression levels, and SA-β-gal positive areas in the indicated groups (n = 10). ∗∗∗∗P < 0.0001. 3.5. Aneurysmal stimuli markedly upregulate CD147 expression in VSMCs and significantly enhance its nuclear localization We next investigated the role of CD147-mediated VSMC phenotypic transition in aortic aneurysm (AA). We first analyzed single-cell RNA sequencing data of healthy aortic tissue (HC) and AngII-induced aneurysmal tissue (AA) obtained from the GEO database. Based on the expression patterns of cell type-specific marker genes, we identified four major vascular cell populations: VSMCs, fibroblasts, monocyte/macrophages, and endothelial cells (EC) ([171]Supplementary Fig. 11A and 11B). According to previous studies [[172]3] and gene ontology (GO) enrichment analysis of feature genes, VSMCs were classified into six principal functional subpopulations: contractile, extracellular matrix-producing, inflammatory, contractile/proliferative, stressed/adaptive, and EC function-associated SMCs ([173]Supplementary Fig. 11C–11E). Notably, cell distribution analysis revealed a significant expansion of inflammatory SMCs, alongside a marked reduction in proliferative and contractile SMCs in the aneurysm group compared to the control ([174]Fig. 5A). These findings suggest that aneurysmal stimuli induce VSMC phenotypic transition from a contractile state to inflammatory and senescent states. Furthermore, expression analysis revealed a pronounced upregulation of CD147 in VSMCs from the AngII-induced mouse aneurysmal tissues, while no significant changes were observed in endothelial cells or macrophages ([175]Fig. 5B). Moreover, the upregulation of CD147 in response to AngII stimulation was most evident in inflammatory SMCs ([176]Fig. 5B), reinforcing our in vitro findings that CD147 primarily drives VSMCs transition to the inflammatory phenotype. Further Western blot and immunofluorescence co-localization assays demonstrated that aneurysmal stimuli not only upregulated CD147 expression in VSMCs but also facilitated its nuclear translocation ([177]Fig. 5C and D). Collectively, these findings suggest that CD147-driven VSMC phenotypic transition towards an inflammatory and senescent state may play a pivotal role in the pathogenesis and progression of AAA. Fig. 5. [178]Fig. 5 [179]Open in a new tab VSMC-specific knockout of CD147 significantly inhibits AngII-induced AAA formation in an ApoE^−/− background mouse model. (A) Bar chart showing the percentage of cluster populations in aortic smooth muscle cells (SMCs) from HC and AA groups of mice. Fisher’s exact test was performed to examine differences in the cluster proportions between groups. (B) Violin plots depicting the expression levels of CD147 across different cell types in healthy control (HC) and aortic aneurysmal (AA) vascular tissues. Macro/Mono, macrophages/monocytes; EC, endothelial cells. (C) Western blot analysis indicating a significant upregulation of CD147 in mouse suprarenal abdominal aortic aneurysm tissue. ∗∗∗P < 0.001. (D) Immunofluorescence staining illustrating CD147 protein expression and nuclear localization in aneurysmal tissues of HC and AAA mice. The slides were co-stained with the smooth muscle cell marker α-SMA and DAPI. (E) Schematic protocol: ApoE^−/−/CD147^flox/flox and ApoE^−/−/CD147^ΔSMC mice were subcutaneously injected with saline or AngII by a mini osmotic pump for 28 days (n = 21–22 per group). (F) Survival curves of the AngII-infused ApoE^−/−/CD147^flox/flox and ApoE^−/−/CD147^ΔSMC mice. Data were analyzed using the Kaplan-Meier method and compared by log-rank tests. (G) Incidence of AngII-induced aneurysm rupture in the indicated groups (n = 21–22). (H) Representative macroscopic images of AAA formation in the indicated groups. (I) Incidence of AngII-induced AAA in the indicated groups (n = 21–22). Data were analyzed using Fisher’s exact test. ∗P < 0.01; ∗∗∗P < 0.001. (J) Quantification of total aortic weight/body weight (BW) ratio in the indicated groups (n = 7–10). ∗∗∗∗P < 0.0001. 3.6. VSMC-specific CD147 deficiency represses AAA formation In vivo To elucidate the potential role of CD147 in the pathology of AAA, we generated VSMC-specific CD147-knockout mice and their littermate controls in an ApoE knockout background (ApoE^−/−/CD147^ΔSMC and ApoE^−/−/CD147^flox/flox) ([180]Supplementary Fig. 12A and 12B), which were subjected to AngII or saline and maintained on a high-fat diet for 28 days ([181]Fig. 5E). Blood pressure measurements revealed no significant difference between ApoE^−/−/CD147^ΔSMC and ApoE^−/−/CD147^flox/flox control mice ([182]Table S3). However, ApoE^−/−/CD147^ΔSMC mice were less sensitive to AngII induction, as evidenced by significant reductions in mortality rate and aortic ruptures ([183]Fig. 5F and G). Moreover, compared with ApoE^−/−/CD147^flox/flox mice, decreased AAA incidence and the ratio of total aortic weight to body weight were observed in ApoE^−/−/CD147^ΔSMC mice under the condition of AngII infusion ([184]Fig. 5H-J). At the end of the induction period, micro-ultrasound imaging revealed that VSMC-specific CD147 knockout significantly attenuated the AngII-induced enlargement of maximal aortic diameter ([185]Supplementary Fig. 12C). Morphological and histological analyses further showed that CD147 deletion in VSMCs mitigated elastic fiber degradation and decreased collagen deposition in the aortas of AngII-infused mice ([186]Supplementary Fig. 12D). Western blot analysis revealed that in response to AngII stimulation, the expression of contractile phenotype markers in abdominal aortas was significantly upregulated in ApoE^−/−/CD147^ΔSMC mice compared to ApoE^−/−/CD147^flox/flox controls ([187]Fig. 6A and B). In addition, CD147 deficiency suppressed the expression of inflammatory cytokines, including interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), in the abdominal aorta ([188]Fig. 6C and D). Furthermore, CD147 deficiency markedly attenuated AngII-induced MMP activation, as evidenced by the significant reduction in MMP-2 expression in aortic tissue and MMP activity both in vivo and in cultured VSMCs ([189]Fig. 6C and D; [190]Fig. 6G and H). Immunostaining demonstrated lower levels of IFN-I components (IRF7, IFNβ) and senescence marker P21 in aneurysmal tissue, indicating improved cellular function following CD147 deletion ([191]Fig. 6E and F). In an AngII-induced in vitro VSMC model, we further confirmed that CD147 deficiency significantly suppressed IFN-I-related inflammatory signaling activation and cellular senescence, while promoting the transition toward a contractile VSMC phenotype through protein quantification ([192]Supplementary Fig. 13A–13D) and RNA-Seq ([193]Supplementary Fig. 13E–13I). Fig. 6. [194]Fig. 6 [195]Open in a new tab CD147 knockout significantly alleviated AngII-induced VSMC phenotypic transition, IFN-I response, and MMP activation. (A–B) Representative Western blot and quantification of STAT2, IRF7, α-SMA, SM22, CNN1, and MYH11 protein levels in abdominal aortas in the indicated groups of mice on day 28 following AngII infusion. ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. (C–D) Representative immunohistochemistry images and quantification of typical matrix-degrading enzyme MMP2 and inflammatory markers (IL-6, TNF-α) protein expression levels in aneurysmal tissues from the indicated groups of mice on day 28 following AngII infusion (n = 5). ∗P < 0.05. (E–F) Representative immunofluorescence images and quantification of fluorescence intensity for protein expression of IFN-I components (IRF7, IFNβ) and senescence marker (P21) in aneurysmal tissues from the indicated groups of mice on day 28 following AngII infusion. ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. (G) Representative images and quantification of active MMP in aneurysmal tissues from the indicated groups of mice on day 28 following AngII infusion (n = 6). ∗∗∗P < 0.001. (H) MMP activity measured by gelatin zymography in isolated VSMCs treated with AngII (1 μM). ∗∗P < 0.01. To further validate the pathogenic role of VSMC-derived CD147 in AAA development, we employed an alternative model of AAA induced by CaPO[4] ([196]Supplementary Fig. 14A). Compared with their CD147^flox/flox littermates, CD147^ΔSMC mice exhibited markedly attenuated aneurysm formation and a reduced maximal diameter of the infrarenal abdominal aorta ([197]Supplementary Fig. 14B and 14C). Histopathological analysis revealed significantly less elastic fiber degradation and collagen accumulation in CD147^ΔSMC mice compared with littermate CD147^flox/flox controls ([198]Supplementary Fig. 14D–14F). Collectively, these findings underscore the critical role of VSMC-derived CD147 in driving AAA pathogenesis and highlight its therapeutic potential. 3.7. CD147 natural small molecule inhibitor Myr ameliorates H[2]O[2]-induced VSMC phenotypic transition, IFN-I response and senescence We further assessed the effects of pharmacological inhibition of CD147 in vitro. Myricetin (Myr) is a natural small-molecule inhibitor with the highest affinity for CD147 discovered to date and possesses significant medicinal value [[199]48,[200]49]. We next investigated the impact of Myr on CD147 expression in VSMCs and its role in VSMC phenotypic transition and senescence. Molecular docking was conducted to evaluate the binding affinity and target binding sites between Myr and CD147. The crystal structure of monomer full-length CD147 was predicted by Alphafold3 and that of Myr were obtained from PubChem chemistry database, respectively. As shown in [201]Supplementary Fig. 15A and 15B, Myr bound to the N-terminal domain of CD147, forming five hydrogen bonds with amino acid residues GLU-53, TRP-56, LEU-70, ASP-72, and ALA-83. The docking score of −6.408 kcal/mol indicates a strong affinity between Myr and CD147. These data confirmed the stable interaction between Myr and CD147. Further Western blot analyses revealed that Myr downregulated CD147 protein expression in a dose-dependent manner in VSMCs ([202]Supplementary Fig. 15C–15E). Moreover, pre-treatment with the proteasome inhibitor MG-132 significantly reversed Myr-induced degradation of CD147 protein ([203]Supplementary Fig. 15F and 15G). These results suggest that Myr exhibits a strong binding affinity for CD147 and downregulates its protein levels through proteasome-dependent degradation. We next evaluated the cytotoxicity of Myr in VSMCs. CCK-8 assays demonstrated that even at a higher concentration (20 μM), Myr did not significantly inhibit VSMC cellular viability ([204]Supplementary Fig. 15H). We then investigated Myr’s potential protective effects on VSMCs under pathological stimuli. Results showed that Myr treatment significantly ameliorated the downregulation of contractile genes induced by H[2]O[2] ([205]Fig. 7A and B; [206]Supplementary Fig. 15I). Additionally, compared to the control group, Myr treatment markedly suppressed H[2]O[2]-induced upregulation of IRF7 and activation of the IFN-I signaling ([207]Fig. 7C and D; [208]Supplementary Fig. 15I). Quantitative analysis of senescence markers (P21 and P16) and SA-β-gal staining revealed that Myr significantly inhibited H[2]O[2]-induced cellular senescence ([209]Fig. 7E and F; [210]Supplementary Fig. 15I). These findings suggest that Myr treatment significantly attenuates H[2]O[2]-induced VSMC phenotypic transition, activation of the IFN-I signaling pathway, and cellular senescence. Fig. 7. [211]Fig. 7 [212]Open in a new tab Myricetin significantly attenuates H[2]O[2]-induced VSMC phenotypic transition, IFN-I activation, and senescence. (A–B) Western blot and quantification of α-SMA, SM22, CNN1, and MYH11 protein levels in VSMCs treated with myricetin (10 μM) or DMSO, followed by exposure to PBS or H[2]O[2] (100 μM) (n = 6). ∗∗P < 0.01; ∗∗∗∗P < 0.0001. (C–D) Western blot and quantification of IRF7, IFNα, IFNβ, and P21 protein levels in VSMCs treated with myricetin (10 μM) or DMSO, followed by exposure to PBS or H[2]O[2] (100 μM) (n = 6). ∗∗∗∗P < 0.0001. (E) Representative immunofluorescence images of IRF7, P16, and P21 expression, along with SA-β-gal staining, in VSMCs treated with myricetin (10 μM) or DMSO, followed by exposure to PBS or H[2]O[2] (100 μM). The slides were co-stained with the SMC marker α-SMA and nuclear stain DAPI. (F) Quantification of IRF7, P16, P21 fluorescence intensity, and SA-β-gal-positive areas in the indicated groups (n = 7–10). ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. 3.8. Myr treatment significantly inhibits AAA formation In vivo To explore the potential therapeutic effects of Myr in AAA, we first administered a gradient of Myr concentrations via daily intraperitoneal injections to mice for 7 consecutive days ([213]Supplementary Fig. 16A). Results showed that a dose of 10 mg/kg effectively inhibited CD147 protein expression in vascular tissues without causing any noticeable toxicity ([214]Supplementary Fig. 16B). Subsequently, we performed AngII infusion in ApoE^−/− mice and administered 10 mg/kg/day of Myr via intraperitoneal injection for 28 days ([215]Fig. 8A). Compared to the vehicle-treated group, Myr treatment improved the survival rate of aneurysmal mice ([216]Fig. 8B), and reduced AngII-induced aortic rupture, AAA formation, and the ratio of total aortic weight to body weight in ApoE^−/− mice ([217]Fig. 8C-F). Ultrasound analysis confirmed that the maximal aortic diameter was significantly lower in Myr-treated mice than that in Vehicle-treated littermates infused with AngII ([218]Fig. 8G). Morphologically, histological analysis revealed that Myr treatment obviously mitigated elastin degradation and pathological collagen deposition in AngII-administered mouse aortas ([219]Fig. 8G). Fig. 8. [220]Fig. 8 [221]Open in a new tab Administration of the CD147 natural inhibitor myricetin protects against AAA formation. (A) Schematic protocol: ApoE^−/− mice were infused with AngII or saline and intraperitoneally injected with vehicle or Myricetin (10 mg/kg/day) for 28 days. (B) Survival curves of the vehicle- or Myr-treated ApoE^−/− mice infused with AngII (n = 18–20). Data were analyzed using the Kaplan-Meier method and compared by log-rank tests. (C) Incidence of AngII-induced aneurysm rupture in the indicated groups (n = 18–20). (D) Representative macroscopic images of AAA formation in the indicated groups. (E) Incidence of AngII-induced AAA in the indicated groups (n = 18–20). Data were analyzed using Fisher’s exact test. ∗P < 0.05; ∗∗∗P < 0.001. (F) Quantification of total aortic weight/body weight (BW) ratio in the indicated groups (n = 6–9). ∗∗P < 0.01; ∗∗∗∗P < 0.0001. (G) Left: Representative ultrasound and histopathological images of abdominal aortas in mice after 28 days of saline or AngII infusion. Right: Quantification of the maximal diameter of abdominal aortas measured by B-mode ultrasound imaging (n = 6–11), elastin degradation, and collagen content based on histopathological staining (n = 6–7). ∗P < 0.05; ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. Compared with the vehicle group, Myr treatment significantly upregulated the expression of VSMC contractile phenotype markers in abdominal aortas upon AngII stimulation ([222]Fig. 9A and B). Consistently, Myr administration markedly reduced the protein levels of pro-inflammatory cytokines (TNF-α and IL-6) in the vascular wall and suppressed the expression of type I interferons (IFN-α and IFN-β) as well as the senescence marker P21 in aneurysmal tissues ([223]Fig. 9C-F). Furthermore, Myr treatment robustly attenuated AngII-induced MMP activation both in vivo and in cultured VSMCs ([224]Fig. 9G-H). Fig. 9. [225]Fig. 9 [226]Open in a new tab Administration of the CD147 natural inhibitor Myricetin attenuates AngII-induced VSMC phenotypic transition, IFN-I response, and MMP activation. (A–B) Western blot and quantification of STAT2, IRF7, α-SMA, SM22, CNN1, and MYH11 protein levels in abdominal aortas in the indicated groups of mice on day 28 following AngII infusion. ∗∗P < 0.01; ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. (C–D) Representative immunohistochemistry images and quantification of typical matrix-degrading enzyme MMP2 and inflammatory markers (IL-6, TNF-α) protein expression levels in aneurysmal tissues from the indicated groups of mice on day 28 following AngII infusion (n = 5). ∗P < 0.05; ∗∗∗P < 0.001. (E–F) Representative immunofluorescence images and quantification of fluorescence intensity for protein expression of IFN-I components (IFNα, IFNβ) and senescence marker (P21) in aneurysmal tissues from the indicated groups of mice on day 28 following AngII infusion (n = 5). ∗∗∗P < 0.001; ∗∗∗∗P < 0.0001. (G) Representative images and quantification of active MMP in aneurysmal tissues from the indicated groups of mice on day 28 following AngII infusion (n = 6). ∗P < 0.05. (H) MMP activity measured by gelatin zymography in isolated VSMCs treated with AngII (1 μM). ∗∗∗∗P < 0.0001. In the CaPO[4]-induced AAA model ([227]Supplementary Fig. 16C), Myr treated mice exhibited markedly attenuated aneurysm formation, reduced maximal diameter, less elastic fiber degradation and collagen accumulation of the infrarenal abdominal aorta compared with Vehicle group, ([228]Supplementary Fig. 16D–16H). These data indicate that Myr significantly mitigates the occurrence and progression of AngII- and CaPO[4]-induced AAA in vivo, highlighting its potential clinical translational value for the treatment of AAA. 4. Discussion This study identified a previously unrecognized role of VSMC-derived CD147 in the pathogenesis of AAA and VSMC phenotypic transition, providing several novel insights. First, CD147 was highly expressed in the nucleus of VSMCs and exhibited remarkable nuclear translocation upon pathological stimuli in a ROS-dependent manner, where it transcriptionally activated the IFNα/β signaling pathway through interaction with the STAT1/STAT2 transcriptional complex. This ROS-activated CD147-IFN-I axis promoted the phenotypic transition of healthy contractile VSMCs into pro-inflammatory and senescent states. Second, VSMC-specific knockout of CD147 protected mice against AAA formation in both AngII- and CaPO[4]-induced AAA models. Third, supplementation with myricetin, a natural CD147 inhibitor abundantly found in flavonoid-rich foods, effectively inhibited H[2]O[2]-induced VSMC phenotypic transition in vitro and AngII/CaPO[4]-induced AAA formation in vivo. Collectively, our study provides the first evidence identifying a novel ROS-activated CD147–STAT1/2–type I interferon signaling axis, which plays a pivotal role in driving VSMC fate transition and promoting AAA pathogenesis. Protein subcellular localization is dynamic, cell-specific, and functionally diverse, often influenced by tissue and cellular microenvironments to meet diverse biological demands [[229]50,[230]51]. For instance, FGFR3, a transmembrane glycoprotein typically localized to the plasma membrane in normal cells, is aberrantly expressed in the nucleus of pancreatic cancer cells, where its nuclear presence has been associated with poor prognosis [[231]52]. Beyond its pyruvate kinase activity in the cytoplasm, PKM2 also translocates to the nucleus in response to various signals, functioning as a transcriptional regulator by acting as a protein kinase [[232]53,[233]54]. Similarly, as a highly glycosylated protein, CD147 has been reported predominantly localized to the cytoplasm and plasma membrane in most mammalian cells [[234]55]. In this study, we provide the evidence that CD147 is highly expressed in the nucleus of VSMCs in an NLS ‘KRRK’-dependent manner. Interestingly, earlier study suggested that in hepatocellular carcinoma cells, a C-terminus domain of CD147 could be cleaved by γ-secretase and translocated into the nucleus [[235]56]. However, our data suggest that nuclear CD147 in VSMCs likely exhibits as the full-length protein, as its molecular weight is comparable between nuclear and cytoplasmic fractions. Moreover, external stimuli, such as PDGF-BB, H[2]O[2], and AngII, significantly enhance the nuclear expression of CD147 as a result of elevated ROS levels, suggesting a potential regulatory role in cellular redox processes. This finding expands our understanding of CD147’s cellular dynamics, and elucidating the mechanisms driving CD147 nuclear translocation could provide the foundation for targeting its nuclear-specific functions as a strategy to mitigate VSMC phenotypic switching and AAA progression. Proper subcellular localization is fundamental to the functionality of biomacromolecules, including proteins and RNAs. CD147 is predominantly localized to the cytoplasm and plasma membrane in most mammalian cells, where it mediates cell-cell interactions [[236]13], lactate transport [[237]11], and signal transduction [[238]57]. In contrast, proteins highly expressed in the nucleus are typically involved in transcriptional regulation, DNA replication and repair, chromatin remodeling. In this study, we utilized multi-omics analyses (RNA-seq, CUT&Tag, and protein interactome) to uncover a novel signaling pathway in which nuclear CD147 interacts with the STAT1/2 complex to transcriptionally activate the IRF7-IFNα/β signaling axis under H[2]O[2]/AngII exposure in VSMCs. The IFN-I pathway was initially identified for its critical antiviral role; however, excessive IFN-I production has been implicated in autoinflammatory diseases [[239]58]. In recent years, this pathway has attracted increasing attention for its involvement in a broad range of pathological conditions, including cardiovascular and oncological diseases [[240]59,[241]60]. For example, cardiomyocytes, rather than immune cells, have been identified as primary initiators of the pathological IFN-I response in the infarct border zone following myocardial infarction, positioning IFN-I as a potential therapeutic target in ischemic cardiomyopathy [[242]61,[243]62]. VSMCs not only express and secrete IFN-I in response to injury stimuli but also act as IFN-I-responsive cells. Through autocrine and paracrine signaling, IFN-I promotes VSMC phenotypic switching and senescence [[244]6,[245]9,[246]63]. Similarly, our study demonstrated that ROS-activated CD147-type I interferon signaling axis orchestrates the phenotypic transition of VSMCs toward a pro-inflammatory and senescent state, and VSMC-specific knockout of CD147 ameliorates AngII-induced AAA development and suppresses pathological VSMC fate transition. Although we did not directly manipulate nuclear CD147 in vivo, our in vitro rescue experiments, by overexpressing either CD147-WT or CD147-ΔNLS in CD147-deficient VSMCs, demonstrated that the pro-phenotypic switching and IFN-I signaling activation effects of CD147 largely depend on its nuclear localization sequence. These findings suggest that ROS-induced nuclear translocation of CD147 likely serves as the key mechanism by which CD147 promotes AAA pathogenesis. This discovery not only highlights CD147 as a versatile regulator with expanded functional repertoire, but also positions it as a mechanistic link between ROS and type I interferon signaling in VSMC, thereby bridging the redox-regulated immune-metabolic control of VSMC plasticity and AAA pathogenesis. Moreover, it offers a novel therapeutic avenue to inhibit VSMC plasticity and attenuate aneurysmal development. As a promising therapeutic target, CD147 is considered as a key driver in various diseases, including cancer [[247]11,[248]64], cardiovascular diseases [[249]16], and infectious diseases [[250]15,[251]65]. Despite the initial success of monoclonal antibodies and chemically synthesized small molecule inhibitors targeting CD147, their widespread clinical application remains limited due to concerns over biosafety, biocompatibility, high costs and low affinity [[252][66], [253][67], [254][68], [255][69]]. Natural products, however, offer advantages such as high bioavailability, low toxicity, and diverse pharmacological activities, making them an important resource for the development of targeted therapies for human diseases [[256]70]. Myricetin, a natural flavonoid abundant in various plants or foods and available in dietary supplement form, is currently the small molecule with the highest known binding affinity for CD147, and has been considered to possess considerable medicinal value [[257]49,[258]71]. It exhibits diverse pharmacological effects, including antioxidant, anti-inflammatory, antimicrobial, anticancer, antidiabetic, and hepatoprotective properties [[259]48]. Healthcare products containing myricetin have been developed and marketed for their antioxidant and anti-inflammation effects, although it is not yet approved as a new drug [[260]48]. Accumulating evidence suggests a strong association between oxidative stress and both VSMC phenotypic modulation and AAA development, highlighting antioxidant intervention as a promising AAA therapeutic approach [[261]7,[262]72]. For example, dietary intake of antioxidant vitamins or fruits rich in antioxidative compounds has been shown to confer protective effects against AAA [[263]73,[264]74]. Consistently, our data show that antioxidant myricetin effectively inhibits oxidative stress-induced VSMC phenotypic transition and cellular senescence in vitro, as well as AngII- and CaPO[4]-induced AAA formation and progression in vivo, at least in part by targeting CD147-type I interferon signaling. These findings provide new mechanistic insights into antioxidant-based strategies for AAA prevention and treatment. Several limitations of this study should be acknowledged. First, although ROS may serve as a key initiator, the molecular mechanisms underlying CD147 nuclear translocation in VSMCs remain unclear. Further investigation into this process may provide novel opportunities to directly target nuclear CD147. Second, while our findings suggest myricetin as a potential therapeutic agent for AAA, additional clinical research is needed to confirm the efficacy and clinical benefits of myricetin in AAA treatment. Third, although our study confirmed the detrimental role of VSMC-derived CD147 in AAA development using two distinct murine models—subcutaneous AngII infusion and adventitial CaPO[4] application—both of which have inherent limitations in recapitulating the complex pathogenesis of human AAA. Moreover, although Tagln-Cre is widely used for VSMC-specific gene manipulation, its cellular specificity remains a matter of debate [[265]75]. Given that both our study and previous reports support CD147 as a therapeutically targetable molecule in AAA [[266]24], the potential contributions of CD147 derived from non-VSMC populations warrant further investigation. 5. Conclusion In summary, our study unveils a previously undescribed ROS-activated CD147-STAT1/2-IFNα/IFNβ axis in promoting VSMC inflammatory phenotype and senescence, and provides proof-of-concept evidence indicating the therapeutic value of targeting CD147 inhibition in AAA prevention and treatment. CRediT authorship contribution statement Fangyuan Zhong: Writing – review & editing, Writing – original draft, Software, Methodology, Investigation, Data curation, Conceptualization. Hengyuan Zhang: Writing – original draft, Software, Methodology, Investigation. Xinning Guo: Writing – original draft, Visualization, Software, Methodology, Investigation. Yichao Zhao: Writing – review & editing, Conceptualization. Yufei Wang: Methodology, Investigation. Wenli Li: Methodology, Investigation. Yuyan Lyu: Writing – review & editing, Resources, Conceptualization. Heng Ge: Writing – review & editing, Visualization, Supervision, Resources, Funding acquisition, Conceptualization. Xiyuan Lu: Writing – review & editing, Writing – original draft, Validation, Supervision, Conceptualization. Jun Pu: Writing – review & editing, Writing – original draft, Supervision, Resources, Funding acquisition, Conceptualization. Declaration of competing interest There are no conflicts of interest to declare. Acknowledgments