Abstract Beyond dyslipidemia, inflammation contributes to the development of atherosclerosis. However, intrinsic factors that counteract vascular inflammation and atherosclerosis remain scarce. Here we identify insulin-like growth factor binding protein 6 (IGFBP6) as a homeostasis-associated molecule that restrains endothelial inflammation and atherosclerosis. IGFBP6 levels are significantly reduced in human atherosclerotic arteries and patient serum. Reduction of IGFBP6 in human endothelial cells by siRNA increases inflammatory molecule expression and monocyte adhesion. Conversely, pro-inflammatory effects mediated by disturbed flow (DF) and tumor necrosis factor (TNF) are reversed by IGFBP6 overexpression. Mechanistic investigations further reveal that IGFBP6 executes anti-inflammatory effects directly through the major vault protein (MVP)–c-Jun N-terminal kinase (JNK)/nuclear factor kappa B (NF-κB) signaling axis. Finally, IGFBP6-deficient mice show aggravated diet- and DF-induced atherosclerosis, whereas endothelial-cell-specific IGFBP6-overexpressing mice protect against atherosclerosis. Based on these findings, we propose that reduction of endothelial IGFBP6 is a predisposing factor in vascular inflammation and atherosclerosis, which can be therapeutically targeted. __________________________________________________________________ Atherosclerosis is the hallmark pathology of coronary artery disease (CAD) and has long been a significant cause of death globally^[73]1. Atherosclerotic plaques typically develop in areas of disturbed flow (DF), such as arterial branches or vascular bends. DF alters the morphology and cytoskeleton of endothelial cells (ECs) through altered biochemical signaling and gene expression, ultimately leading to endothelial dysfunction. Conversely, unidirectional laminar flow (UF) usually occurs in the straight part of the blood vessel, and vessels exposed to this area are protected against atherosclerosis^[74]2–[75]9. DF instigates endothelial dysfunction partially through inducing inflammation in ECs^[76]10. Inflammation leads to endothelial injury, which, in turn, promotes the expression of a variety of adhesion molecules, fueling the adhesion and migration of circulating leukocytes to the activated endothelium and ultimately leading to plaque formation^[77]11,[78]12. Therefore, targeting inflammation has been recognized as a necessary means to combat atherosclerosis^[79]13. More recently, in a retrospective analysis of 31,245 patients receiving statin therapy, residual inflammation risk as assessed by high-sensitivity C-reactive protein (hs-CRP) was a stronger predictor of risk for future cardiovascular events and death than residual cholesterol risk as assessed by low-density lipoprotein cholesterol (LDL-C)^[80]14,[81]15. In addition, low-dose colchicine reduces the incidence of major adverse cardiovascular events (MACE) in patients with cardiovascular disease^[82]16. Likewise, monoclonal antibodies targeting the inflammatory cytokines interleukin (IL)-6 and IL-1β (by ziltivekimab and canakinumab, respectively) have shown promising anti-atherosclerotic effects in clinical trials^[83]17,[84]18. Therefore, the identification of novel anti-inflammatory therapies holds promise for reducing residual inflammation and cardiovascular risk. To identify novel anti-inflammatory targets in ECs, we selected RNA sequencing (RNA-seq) datasets from human umbilical vein endothelial cells (HUVECs) exposed to atherosclerotic protective therapy (statins and UF) as well as expression datasets from patients with CAD with unstable plaques. We discovered that insulin-like growth factor binding protein 6 (IGFBP6) may serve as a potential vascular homeostasis-associated molecule. Previous studies demonstrated that IGFBP6 plays a crucial role in regulating the cell cycle, cell proliferation and migration^[85]19–[86]21. Also, compared to stable plaques from patients with CAD, the expression level of IGFBP6 in unstable plaques was decreased^[87]22. However, the precise role and mechanism of IGFBP6 in endothelial homeostasis and atherosclerosis remain unclear. Here we demonstrate that global or EC-specific deficiency of Igfbp6 accelerates the progression of atherosclerosis in mice, whereas EC-specific overexpression of Igfbp6 ameliorates atherosclerosis. Mechanistically, IGFBP6 inhibits endothelial cell inflammation and monocyte adhesion through the major vault protein (MVP)–c-Jun N-terminal kinase (JNK)/nuclear factor kappa B (NF-κB) pathway. Of translational impact, the expression of IGFBP6 is decreased in serum and atherosclerotic lesions of patients with CAD. Overall, these data suggest that IGFBP6 is mechanoresponsive, anti-inflammatory and atheroprotective. Results IGFBP6 is decreased in human atherosclerotic plaques and regulated by blood flow To identify novel regulators of endothelial homeostasis, we treated HUVECs with atorvastatin for transcriptomic sequencing ([88]GSE176531) and selected differentially expressed genes (DEGs) with P < 0.05 and log[2]fold change (FC) > 1.5. In addition, DEGs in sequencing datasets from UF-treated HUVECs ([89]GSE20739 and [90]GSE87534) and CAD patient samples ([91]GSE163154 and [92]GSE41571) were compared. The combined analysis of five datasets yielded a total of three genes: ADAM metallopeptidase with thrombospondin type 1 motif 1 (ADAMTS1), cellular communication network factor 3 (CCN3, also known as NOV) and IGFBP6. ADAMTS1 and NOV were reported to be involved in endothelial inflammation and atherosclerosis^[93]23,[94]24. However, the role and mechanism of IGFBP6 in endothelial homeostasis and atherosclerosis remain obscure. Thus, in the present study, we focused on the role of IGFBP6 as a potential regulator of endothelial homeostasis ([95]Fig. 1a). Fig. 1 |. IGFBP6 is decreased in human atherosclerotic plaques and regulated by blood flow. Fig. 1 | [96]Open in a new tab a, Identification of IGFBP6 as a novel endothelial homeostasis-associated molecule. Statin-upregulated genes ([97]GSE176531) and UF-upregulated genes ([98]GSE20739 and [99]GSE87534) were overlapped with downregulated genes in unstable atherosclerotic plaques ([100]GSE163154 and [101]GSE41571). b, The expression of IGFBP6 in macroscopically intact tissues versus atheroma plaques, stable plaques versus unstable plaques in public datasets from patients ([102]GSE43292 (n = 32) or [103]GSE163154 (stable plaques: n = 16; unstable plaques: n = 27)). c, Expression of IGFBP6 in tissues and organs of patients with CAD (n = 600) and healthy individuals (n = 250) in the STARNET database. AOR, atherosclerotic aortic wall; LIV, liver; SF, subcutaneous fat; SKLM, skeletal muscle; VAF, visceral fat. d, Representative images of immunofluorescence staining of IGFBP6 (red), VE-cadherin (green) and DAPI (blue). The expression of IGFBP6 was downregulated in the endothelium of human aortic atherosclerotic lesions compared to normal controls (n = 3). Scale bar, 25 μm. e, Serum samples were collected from normal controls and patients with CAD to verify the IGFBP6 content by ELISA (control: n = 12; CAD: n = 19). f, IGFBP6 mRNA expression was downregulated comparing DF to UF in the public microarray in HUVECs (n = 3, [104]GSE20739). g, HUVECs were subjected to DF or UF for 24 h. KLF2, KLF4 and IGFBP6 mRNA expression were downregulated in DF-treated HUVECs (n = 3). h, Schematic representation of a mouse model with PCL. i, Igfbp6 was decreased in the intimal RNA of mouse carotid arteries 4 d after PCL surgery (n = 5). j, Immunofluorescence staining for IGFBP6 (red) and DAPI (blue) showed that IGFBP6 was reduced in the carotid arteries of mice at 4 weeks after PCL surgery (n = 5). Green: auto-fluorescence of the elastic lamina (EL). Scale bar, 25 μm. k, Immunoblotting showing that the IGFBP6 was lower in the inner curvature of the AA than in the TA in C57BL/6J mouse aortas (n = 6). l, En face immunofluorescence staining for IGFBP6 (red), VE-cadherin (green) and DAPI (blue) in C57BL/6J mouse aortas, showing decreased IGFBP6 expression in the AA compared to the TA (n = 10). Scale bar, 50 μm. Data are shown as mean ± s.d. Statistical analysis was performed by two-tailed Student’s t-test for b, e, k and l, by Welch’s t-test for c and f and by paired t-test for g and i. By mining two additional and independent published datasets ([105]GSE43292 and [106]GSE163154), we observed that IGFBP6 mRNA expression was reduced in carotid artery plaques compared to the adjacent non-plaque tissues. Additionally, unstable plaques had lower levels of IGFBP6 mRNA than stable plaques ([107]Fig. 1b). We subsequently validated IGFBP6 expression in tissues and organs of patients with CAD (n = 600) and healthy individuals (n = 250) from the Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task (STARNET) database^[108]25–[109]27. The results showed that IGFBP6 expression was significantly lower in atherosclerotic aortic tissues of patients with CAD compared to healthy controls ([110]Fig. 1c). To extend the translational significance of our study in human patients, we analyzed the arterial expression of IGFBP6 in control subjects and patients with CAD. In comparison to healthy coronary arteries, the expression level of IGFBP6 in the intima of human coronary arteries with atherosclerotic plaques was decreased ([111]Fig. 1d). Furthermore, the level of IGFBP6 was reduced in the serum of patients with atherosclerotic plaques compared to that in healthy individuals ([112]Fig. 1e). Because the expression of IGFBP6 is upregulated by UF, we next explored the potential role of IGFBP6 in responding to different types of shear stress. We first examined its expression in HUVECs by mining microarray data of ECs treated with DF and UF ([113]GSE20739). The mRNA expression of IGFBP6 was lower in HUVECs treated with DF compared to those treated with UF ([114]Fig. 1f). We also validated the downregulation of IGFBP6 mRNA expression in HUVECs exposed to DF compared to UF using quantitative real-time polymerase chain reaction (qRT–PCR) ([115]Fig. 1g). Because the IGFBP family has seven members, we next examined the expression of IGFs and different IGFBPs in UF-treated and DF-treated HUVECs. Our results showed significant increases in IGFBP5 and IGFBP6 ([116]GSE20739) ([117]Extended Data Fig. 1a,[118]b). We also measured the mRNA levels of IGFBPs in HUVECs and found that IGFBP6 had a higher expression level than other IGFBPs ([119]Extended Data Fig. 1c). Because our previous study reported that IGFBP5 is mechanoresponsive and anti-inflammatory^[120]28, in the present study our goal was to explore the potential role of IGFBP6 in regulating endothelial function and atherosclerosis. To verify blood flow-dependent regulation of IGFBP6 expression in vivo, we obtained intimal RNA from carotid arteries of mice after partial carotid ligation (PCL) surgery for 4 d ([121]Fig. 1h). qRT–PCR analysis showed that the expression of Igfbp6 mRNA in the endothelium of the left carotid artery (LCA; exposed to DF) was decreased compared to LCA from sham group after 4 days of PCL ([122]Fig. 1i). Consistently, immunofluorescence staining demonstrated a reduction in IGFBP6 protein expression after PCL ([123]Fig. 1j). IGFBP6 protein expression was also quantified in different regions of the aorta, specifically the inner curvature of aortic arch (AA; region of DF) and thoracic aorta (TA; region of UF). We found that IGFBP6 protein expression was lower in the AA than in the TA ([124]Fig. 1k). En face staining of the mouse aortic endothelium also showed lower expression of IGFBP6 in the AA than in the TA ([125]Fig. 1l). Collectively, these results suggest a potential role of IGFBP6 as a flow-responsive gene that may execute important biological functions in vascular homeostasis and atheroprotection. IGFBP6 deficiency increases atherosclerotic plaque formation in mice receiving PCL surgery To validate whether IGFBP6 suppresses atherosclerosis in vivo, we first constructed Igfbp6 global knockout mice (Igfbp6^−/−) by deleting the exon 2 of Igfbp6. We confirmed Igfbp6 knockout by genotyping ([126]Extended Data Fig. 2a) and qRT–PCR. Decreased Igfbp6 mRNA expression was observed in tested mouse tissues and organs (adipose tissue, liver, heart, lung, colon and aorta) of Igfbp6^−/− mice compared to wild-type (WT) mice ([127]Fig. 2a). Male 8-week-old Igfbp6^−/− mice and their WT littermate controls were subsequently infected with AAV8-Pcsk9^D377Y (to reduce LDLR levels in mouse livers and induce atherosclerosis; [128]Extended Data Fig. 2b) via tail vein injection concurrent with feeding with a high-cholesterol diet (HCD) for 2 weeks. Afterwards, we performed PCL surgery on the mice and continued HCD feeding for 3 weeks ([129]Fig. 2b). Fig. 2 |. IGFBP6 deficiency increases atherosclerosis in mice receiving PCL surgery. Fig. 2 | [130]Open in a new tab a, Igfbp6 transcript level was successfully deleted in various tissues and organs in Igfbp6^−/− mice (n = 3). b, Schematic diagram illustrating the experimental scheme. c, Mouse serum was used to verify IGFBP6 deletion by ELISA in Igfbp6^−/− mice (n = 4). d, Immunofluorescence staining for IGFBP6 (red) and DAPI (blue) showed that IGFBP6 was reduced in the RCAs of Igfbp6^−/− mice (n = 3). EL denotes elastic lamella that display auto-fluorescence (green). Scale bar, 25 μm. e, Increased plaque formation in male Igfbp6^−/− mice receiving PCL (WT: n = 15; Igfbp6^−/−: n = 13). Scale bar, 2 mm. f, Representative Oil Red O–stained carotid arteries of male WT and Igfbp6^−/− mice. The panel on the right shows the quantification of the Oil Red O–positive plaque area (WT: n = 15; Igfbp6^−/−: n = 10). Scale bars, 50 μm. g, Immunofluorescence staining for VCAM-1 (green) and DAPI (blue) showed that VCAM-1 was increased in carotid cryosections from Igfbp6^−/− mice (n = 6). Scale bars, 100 μm and 25 μm. Data are shown as mean ± s.d. Statistical analysis was performed by two-tailed Welch’s t-test for a, by Mann–Whitney U-test for c and by Student’s t-test for e and f. W, weeks. No significant differences were observed between Igfbp6^−/− mice and their WT littermates regarding the levels of plasma total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), LDL-C, aspartate aminotransferase (AST) or alanine aminotransferase (ALT) ([131]Extended Data Fig. 2c). As IGFBP6 is a secreted protein, we used ELISA to measure IGFBP6 levels in the serum of Igfbp6^−/− mice. IGFBP6 was depleted in the knockout mice ([132]Fig. 2c). Immunofluorescence staining also showed that IGFBP6 was drastically reduced in the RCA of Igfbp6^−/− mice ([133]Fig. 2d). Most notably, compared to WT mice, the area of atherosclerotic plaques in the LCA of Igfbp6^−/− mice was increased ([134]Fig. 2e). Oil Red O and immunofluorescence staining showed an increase in the area of lipid accumulation and vascular cell adhesion molecule-1 (VCAM-1) positivity in the endothelium layer of the LCA in Igfbp6^−/− mice ([135]Fig. 2f,[136]g). These findings indicate that the absence of Igfbp6 increases atherosclerotic lesions induced by DF in mice. EC-specific knockout of IGFBP6 aggravates atherosclerosis in mice To further explore the role of EC-derived IGFBP6 in atherosclerosis development in hypercholesterolemic mice, we infected male and female Igfbp6^ECKO (Cdh5-Cre; Igfbp6^flox/flox) mice and Igfbp6^WT (Igfbp6^flox/flox) littermate controls with AAV8-Pcsk9^D377Y ([137]Extended Data Fig. 3a,[138]b). En face staining of the aortic intima of mice showed a specific ablation of IGFBP6 in the ECs of Igfbp6^ECKO mice compared to Igfbp6^WT control mice ([139]Fig. 3b). Compared to control mice, Igfbp6 mRNA levels in the intimal layer but not in the medial layer of Igfbp6^ECKO mice aorta were decreased ([140]Fig. 3c). ELISA also indicated that serum levels of IGFBP6 in Igfbp6^ECKO mice were significantly reduced by approximately 70% ([141]Fig. 3d), whereas EC-specific deletion of Igfbp6 did not induce changes in mRNA expression of other members of the Igfbps in the aortic intima ([142]Extended Data Fig. 3c), consolidating the specificity of IGFBP6 knockout in mouse aortic endothelium. Fig. 3 |. EC-specific knockout of IGFBP6 aggravates atherosclerosis in mice. Fig. 3 | [143]Open in a new tab a, Schematic figure showing the experimental strategy. b, En face immunofluorescence staining for IGFBP6 (red), VE-cadherin (green) and DAPI (blue) in the aortas of mice. Decreased IGFBP6 expression in the intima of Igfbp6^ECKO mice was observed (n = 5). Scale bar, 50 μm. c, The expression of Igfbp6 mRNA in the intima and non-intima lysate of the aorta was detected by qRT–PCR (n = 5). d, IGFBP6 level in the serum of Igfbp6^ECKO mice was detected by ELISA (n = 4). e,f, Representative en face images of Oil Red O staining of atherosclerotic lesions of the aorta in male (e) (Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 7) and female Igfbp6^ECKO mice and respective controls (f) (Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 10). Scale bar, 2 mm. g,h, Representative images of atherosclerotic lesions in the aortic root stained with Oil Red O in male (g) (Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 7) and female Igfbp6^ECKO mice and respective controls (h) (Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 11). Scale bar, 200 μm. i, Representative Oil Red O–stained cryosections of brachiocephalic arteries from male Igfbp6^ECKO mice and control mice. The quantification of the Oil Red O–positive plaque area is shown in the right panel (Igfbp6^WT: n = 7; Igfbp6^ECKO: n = 9). Scale bar, 50 μm. j, Massonʼs trichrome staining showed collagen expression in plaques, and decreased collagen expression in the aortic roots of male Igfbp6^ECKO mice was observed as compared to control mice (Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 11). Scale bar, 200 μm. k, H&E staining of aortic root lesions was performed in male Igfbp6^ECKO mice and control mice, and the area of the necrotic plaque core was quantified (Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 11). Scale bar, 200 μm. l,m, Immunofluorescence staining of α-SMA (red) and CD68 (green) in the diseased area of the aortic root in male (l, Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 7) and female Igfbp6^ECKO mice as compared to control mice (m, Igfbp6^WT: n = 6; Igfbp6^ECKO: n = 8). Scale bar, 50 μm. Data are shown as mean ± s.d. Statistical analysis was performed by two-tailed Student’s t-test for c (left panel), e–k, l (right panel) and m (left panel), by Mann–Whitney U-test for c (right panel) and by Welch’s t-test for d and l (left panel) and m (right panel). W, weeks. At the end of 12 weeks of HCD feeding, no phenotypic differences were observed between Igfbp6^ECKO mice and Igfbp6^WT littermates regarding body weight, blood glucose or the levels of plasma TC, TG, HDL-C, LDL-C, AST or ALT ([144]Extended Data Fig. 3d). However, Oil Red O staining of mouse aorta and aortic sinus revealed a significant increase in atherosclerotic plaque area in both male and female Igfbp6^ECKO mice compared to the respective controls ([145]Fig. 3e–[146]h). In addition, EC-specific deletion of Igfbp6 also increased the accumulation of plaques in the brachiocephalic artery ([147]Fig. 3i). Massonʼs trichrome staining revealed a decrease in collagen content in the plaques of Igfbp6^ECKO mice ([148]Fig. 3j). Hematoxylin and eosin (H&E) staining of the aortic root showed that Igfbp6 ablation in ECs increased the size of lesional necrotic core ([149]Fig. 3k). In addition, α-SMA expression was decreased in the aortic plaques of Igfbp6^ECKO mice compared to the control group, whereas CD68^+ macrophage content was increased ([150]Fig. 3l,[151]m). These data suggest that EC-specific knockout of Igfbp6 increases atherosclerosis in both male and female mice and increases features of plaque instability. EC-specific overexpression of IGFBP6 ameliorates atherosclerosis in ApoE^−/− mice To correlate IGFBP6 level and plaque progression, we established a mouse model of progressive atherosclerosis from early (HCD feeding for 6 weeks) to advanced stage (HCD feeding for 18 weeks) of atherosclerosis. The plaque area of the en face aorta and aortic sinus of ApoE^−/− mice increased significantly with plaque progression compared to C57BL/6J control mice ([152]Fig. 4a). We observed that the expression of IGFBP6 in plaques of ApoE^−/− mice was reduced compared to healthy aortas ([153]Fig. 4b). Similar data were obtained in the serum of ApoE^−/− mice fed an HCD for 6–18 weeks ([154]Fig. 4c). These data implicate decreased expression level of IGFBP6 during atheroprogression. Fig. 4 |. EC-specific overexpression of IGFBP6 ameliorates atherosclerosis in ApoE^−/− mice. Fig. 4 | [155]Open in a new tab a, Representative images of atherosclerosis plaque development in the gross whole aorta and aortic sinus (stained with Oil Red O) of male C57BL/6J mice fed with a normal chow diet for 6 weeks and male ApoE^−/− mice fed with an HCD for 6 weeks or 18 weeks (n = 5). Scale bars, 1 mm and 200 μm. b, Immunofluorescence staining of IGFBP6 (red), CD31 (green) and DAPI (blue) in the diseased area of the aortic root in male C57BL/6J and ApoE^−/− mice (n = 5). Scale bar, 25 μm. c, Serum samples were collected from male C57BL/6J and ApoE^−/− mice to verify IGFBP6 levels by ELISA (n = 5). d, Schematic figure showing the experimental strategy. e, Immunofluorescence staining of IGFBP6 (red) in the diseased area of the aortic root in male AAV9-EC-Igfbp6 mice (n = 5). Scale bar, 50 μm. f, The expression of Igfbp6 mRNA in the intima and non-intima lysate of the aorta was detected by qRT–PCR (n = 5). g–i, Representative images of Oil Red O–stained aortas (g, n = 14), sections of aortic sinus (h, n = 9) and sections of brachiocephalic arteries (i, n = 10). Scatter plots show the statistical evaluation of the Oil Red O–positive areas. Scale bars, 2 mm (g and i) and 200 μm (h). j, Massonʼs trichrome staining showed increased collagen expression in the aortic roots of male AAV9-EC-Igfbp6 mice (n = 9). Scale bar, 200 μm. k, H&E staining of aortic root lesions was performed in male AAV9-EC-Igfbp6 mice, and the area of the necrotic core of the plaque was quantified (AAV9-EC-Con: n = 9; AAV9-EC-Igfbp6: n = 11). Scale bar, 200 μm. l,m, Immunofluorescence staining of α-SMA (red) and CD68 (green) in the diseased area of the aortic root and brachiocephalic arteries in male AAV9-EC-Igfbp6 mice (l, AAV9-EC-Con: n = 9 or n = 13; AAV9-EC-Igfbp6: n = 12; m, AAV9-EC-Con: n = 7 or n = 10; AAV9-EC-Igfbp6: n = 11 or n = 9). Scale bar, 50 μm. Data are shown as mean ± s.d. Statistical analysis was performed by two-tailed one-way ANOVA followed by Bonferroni’s test for c, by Student’s t-test for f–j and l (right panel), by Welch’s t-test for k and by Mann–Whitney U-test for l (left panel) and m. W, weeks. To determine whether IGFBP6 overexpression in ECs ameliorates atherosclerosis, we employed an EC-directed AAV9 vector driven by the ICAM-2 promoter to achieve EC-specific overexpression of Igfbp6 (AAV9-EC-Igfbp6) in 8-week-old male ApoE^−/− mice. Littermate controls were injected with a control virus (AAV9-EC-Con). Concurrently, all mice were fed an HCD ([156]Fig. 4d) for 12 weeks. Increased IGFBP6 levels were observed in aortic root and aortic intima in AAV9-EC-Igfbp6 mice compared to control mice ([157]Fig. 4e,[158]f and [159]Extended Data Fig. 4a). At the same time, EC-specific overexpression of Igfbp6 did not induce changes in mRNA expression of other members of the IGFBPs in the aortic intima ([160]Extended Data Fig. 4b). Terminally, no notable phenotypic differences were observed between the AAV9-EC-Igfbp6 mice and the control mice in terms of body weight, blood glucose and the levels of plasma TC, TG, HDL-C, LDL-C and AST, whereas the level of ALT tended to decrease ([161]Extended Data Fig. 4c). Oil Red O staining of the mouse aorta, aortic root and brachiocephalic arteries showed that EC-specific overexpression of Igfbp6 reduced the atherosclerotic plaque area ([162]Fig. 4g–[163]i). Massonʼs trichrome staining revealed that EC-specific overexpression of Igfbp6 increased collagen expression in atherosclerotic plaques ([164]Fig. 4j). In addition, H&E staining showed that EC-specific overexpression of Igfbp6 reduced the size of the necrotic core area in plaques ([165]Fig. 4k). AAV9-EC-Igfbp6 mice also exhibited increased α-SMA expression in the brachiocephalic arteries and aortic root as well as decreased CD68^+ macrophage infiltration ([166]Fig. 4l,[167]m). Collectively, these results suggest that EC-specific overexpression of Igfbp6 ameliorates atherosclerotic lesions in mice and enhances several features of plaque stability. IGFBP6 inhibits endothelial inflammation Next, we further explored the role of IGFBP6 in endothelial inflammation. After treatment with various pro-inflammatory factors (IL-1α, IL-1β, tumor necrosis factor (TNF), interferon-gamma (IFN-γ) and lipopolysaccharide (LPS)), we observed significant downregulation of IGFBP6 mRNA levels under various inflammatory factor treatments ([168]Fig. 5a). To assess whether IGFBP6 exerts anti-inflammatory effects in ECs, we first conducted experiments on HUVECs and human aortic endothelial cells (HAECs). We used siRNA (si-IGFBP6) or IGFBP6 overexpression adenovirus (Ad-IGFBP6) to knock down or overexpress IGFBP6, respectively, and assessed their impacts on TNF-induced inflammatory response. First, we confirmed successful IGFBP6 knockdown by ELISA ([169]Fig. 5b). In IGFBP6-silenced HUVECs, both the mRNA and protein levels of VCAM-1, intercellular adhesion molecule-1 (ICAM-1) and E-selectin (SELE) were significantly elevated ([170]Fig. 5c,[171]d). In contrast, in IGFBP6-overexpressing HUVECs, both the mRNA and protein levels of VCAM-1, ICAM-1 and SELE were decreased compared to control ([172]Fig. 5e,[173]f). Of functional relevance, knockdown of IGFBP6 increased the adhesion of THP-1 monocytes to HUVECs ([174]Fig. 5g), whereas overexpression of IGFBP6 inhibited THP-1 monocyte adhesion to HUVECs ([175]Fig. 5h). The anti-adhesive effect of IGFBP6 was recapitulated in HAECs ([176]Extended Data Fig. 5a–[177]f). Fig. 5 |. IGFBP6 regulates EC inflammation. Fig. 5 | [178]Open in a new tab a, HUVECs were treated with various inflammatory stimuli (IL-1α, IL-1β, TNF, IFN-γ and LPS) for 24 h, and the mRNA expression of VCAM-1 and IGFBP6 was detected (n = 3–4). b, HUVECs were transfected with si-NC or si-IGFBP6 for 48 h and then treated with TNF for 6 h. The expression of IGFBP6 was determined by ELISA (b, n = 3). c,d, The expression of VCAM-1, ICAM-1 and SELE was determined by qRT–PCR (c, n = 5) and western blot (d, VCAM-1 and ICAM-1: n = 9; SELE: n = 3). e,f, HUVECs were transfected with Ad-NC or Ad-IGFBP6 for 24 h and then treated with TNF for 6 h. The expression of IGFBP6/Flag, VCAM-1, ICAM-1 and SELE was determined by qRT–PCR (e, n = 3) and western blot (f, VCAM-1 and ICAM-1: n = 5; SELE: n = 3). g, Number of adherent THP-1 cells to HUVECs treated with si-NC or si-IGFBP6 in the presence of TNF (n = 6). Scale bar, 50 μm. h, Number of adherent THP-1 cells to HUVECs treated with Ad-NC or Ad-IGFBP6 in the presence of TNF (n = 6). Scale bar, 50 μm. i,j, HUVECs were transfected with Ad-NC or Ad-IGFBP6 for 24 h and then treated with UF or DF for 24 h, respectively. The expression of VCAM-1 and ICAM-1 was detected by qRT–PCR (i, n = 3) and western blot (j, n = 4). k, Number of adherent THP-1 cells to HUVECs treated with Ad-NC or Ad-IGFBP6 in the presence of DF or static (n = 4). Scale bar, 50 μm. l, After intraperitoneal injection of murine TNF (500 ng per mouse) for 4 h, the leukocytes of mice were stained with Rhodamine 6G, and the number and migration rate of leukocytes in mesenteric vessels were observed and counted (n = 3). Scale bar, 100 μm. Data are shown as mean ± s.d. Statistical analysis was performed by two-tailed Student’s t-test for l (left and middle panel), by paired t-test for a, b, d and f, by two-way ANOVA (repeated measures) followed by Bonferroni’s test for c, e, g, i and j, by two-way ANOVA followed by Tukey’s test for h and k and by Welch’s t-test for l (right panel). Veh, vehicle. Furthermore, RNA-seq data indicate that IGFBP6 overexpression led to changes of DEGs related to TNF signaling pathway, fluid shear stress and atherosclerosis ([179]Extended Data Fig. 5g,[180]h). Specifically, overexpression of IGFBP6 in ECs reduced the mRNA expression of adhesion molecules (VCAM-1, ICAM-1 and SELE) and chemokines (C-C motif chemokine ligand 2 (CCL2), C-X-C motif chemokine ligand 1 (CXCL1), CXCL3, CXCL6 and CXCL8) ([181]Extended Data Fig. 5i). Subsequently, we investigated whether IGFBP6 exerts anti-inflammatory effects in ECs exposed to DF. Compared to UF, exposure to DF upregulated the expression of ICAM-1 and VCAM-1 genes in HUVECs, which was reversed by IGFBP6 overexpression ([182]Fig. 5i). In parallel, DF-induced VCAM-1 protein upregulation was reversed by IGFBP6 overexpression ([183]Fig. 5j). As a consequence, overexpression of IGFBP6 inhibited the adhesion of THP-1 monocytes to HUVECs exposed to DF ([184]Fig. 5k). Finally, we evaluated whether Igfbp6 impacts leukocyte adhesion in vivo by intravital microscopy. We injected TNF into the peritoneal cavity of male Igfbp6^ECKO mice and Igfbp6^WT control mice at 4–6 weeks of age to induce leukocyte adhesion. We observed that the EC-specific knockout of Igfbp6 increased the number of rolling and adhering leukocytes in the mesenteric vessels of mice and slowed the movement of leukocytes ([185]Fig. 5l). These data indicate that IGFBP6 alleviates endothelial inflammation induced by inflammatory factors and prevents leukocyte adhesion to activated endothelium, further consolidating the protective role of IGFBP6 against endothelial dysfunction. IGFBP6 is a transcriptional target of KLF2 KLF2 is a master transcription factor responsive to shear stress and regulates the expression of a battery of mechanosensitive genes associated with vascular homeostasis^[186]29–[187]31. By analyzing the human IGFBP6 promoter region, we identified multiple putative binding sites of KLF2. Therefore, we examined the possibility of whether KLF2 could transcriptionally regulate IGFBP6 expression in ECs. We thus treated HUVECs with three different statins (atorvastatin, simvastatin and rosuvastatin) as well as resveratrol at different concentrations for 24 h. We observed a concentration-dependent upregulation of KLF2 and IGFBP6 mRNA expression in statin-treated and resveratrol-treated ECs ([188]Fig. 6a–[189]d). Fig. 6 |. IGFBP6 is a transcriptional target of KLF2. Fig. 6 | [190]Open in a new tab a–d, HUVECs were treated with escalating concentrations of indicated chemicals (atorvastatin (a), simvastatin (b), rosuvastatin (c) and resveratrol (d)) for 24 h. Then, the mRNA expression of KLF2 and IGFBP6 was detected (n = 5). e, HUVECs were treated with si-NC or si-KLF2 for 48 h, and the mRNA expression of KLF2 and IGFBP6 was detected (n = 3). f, HUVECs were treated with siRNA or si-KLF2 for 24 h and then treated with DMSO or atorvastatin for 24 h, and the mRNA expression of KLF2 and IGFBP6 was detected (n = 5). g,h, HUVECs were treated with Ad-NC or Ad-KLF2 for 24 h, and the expression of KLF2 and IGFBP6 was determined by qRT–PCR (g, n = 4) and ELISA (h, n = 3). i, Dual-luciferase reporter assay showed (293T) that KLF2 regulated IGFBP6 promoter activity in HEK293T cells transfected with different mutants of IGFBP6 promoters (n = 5). j, In Ad-KLF2-transduced HUVECs, chromatin immunoprecipitation–PCR showed that KLF2 bound to the IGFBP6 promoter (n = 3). Data are shown as mean ± s.d. Statistical analysis was performed by two-tailed one-way ANOVA (repeated measures) followed by Bonferroni’s test for a and b (left panel) and d and f, by Kruskal–Wallis test for b (right panel), by Mann–Whitney U-test for g (right panel), by Welch’s t-test for i (Ad-NC WT versus Ad-KLF2 WT), by one-way ANOVA followed by Tukey’s test for i (Ad-KLF2 WT versus Ad-KLF2 MT1 versus Ad-KLF2 MT2), by paired t-test for c, e and g (left panel) and j and by Student’s t-test for h. We then employed siRNA (si-KLF2) or Flag-tagged adenovirus (Ad-KLF2) to knock down or overexpress KLF2 in HUVECs, respectively. We found that KLF2 positively regulates the mRNA expression of IGFBP6 ([191]Fig. 6e–[192]h). Also, we found that KLF2 depletion partially reversed the upregulation of IGFBP6 mRNA expression induced by atorvastatin ([193]Fig. 6f). Next, we constructed various plasmid vectors (including empty vector (basic), vector containing the IGFBP6 promoter (WT) and vectors containing mutations that deleted all CACCC (MT1) or GGGTG (MT2) sequences in the IGFBP6 promoter) for the dual-luciferase reporter assay. The results showed that overexpression of KLF2 increased the activity of the WT IGFBP6 promoter and the MT1 IGFBP6 promoter but not the MT2 IGFBP6 promoter, indicating that KLF2 could enhance the activity of the IGFBP6 promoter and bind to the GGGTG sequence in the IGFBP6 promoter ([194]Fig. 6i). Chromatin immunoprecipitation–PCR also confirmed that KLF2 binds to the IGFBP6 promoter region ([195]Fig. 6j). Taken together, these findings collectively indicate that IGFBP6 is a bona fide transcriptional target of KLF2. Because KLF4 regulates a plethora of overlapping genes as KLF2, we also confirmed that KLF4 also regulates the expression of IGFBP6. The results suggest that IGFBP6 may also be a transcriptional target of KLF4 ([196]Extended Data Fig. 6a,[197]b). IGFBP6 interacts with MVP and suppresses endothelial inflammation via MVP-dependent inhibition of p65 and JNK phosphorylation To unravel the direct mechanism of the anti-inflammatory effects of IGFBP6 in ECs, we precipitated IGFBP6 from overexpressed ECs followed by mass spectrometry. Based on protein-specific peptide segments and scores, MVP had a high ranking score ([198]Fig. 7a). Co-immunoprecipitation assays revealed an interaction between exogenous IGFBP6 and endogenous MVP in ECs ([199]Fig. 7b). Co-transfection of Flag-tagged IGFBP6 and HA-tagged MVP in HEK293T cells revealed that IGFBP6 interacted with MVP ([200]Fig. 7c). Immunofluorescence staining also showed that IGFBP6 co-localized with MVP in the cytoplasm of ECs ([201]Fig. 7d). Through surface plasmon resonance (SPR) experiments, we found that MVP can directly bind to the IGFBP6 protein in a concentration-dependent manner ([202]Fig. 7e). Fig. 7 |. IGFBP6 interacts with MVP and suppresses endothelial inflammation via MVP-dependent inhibition of p65 and JNK phosphorylation. Fig. 7 | [203]Open in a new tab a, HUVECs were treated with Ad-NC or Ad-IGFBP6 for 24 h before treatment with TNF for 6 h. Lysates were immunoprecipitated with anti-Flag-M2 beads. List of IGFBP6-binding proteins identified by LC–MS/MS. b,c, Immunoprecipitation (IP) was performed with the indicated antibodies in HUVECs (b) and HEK293T cells (c). d, Immunofluorescence staining for IGFBP6 (red), MVP (green) and DAPI (blue) in HUVECs (n = 4). Scale bar, 10 μm. e, SPR analysis of the direct interaction of MVP and IGFBP6. f, HUVECs were transfected with si-NC or si-MVP for 48 h and treated with TNF for 6 h. Whole cell lysate of treated cells was immunoblotted to detect MVP, ICAM-1, P-p65, p65, P-JNK and JNK (n = 3). g,i, ECs were transfected with si-NC or si-IGFBP6 for 48 h and then treated with TNF for different times. The protein expression of P-p65, p65, P-JNK and JNK was determined in HAECs (g) and HUVECs (i) (n = 3). h,j, ECs were transfected with Ad-NC or Ad-IGFBP6 for 24 h and then treated with TNF for different times. The protein expression of P-p65, p65, P-JNK and JNK was determined in HAECs (h) and HUVECs (j) (n = 3). k, HUVECs were transfected with adenovirus (Ad-NC or Ad-IGFBP6) and siRNA (si-NC or si-MVP) for 48 h and then treated with TNF for 6 h. THP-1 monocyte adhesion assay was performed (n = 3). Scale bar, 50 μm. l, The protein expression of Flag-tagged IGFBP6, MVP, ICAM-1, P-p65, p65, P-JNK and JNK was determined in HUVECs (n = 3). m,n, Immunofluorescence staining of P-JNK (red) and JNK (green) in the diseased area of the aortic root in male Igfbp6^ECKO mice (m, Igfbp6^WT: n = 8; Igfbp6^ECKO: n = 13) and male AAV9-EC-Igfbp6 mice (n, AAV9-EC-Con: n = 8; AAV9-EC-Igfbp6: n = 6). Scale bar, 50 μm. Data are shown as mean ± s.d. Statistical analysis was performed by two-tailed Student’s t-test for k, by Mann–Whitney U-test for m and by Welch’s t-test for n. Previous studies demonstrated that MVP suppresses inflammation by inhibiting the TNF receptor-associated factor 6 (TRAF6)–NF-κB and apoptosis signal-regulating kinase 1 (ASK1)–JNK signaling pathways in macrophages, leading to protection against atherosclerosis in mice^[204]32,[205]33. We demonstrated that MVP silencing in HUVECs increased ICAM-1 and SELE mRNA expression levels, increased monocyte adhesion to HUVECs and increased the phosphorylation of JNK and p65 ([206]Fig. 7f and [207]Extended Data Fig. 6c,[208]d). Moreover, we induced endothelial cell inflammation by treating HUVECs and HAECs with TNF at different timepoints. In IGFBP6-silenced HAECs, the expression of phosphorylated JNK at serine 183 and threonine 185 was increased, whereas the overexpression of IGFBP6 significantly decreased the expression of phosphorylated JNK and p65 ([209]Fig. 7g,[210]h). In HUVECs, similar results were obtained ([211]Fig. 7i,[212]j). Interestingly, MVP silencing reversed the preventive effect of IGFBP6 on monocyte adhesion ([213]Fig. 7k) and JNK and p65 phosphorylation ([214]Fig. 7l). Immunofluorescence staining of the mouse aortic sinus revealed that EC-specific knockout of Igfbp6 increased JNK phosphorylation in plaques, whereas EC-specific overexpression of Igfbp6 exhibited the opposite effects ([215]Fig. 7m,[216]n). To identify the specific mechanism whereby IGFBP6 inhibits inflammation through MVP, we explored the mechanistic details downstream of IGFBP6–MVP interaction. In previous reports, MVP in macrophages inhibited downstream JNK and p65 activation by inhibiting the dimerization of ASK1 and TRAF6 (refs. [217]32,[218]33). We first verified in HUVECs that MVP can also bind to ASK1 ([219]Extended Data Fig. 7a). The interaction between exogenous IGFBP6 and endogenous ASK1 in ECs was further validated by co-immunoprecipitation ([220]Extended Data Fig. 7b). We next evaluated the effect of IGFBP6 on the dimerization of ASK1. Our findings verified the exogenous interaction between ASK1 and MVP and the inhibition of ASK1 dimerization by MVP. IGFBP6 also binds with ASK1 and increases the interaction between MVP and ASK1. Most importantly, IGFBP6 promoted the binding of MVP to ASK1, further inhibiting the ASK1 dimerization ([221]Extended Data Fig. 7c). As a result, IGFBP6 downregulates the phosphorylation of JNK downstream of ASK1 dimerization. We then employed AlphaFold 3–based molecular docking to predict the binding sites of IGFBP6 and MVP and pre-selected three possible binding sites according to screening conditions, such as hydrogen bond distance ([222]Extended Data Fig. 7d). We constructed four mutants of IGFBP6, including three mutants with a single point mutation to alanine (E48A/R65A/R103A) and one mutant carrying triple point mutations (E48A + R65A + R103A). All these plasmid vectors carried flag tags and were transfected into HEK293T cells together with HA-MVP vector. Our results showed that a single mutation at E48 or R65 or R103 could slightly reduce the interaction between IGFBP6 and MVP with varying potencies, whereas mutation of all three potential binding sites could significantly dampen the binding between IGFBP6 and MVP ([223]Extended Data Fig. 7e). We then explored whether the anti-inflammatory effect of IGFBP6 depended on these key binding sites. We constructed IGFBP6 adenovirus with mutations at three binding sites and transfected into HUVECs for downstream functional assays. The results showed that overexpression of WT IGFBP6 could significantly inhibit the inflammation of ECs, whereas this anti-inflammatory effect was weakened in ECs infected with the triple mutants of IGFBP6, indicating that these three binding sites were at least partially involved in relaying the anti-inflammatory IGFBP6–MVP signaling axis ([224]Extended Data Fig. 7f). These results suggested that IGFBP6 exerted anti-inflammatory effects by binding to MVP. It was reported that MVP can be detected in human serum as a biomarker of disease^[225]34. We speculate whether IGFBP6 is secreted extracellularly with MVP to play an anti-inflammatory role. First, we detected MVP protein extracellularly in conditioned media from MVP-overexpressed ECs ([226]Extended Data Fig. 8a). We next explored the possibility of the extracellular interaction between IGFBP6 and MVP. Our data demonstrated that MVP and IGFBP6 secreted in concentrated endothelial conditioned media do not interact with each other ([227]Extended Data Fig. 8a,[228]b). To further validate whether secreted IGFBP6 exerts anti-inflammatory effects, we treated HUVECs with different amounts of recombinant human IGFBP6 protein before stimulation with TNF. Our data showed that extracellular IGFBP6 had no observable anti-inflammatory effects. The phosphorylation of JNK and p65 was not affected either ([229]Extended Data Fig. 8c,[230]d). Similarly, the conditioned medium from HUVECs overexpressing IGFBP6 did not elicit appreciable anti-inflammatory effects ([231]Extended Data Fig. 8e). Subsequently, we further evaluated the anti-inflammatory effect of extracellular MVP and found that different concentrations of recombinant MVP had no anti-inflammatory effect on ECs ([232]Extended Data Fig. 8f). In summary, these findings indicate that IGFBP6 exerts its anti-inflammatory effects mainly by interacting with intracellular MVP rather than extracellular MVP. Taken together, IGFBP6 inhibits JNK and p65 through its interaction with intracellular MVP, thereby reducing the expression of adhesion molecules and inhibiting monocyte adhesion to ECs, ultimately combating endothelial inflammation. Discussion In this study, we demonstrated that IGFBP6 is a negative regulator of endothelial inflammation and leukocyte adhesion, thereby inhibiting the development of atherosclerosis (Graphic Abstract and [233]Extended Data Fig. 9). This conclusion is backed by multiple findings from cultured human ECs, various mouse models and human patients. The salient findings of the present study can be summarized as follows: (1) the expression of IGFBP6 is decreased in serum and atherosclerotic tissues of patients with CAD; (2) gain and loss of function of endothelial Igfbp6 in atheroprone mouse models suggest that Igfbp6 ameliorates the progression of atherosclerosis in mice; and (3) IGFBP6 is a transcriptional target of KLF2 that inhibits EC inflammation and monocyte adhesion through the MVP–JNK/NF-κB pathway. KLF2 is a key regulator of endothelial homeostasis and atheroprotection. Some chemicals have been shown to increase KLF2 expression or activity, such as statins and resveratrol^[234]30,[235]35,[236]36. In the present study, we found that both statins and resveratrol upregulate IGFBP6 expression in ECs by activating KLF2, suggesting that IGFBP6 is druggable and that its upregulation may contribute to the pleiotropic and atheroprotective effects of statins and resveratrol. To elucidate the molecular mechanism by which IGFBP6 suppresses inflammation, we identified MVP as an IGFBP6-interacting protein that mediates the anti-inflammatory effects of IGFBP6. MVP was shown in macrophages to limit atherosclerosis through the ASK1–JNK and NF-κB signaling pathway^[237]32,[238]33. Similar to the role of MVP in macrophages, we reported that IGFBP6 also decreased the phosphorylation of JNK and NF-κB (p65) dependent on MVP in endothelial cells. We further found that IGFBP6 also interacts with ASK1, and IGFBP6 mitigates the dimerization of ASK1 by binding to MVP. We predicted the potential binding sites of IGFBP6 and MVP by molecular docking and confirmed the key binding domains in this interaction. All these lines of evidences collectively pinpoint the anti-inflammatory effects of IGFBP6 in ECs through binding with MVP. The IGFBP family consists of six high-affinity proteins (IGFBP1–6) and one low-affinity protein (IGFBP7) for IGF, which regulate IGF utilization, cell proliferation and differentiation^[239]37. The role of IGFBPs in atherosclerosis is being recognized. For example, IGFBP1 attenuates atherosclerosis in mice through the PI3K–Akt–eNOS pathway^[240]38. IGFBP2 is a positive predictor of MACE in patients with acute coronary syndrome^[241]39. Cell senescence promotes fibrous cap degradation and ultimately leads to atherosclerotic plaque rupture through IGFBP3 (ref. [242]40). When compared to non-lesional areas, IGFBP4 is upregulated in the arterial lesion area of ApoE^−/− mice^[243]41. Our previous study reported that IGFBP5 is mechanoresponsive and anti-inflammatory^[244]28. IGFBP7 expression is increased in patients with atherosclerotic aortic aneurysm^[245]42. It is known that IGFBPs play different roles in cardiovascular diseases, including atherosclerosis, because different IGFBPs have different tissue expression levels and may exert different pathophysiological functions under different disease contexts. The expression levels of IGFBPs in ECs varied greatly, with IGFBP1–5 being lowly expressed in HUVECs, whereas IGFBP6 and IGFBP7 had relatively high expression levels ([246]Extended Data Fig. 1c). According to the previous database mining and the mechanoresponsive nature of IGFBP6, we observed that, compared to IGFBP7, IGFBP6 is an EC-enriched IGFBP member that responds to blood flow shear stress. Nevertheless, the exact role of IGFBP6 in vascular biology and diseases remains largely unknown. Previous studies showed that IGFBP6 is essential for regulating cell cycle, cell proliferation and cell migration^[247]19–[248]21. In ECs, IGFBP6 inhibits angiogenesis independently of IGF2, leading to a significant reduction in the number of blood vessels in zebrafish embryos as well as a marked decrease in the blood vessel density of BALB/c nude mouse xenografts^[249]20. The anti-angiogenic role of IGFBP6 aligns with the atheroprotective effects of IGFBP6 because neovascularization may promote the progression of atherosclerotic disease and renders the plaque unstable^[250]43. Also, it was reported that, compared to stable plaques in patients with CAD, the expression level of IGFBP6 in unstable plaques was decreased^[251]22. However, the direct role and mechanism of IGFBP6 in endothelial homeostasis and atherogenesis remain unclear. In our study, global or EC-specific deletion of Igfbp6 exacerbated HCD-induced or DF-induced atherosclerotic lesion development. In contrast, overexpression of Igfbp6 in ECs ameliorated HCD-induced atherosclerotic lesion development. These whole battery of gain-of-function and loss-of-function studies in mice suggests that endothelial IGFBP6 negatively regulates the progression of atherosclerotic lesions and highlights the potential of IGFBP6-elevating strategies for treating atherosclerotic diseases. We recognize that this study has several limitations. First, IGFBP6 was also expressed in non-ECs of blood vessels, such as vascular smooth muscle cells (VSMCs). However, it remains unexplored in this study whether VSMC-derived IGFBP6 has similar atheroprotective effects. Second, the paracrine functions of IGFBP6 secreted from ECs on neighboring VSMCs or macrophages remain to be explored. Finally, from the translational perspective, liposome nanoparticles carrying IGFBP6 mRNA to deliver IGFBP6 specifically to plaque ECs are needed to achieve EC-targeted IGFBP6 delivery for cardiovascular therapeutics. In conclusion, our study suggests that IGFBP6 is mechanoresponsive, anti-inflammatory and atheroprotective. The elevation of endothelial IGFBP6 genetically or pharmacologically may represent a strategy to reduce residual inflammatory risk for CAD. Methods Experimental mice and treatment Male ApoE^−/− mice at 8 weeks of age were purchased from GemPharmatech (no. T001458). Igfbp6^flox/flox and Igfbp6^−/− mice were sourced from the Shanghai Model Organisms Center (no. NM-KO-205448 and no. NM-CKO-2107216). Igfbp6^ECKO mice were generated by crossing Igfbp6^flox/flox mice with Cdh5-Cre mice (Shanghai Model Organisms Center, no. 017968), resulting in EC-specific deletion of Igfbp6. Eight-week-old male ApoE^−/− mice were subjected to EC-specific overexpression of Igfbp6 using an EC-specific AAV9 vector driven by the ICAM-2 promoter (AAV9-EC-Igfbp6, 1 × 10^12 vg per mouse)^[252]44. In addition to ApoE^−/− mice, hepatic LDLR protein was downregulated by a single dose of AAV8-Pcsk9^D377Y (3 × 10^11 vg per mouse) injected into the tail vein^[253]45. The AAVs were constructed by Vigene Biosciences. All mice were fed an HCD (Research Diets, no. D12108C; 20% fat and 1.25% cholesterol) to induce atherosclerosis. Mice were housed in specific pathogen-free (SPF) standard cages with a 12-h light/dark cycle and allowed to freely consume standard diets with a temperature of 20–24 °C and a humidity of 30–70%. The animal study protocols were approved by the Animal Care and Use Committee of University of Science and Technology of China (USTCACUC212401038) and carried out in accordance with the guidelines for the care and use of laboratory animals from the National Institutes of Health. Human samples Human samples were obtained from The First Affiliated Hospital of the University of Science and Technology of China, with the research involving these samples approved by the hospital’s ethics committee (2023KY-383 and 2024KY-397). Patient tissue samples were obtained from the coronary arteries of heart transplant recipients with heart failure after myocardial infarction (n = 3), and healthy human tissue samples were taken from coronary arteries of abandoned heart transplant donors (n = 3). Serum samples from healthy individuals were obtained from a health examination center (n = 12), whereas those from patients with CAD were obtained from the department of cardiac surgery (n = 19). Serum was used for ELISA experiments, and coronary artery tissues were embedded in formalin and sectioned continuously after embedding in optimal cutting temperature compound (OCT; Sakura, no. 4583) for immunofluorescence staining and other experiments. This study was conducted in accordance with the Declaration of Helsinki, and all participants provided informed consent. Patient clinical characteristics are provided in [254]Supplementary Tables 1 and [255]2. Chemicals and reagents Recombinant human TNF (no. 300–01A), IL-1α (no. 200–01A), IL-1β (no. 200–01B) and IFN-γ (no. 300–02) were purchased from PeproTech, dissolved in sterile distilled water and then diluted with 0.1% BSA (Sigma-Aldrich, no. 9048–46-8) in PBS to a final concentration of 10 ng ml^−1 for in vitro experiments. The amount of recombinant murine TNF (BBI, no. C600052–0005) was 500 ng per mouse for intravital microscopy. LPS (no. [256]T11855), atorvastatin (no. 134523–00-5), simvastatin (no. 79902–63-9), rosuvastatin (no. 287714–41-4) and resveratrol (no. 501–36-0) were purchased from TargetMol. LPS was added to HUVECs at a final concentration of 1 μmol L^−1. Calcein AM (Beyotime Biotechnology, no. 148504–34-1) was used to stain THP-1 monocytes. Rhodamine 6G (Sigma-Aldrich, BCCF1630) was used to stain leukocytes in mice. The antibodies used in this study included anti-IGFBP6 (Abcam, ab219560, lot no. GR3219721–4; 1:1,000), anti-Flag (Sigma-Aldrich, no. F1804; 1:2,000) and anti-VCAM-1 (Zenbio, no. 381014, lot no. L28JU0P; 1:1,000) for immunoblotting; anti-VCAM-1 (Cell Signaling Technology (CST), no. 39036; 1:100) for immunofluorescence; and anti-ICAM-1 (Zenbio, no. 200350-D6; 1:1,000), anti-SELE (BioVision, no. 3631; 1:1000), anti-MVP (Huabio, no. ET1705–69, lot no. HK0904; 1:1,000), anti-HA (Proteintech, no. 81290–1-RR, lot no. 00116648; 1:1,000), anti-P-p65 (CST, no. 3033S, lot no. 19; 1:1,000), anti-p65 (CST, no. 6956S, lot no. 10; 1:1,000), anti-P-JNK (CST, no. 81E11, lot no. 17; 1:1,000), anti-JNK (Proteintech, no. 66210–1-Ig, lot no. 10023911; 1:1,000), anti-ASK1 (CST, no. 3762; 1:1000), anti-CD144 (BD Biosciences, no. 555289 and no. 555661, lot no. 1076252; 1:100), anti-CD31 (BD Biosciences, no. 553370, lot no. 1005853; 1:100), anti-CD68 (BD Biosciences, no. 566386; 1:100), anti-α-SMA (Proteintech, no. 14395–1-AP, lot no. 00102366; 1:100), anti-GAPDH (Proteintech, no. 60004–1-Ig, lot no. 10029187; 1:10,000), anti-β-actin (Proteintech, no. 66009–1-Ig, lot no. 10029234; 1:1,000) and anti-IgG (Proteintech, no. 30000–0-AP; 1:100; B900620 and B900630). Cell culture and shear stress experiments Different donors of HUVECs (cat. no. FC-0003, lot no. 08119/10128/04 608/07815/06290/04827/08478/06279) and HAECs (cat. no. FC-0014, lot no. 02357/01921/04675) were purchased from Lifeline Cell Technology. In this study, 3rd–7th generation HUVECs and HAECs were used^[257]46. The cells were cultured in ECM medium, which contained 1× endothelial cell growth supplement (ScienCell), 5% FBS and 1% penicillin–streptomycin antibiotics. HEK293T cells were purchased from the American Type Culture Collection (ATCC) using high-glucose DMEM containing 10% FBS (Sigma-Aldrich, no. 1943609–65-1) and 1% penicillin–streptomycin antibiotics (Sigma-Aldrich, RNBL3872). The human monocyte line THP-1 was purchased from the ATCC in RPMI 1640 medium supplemented with 10% FBS (Biosharp, no. BL303A). All the above cells were cultured at 37 °C and 5% CO[2]. In the in vitro laminar flow shear stress experiment, confluent HUVEC monolayers were seeded on 100-mm plates, and a cone-and-plate shear stress device (Jiangsu Stoli Instrument Co., Ltd.) was used to generate DF or UF for 24 h^[258]47. The flow system was enclosed in a chamber maintained at 37 °C and ventilated with 95% humidified air and 5% CO[2]. Monocyte adhesion assay HUVECs or HAECs were treated with adenovirus or siRNA. THP-1 cells cultured in RPMI 1640 medium were counted to ensure that the cell density was approximately 10^5 cells per milliliter. THP-1 cells were then stained with the 5 μM fluorescent dye Calcein-AM and incubated at 37 °C in the dark for 30 min. After centrifugation, the cell pellet was washed three times with RPMI 1640 medium, and the THP-1 cells were resuspended and evenly added to each well. The cells were incubated at 37 °C in the dark for 30 min. Non-adherent cells were removed by washing three times with pre-warmed ECM medium and then images were captured under a microscope (Carl Zeiss Vision, Axio Vert.A1) to quantify the number of adherent cells. Intravital microscopy of leukocyte–endothelial interaction in mesenteric veins in mice Four- to six-week-old male Igfbp6^ECKO mice and littermate control Igfbp6^WT mice with body weights ranging from 16 g to 20 g were intraperitoneally injected with recombinant murine TNF (500 ng per mouse) to induce leukocyte adhesion in mice. Four hours later, Rhodamine 6G was injected into the tail vein to stain the leukocytes in the mice. Then, the mice were anesthetized, and the abdomen of the mice was sterilized. A median laparotomy was performed on the mice, and several drops of pre-warmed saline were dropped into the abdominal cavity to keep the tissue moist. Mesenteric veins were spread out in a 100-mm dish, and 200–300-μm mesenteric veins were placed in the center of the field of view of the microscope. Images were then acquired using an inverted microscope (Nikon Ti2-E) equipped with a Yogokawa CSU-W1 Spinning Disk Confocal Scanner Unit^[259]48. Fluorescence was visualized with the 488-nm laser, combined with emission filter (B525/50), recorded by an sCMOS camera (Photometrics, Prime95B). Imaging data were analyzed using NIS-Elements AR software (version 5.20.00)^[260]48. Leukocyte–endothelial interaction was recorded for 1 min in 3–5 different veins from each mouse. The amount of rolling and adherent leukocytes was finally quantified, and the rolling velocity was determined by measuring the time a single leukocyte would roll steadily over the endothelium simultaneously across a 100-μm distance. Serum lipid profile After anesthesia with 2.5% isoflurane gas, blood was collected from the mice through the orbital venous plexus, and serum was collected by centrifugation at 3,000 r.p.m. for 10 min at room temperature. Lipid levels were measured by Servicebio Technology using a detection kit (Ranto) to measure serum TC (no. S03042), TG (no. S03027), HDL-C (no. S03025), LDL-C (no. S03029), ALT (no. S03030) and AST (no. S03040). Partial ligation of the LCA Eight-week-old male Igfbp6^−/− and WT mice were injected with 3 × 10^11 vg per mouse AAV8-Pcsk9^D377Y (Vigene Biosciences, Addgene plasmid no. 58376; [261]http://n2t.net/addgene:58376; RRID: Addgene_58376; gifted from Jacob Bentzon) via the tail vein and concurrent with feeding with an HCD^[262]45,[263]49,[264]50. After 2 weeks, all mice were anesthetized with 2.5% isoflurane gas, and the ventral midline of the neck was opened approximately 3–4 mm. The left external carotid artery (ECA) was ligated first using a 6–0-gauge suture; then, the internal carotid artery (ICA) was ligated together with the occipital artery (OA); and finally, only the superior thyroid artery (STA) was retained, and the mouse wound was sutured. Mice were observed daily for 3–4 d after surgery, and mice with dehiscence of the wound were disinfected. After 3 weeks, the mice were euthanized; the bilateral carotid arteries were fixed with 4% paraformaldehyde (PFA); and the dehydrated carotid arteries were stored under OCT using 30% sucrose and immediately frozen. Frozen sections were stained using Oil Red O or immunofluorescence. Plasmid construction and viral or siRNA transfection Recombinant adenoviruses encoding Flag-tagged human IGFBP6 (Ad-IGFBP6; [265]NM_002178.3), human KLF2 (Ad-KLF2; [266]NM_016270.4), Flag-tagged human IGFBP6 mutation (Ad-IGFBP6[E48A+R65A+R103A]) and control adenovirus (Ad-NC) were constructed by Vigene Biosciences. siRNA (si-IGFBP6, si-KLF2 and si-MVP) or control (si-NC) for human IGFBP6, KLF2 and MVP ([267]NM_017458.3) or control (si-NC) were purchased from RiboBio Co., Ltd. The sequences of all siRNAs are provided in [268]Supplementary Table 3. Lots of plasmids (HA-MVP, Myc-MVP, ASK1-Flag, ASK1-HA and IGFBP6-Flag (E48A, R65A and E103A)) were constructed by Vigene Biosciences. All plasmid sequences are listed in [269]Supplementary Table 4. Adenovirus was used at a titer of multiplicity of infection (MOI) = 1, and the cells showed no obvious toxicity. Lipofectamine 2000 (Invitrogen, no. 11668–019) was used to transfect ECs and HEK293T cells. Cells cultured in 12-well plates were transfected with 20 nmol L^–1 siRNA or 1 μg of plasmid per well, and HEK293T cells were transfected using 4 μg in 100-mm dishes for 24 h. Total RNA extraction, RNA-seq and qRT–PCR RNA was isolated using RNeasy kits (Yishan Biotech). After quality control of RNA, a library for RNA-seq was established at the Beijing Genomics Institute. In brief, mRNAs were separated from total RNA using magnetic beads attached to oligonucleotides (dT). First-strand cDNA was generated by reverse transcription with random hexamer primers, and then the second-strand cDNA was synthesized. The synthesized cDNA was subjected to end repair, and 3′ adenylation was then performed. Adapters were attached to the ends of these 3′ adenylated cDNA fragments. The cDNA fragments were amplified by PCR, purified with AMPure XP Beads (Thermo Fisher Scientific, Agencourt) and dissolved in EB solution. The library was validated on a Bioanalyzer system (Agilent Technologies, no. G2939BA). The double-stranded PCR products were thermatively denatured and circularized using the splint oligo sequence. The single-strand circular DNA (ssCir DNA) was formatted into the final library. The library was amplified using phi29 to generate DNA nanospheres (DNA-NSs), each containing over 300 copies of a single molecule. The DNA-NSs were loaded into the patterned nanoarray, and single-end 50 (paired-end 100/150) base reads were produced by combinatorial probe-anchor synthesis (cPAS). Total RNA was reverse transcribed into cDNA using reverse transcription kits (TaKaRa, no. RR037A). Upon completion of the reverse transcription process, real-time PCR was carried out using a Roche LC96 real-time PCR detection system using AceQ qPCR SYBR Green Master Mix (Vazyme, no. Q111–02). The relative expression of RNA was calculated by the 2^−ΔΔCt method, and GAPDH was used as a normalization control. Primer sequences are shown in [270]Supplementary Table 5. DEG analysis in Gene Expression Omnibus datasets Transcriptome sequencing datasets in this study were collected from the Gene Expression Omnibus (GEO) ([271]https://www.ncbi.nlm.nih.gov/gds/) database and analyzed using the online software GEO2R ([272]https://www.ncbi.nlm.nih.gov/geo/geo2r/). The GEO datasets used in this study included [273]GSE41571, [274]GSE163154, [275]GSE20739, [276]GSE87534 and [277]GSE1176531. Co-immunoprecipitation Whole cell lysates were prepared with lysis buffer (50 mM Tris-HCl (pH 7.4), 140 mM NaCl, 5% glycerol and 1% Triton X-100) containing protease inhibitors (MCI, no. HY-K0010) and phosphatase inhibitors (Yeasen, no. 20109ES05) for 1 h on ice, and proteins were incubated with 2 μg of specific antibodies in continuous rotation at 4 °C overnight. Then, 30 μl of Protein A/G Magnetic (MCE, no. HY-K0202) was added and incubated for 2 h with constant rotation speed at 4 °C. The beads were adsorbed on a magnetic stand and washed three times with wash buffer (50 mM Tris-HCl (pH 7.4), 140 mM NaCl and 0.1% Triton X-100). After the last wash, the supernatant was aspirated and discarded, and precipitated proteins were subsequently resuspended in sample buffer and boiled at 95 °C for 10 min. The material obtained by immunoprecipitation of cell lysates was subjected to western blot analysis. SPR assay The binding affinity was measured by SPR using a BIAcore T200 (GE Healthcare) instrument. Recombinant human IGFBP6 (Sino Bio, no. 13026-H08H) was diluted in sodium acetate solution (pH 5.0) with a final concentration of 20 μg ml^−1. IGFBP6 was immobilized on a CM5 sensor chip (GE Healthcare) by amine coupling to reach appropriate 1,036 resonance units (RU) density. The running buffer contained a 1× HBS buffer. Instrument settings remained at 25 °C. Then, 10 concentrations of the recombinant human MVP (Abnova, no. H00009961-P01) (0 μg ml^−1, 0.01953125 μg ml^−1, 0.0390625 μg ml^−1, 0.078125 μg ml^−1, 0.15625 μg ml^−1, 0.3125 μg ml^−1, 0.625 μg ml^−1, 1.25 μg ml^−1, 2.5 μg ml^−1 and 5 μg ml^−1) were injected at a flow rate of 30 μl min^−1. The contact time was 2 min for IGFBP6 and MVP; the dissociation time was 2 min for IGFBP6 and MVP. A blank immobilization was performed for one of the sensor chip surfaces to use for correction of the binding response. Sensorgrams were analyzed using Igor Pro version 6.1 (WaveMatrix). Western blotting Whole lysates isolated from cell and tissue samples were lyzed with RIPA buffer supplemented with protease inhibitors (MCE, no. HY-K0010) and phosphatase inhibitors (Yeasen, no. 20109ES05) and boiled at 95 °C for 10 min. Samples were separated by SDS–PAGE, transferred to nitrocellulose membranes (Pall) and subsequently incubated at room temperature with blocking buffer (LI-COR) for 1 h. After blocking, the membranes were incubated with the primary antibody at 4 °C overnight. The membranes were then washed with Tris-buffered saline (TBS) containing 0.1% Tween 20 three times for 10 min. The membrane was subsequently incubated with IRDye 680RD goat anti-mouse IgG (no. 925–68070) or IRDye 800CW goat anti-rabbit IgG (no. 926–32211) (LI-COR; 1:15,000 dilution) for 45 min at room temperature. Finally, the blots were visualized using an Odyssey CLx Infrared Imaging System (LI-COR). Immunofluorescence HUVECs cultured in 35-mm glass-bottom dishes were fixed with 4% PFA for 20 min and then washed three times with PBS. The cells were permeabilized with PBS containing 0.1% Triton X-100 for 20 min to facilitate membrane penetration. Afterwards, the cells were blocked with PBS containing 10% goat serum for 1.5 h, followed by incubation with rabbit anti-IGFBP6 and mouse anti-MVP primary antibodies (1:100) overnight at 4 °C. The cells were then washed again and incubated at room temperature for 1 h with secondary antibodies (Alexa Fluor goat anti-rabbit 546 (no. [278]A48254) and Alexa Fluor goat anti-mouse/rat 488 (no. A48255), 1:1,000). After washing with PBS, DAPI staining solution (Beyotime Biotechnology) was added for 0.5 h. A laser scanning confocal microscope (Leica, TCS SP8 X) was used to capture the images. Immunofluorescence quantification was performed using ImageJ software. For immunofluorescence staining of mouse vascular tissue, after OCT removal with PBS for 10 min, the tissue was blocked with PBS containing 10% goat serum for 1.5 h, and subsequent steps and cell staining were consistent. Immunostaining of murine aortic roots and human coronary arteries with isotype control IgG was performed as negative control ([279]Extended Data Fig. 10a,[280]b). En face staining and RNA flushing from aortic endothelium After euthanasia, 12-week-old mice were perfused slowly for 5 min with saline containing 40 USPU ml^−1 heparin injected through the left ventricle, followed by 10 min with pre-cooled PBS containing 4% PFA (pH 7.4). Subsequently, the aorta was dissected from surrounding adipose tissues, opened longitudinally before incubation with PBS containing 0.1% Triton X-100 for 10 min and blocked with goat serum in TBS containing 2.5% Tween 20 for 1 h. Next, aortas were incubated with a 1:100 dilution of VE-cadherin and IGFBP6 antibodies in dilution buffer (TBS containing 3% BSA and 2.5% Tween 20) at 4 °C overnight. After three rinses for 5 min with washing solution (TBS containing 2.5% Tween 20), the aortas were incubated with Alexa Fluor 488 or 546 secondary antibodies (1:1,000 dilution) at room temperature for 1 h. The nuclei were stained with DAPI. Finally, after three more rinses in washing solution, the aorta was spread on a slide with the aortic endothelium facing the lens. A laser scanning confocal microscope (Leica, TCS SP8 X) was used for imaging. In mice, RNA was extracted from ECs collected from the AA or the TA. Specifically, mice were anesthetized and euthanized; the left ventricle was perfused with PBS; and the aorta was separated after removal of adipose tissue. After washing with PBS, the aorta (tunica intima) was rinsed with 300 μl of TRIzol in a 1-ml syringe. The remaining part of the adventitia of the blood vessel was then placed into TRIzol to collect RNA (tunica media). RNA was isolated using the TRIzol method. ELISA ELISA kits (Thermo Fisher Scientific, no. EMIGFBP6 and no. EHIGFBP6) were used to quantify IGFBP6 in mouse serum and cell supernatant as well as in human serum. IL-6 and IL-1β in human serum were quantified using kits (no. VAL102 and no. VAL101). ELISAs were carried out following the manufacturer’s protocol. In brief, samples were added to wells coated with antibodies and incubated at room temperature for 2 h. The solution was discarded, and the plate was washed four times. Detection antibody bound to the fixed target protein was added to each well and incubated at room temperature for 2 h. Excess detection antibody was washed off, followed by the addition of streptavidin-HRP and incubation at room temperature in the dark for 30 min. The plates were washed four times again and incubated for another 30 min at room temperature in the dark after adding substrate. Stop solution was then added, and the absorbance was read at 450 nm after thoroughly mixing the contents. IGFBP6 promoter cloning and dual-luciferase reporter assay To investigate the precise relationship between IGFBP6 and KLF2, several plasmids were used, including the empty plasmid (basic-luc), and the plasmid was generated containing the IGFBP6 promoter (2,000-bp length before the transcription start site) cloned upstream from firefly luciferase (WT-luc). A version of the IGFBP6 promoter reporter was produced with deletion mutations of all CACCC KLF2 consensus sites (MT1-luc), and a version was produced with deletion mutations of all GGGTG sites (MT2-luc). All reporter plasmids were produced by Genomeditech. Primers for reporter and mutant genes are provided in [281]Supplementary Table 4. HEK293T cells were transfected with a mixture of luciferase reporter plasmid and control plasmid containing basally regulated renilla luciferase, and the cells were transfected with Lipofectamine 2000 (Thermo Fisher Scientific, no. 11668019) and cultured for 1 d. Then, the cells were collected and lysed, and the Dual-Luciferase Reporter Assay Kit (Vazyme, no. DL101–01) was tested by a multifunctional microplate reader (Molecular Devices, SpectraMaxiD3). Luciferase activity was measured using SoftMax Pro 7.1 software. The relative luciferase activity was determined by calculating the ratio of firefly luciferase to renilla luciferase values. Chromatin immunoprecipitation The EZ-Magna ChIP A/G Chromatin Immunoprecipitation Kit (Sigma-Aldrich, 17–10086) was used to prepare chromatin from Ad-KLF2-treated or Ad-NC-treated HUVECs. In brief, cells were fixed with 1% formaldehyde for 10 min to crosslink DNA to proteins, and the reaction was quenched with glycine. Next, the nuclei were lysed with nuclear lysis buffer and passed through an ultrasonic cell grinder (Ningbo Scientz Biotechnology, SCIENTZ-IID) to cleave chromatin into small 200–1,000-bp fragments. Immunoprecipitation with Flag antibody and Protein A/G beads was then performed by rotating incubation at 4 °C overnight. The next day, the magnetic beads were washed using a magnetic rack, followed by elution of chromatin and reverse crosslinking (incubation at 62 °C for 2 h with shaking), followed by boiling at 95 °C for 10 min. Finally, the DNA was purified for PCR analysis. The IGFBP6 promoter was amplified by PCR using multiple primers, followed by nucleic acid gel electrophoresis. After gel extraction, the DNA was quantified by qPCR. Details of the primers used are provided in [282]Supplementary Table 5. Analysis of plaque composition To determine the extent of atherosclerotic plaques in mice, mouse hearts that had been perfused from the left ventricle with PBS were collected and fixed in 4% PFA for 1 d before dehydration in 30% sucrose for 1 d. Subsequently, OCT-embedded hearts were sliced horizontally along the aortic axis in the direction of the AA. H&E and Massonʼs trichrome staining were performed to determine the necrotic core area of the aortic root and plaque collagen expression. ImageJ software was applied to determine the necrotic core area and collagen expression of the plaque, and this area divided by the plaque area was used to calculate the plaque lesions. En face Oil Red O staining Mice were euthanized and aorta were isolated. Aorta were cut open in PBS under the dissection microscope to expose atherosclerotic plaques. After fixation with 4% PFA for 1 d, the mixture was removed and washed with PBS for 5 min. The mixture was mixed with 60% isopropanol (Sangon Biotech, no. 67–63-0), soaked for a short time and stained with freshly prepared filtered Oil Red O (Poly Scientific, no. s1849–32OZ) at 37 °C for 15 min. The solution was discarded and then washed with PBS after washing with 60% isopropanol. The en face aorta was mounted onto tissue slides with the intimal layer facing up. An MShot Image Analysis System (Mingmei Photoelectric Technology) recorded images. The plaque area was determined using ImageJ software, and the percentage of plaque area was calculated by the ratio of plaque area to total vessel area. Conditioned medium preparation Conditioned media from ECs infected with Ad-IGFBP6 were collected. Media were concentrated using Macrosep Advance (Pall, no. MAP010C37). Co-immunoprecipitation was performed using the concentrated media. Liquid chromatography with tandem mass spectrometry analysis Liquid chromatography with tandem mass spectrometry (LC–MS/MS) analysis was conducted using a timsTOF Pro mass spectrometer (Bruker) coupled with a nanoElute (Bruker). The peptides were loaded onto a C18 reversed-phase analytical column (Thermo Fisher Scientific, Easy Column, 25 cm long, 75 μm inner diameter, 1.9 μm resin) in 95% buffer A (0.1% formic acid in water) and separated using a linear gradient of buffer B (99.9% acetonitrile and 0.1% formic acid) at a flow rate of 300 nl min^−1. The mass spectrometer was operated in positive ion mode with an electrospray voltage of 1.5 kV. Precursors and fragments were analyzed using the TOF detector within a mass range of m/z 100–1,700. The timsTOF Pro was operated in parallel accumulation serial fragmentation (PASEF) mode, with PASEF mode data collection under the following conditions: ion mobility coefficient (1/KO) value was set from 0.6 Vs cm^−2 to 1.6 Vs cm^−2; one MS and 10 MS/MS PASEF scans were performed. Active exclusion was enabled with a release time of 24 s. Molecular docking prediction In this study, the three-dimensionl structures of both IGFBP6 and MVP proteins were derived from predictive models generated by the AlphaFold 3 online tool ([283]https://alphafoldserver.com/). Protein–protein molecular docking was performed using the online tool HDOCK SERVER, with detailed parameters referring to the provided example tutorials^[284]51. The Protein Interfaces, Surfaces and Assemblies (PISA) service, available at the European Bioinformatics Institute, can be accessed via the following link: [285]http://www.ebi.ac.uk/pdbe/prot_int/pistart.html. The final results were analyzed and visualized using PyMOL version 2.6 software. Statistical analysis Data are presented as the mean ± s.d. The Shapiro–Wilk normality test was used to assess the distribution of the data. For comparisons between two groups with normally distributed data, Student’s t-test was applied when variances were equal, whereas Welch’s correction was used if the F-test indicated unequal standard deviations. For the experiments using HUVECs/HAECs from different donors, the two-sided paired t-test was adopted because of the large differences among different donors. Comparisons among three or more groups were conducted using one-way or two-way ANOVA followed by Tukey’s or Bonferroni’s multiple comparison test to evaluate differences among multiple groups. For data that were not normally distributed, non-parametric tests were used. The Mann–Whitney test was used for comparisons between two independent groups, and the Kruskal–Wallis test and Dunn’s multiple comparison test were used for comparisons among multiple groups. For experiments involving HUVECs/HAECs with different donors, one-way or two-way ANOVA (repeated measures) followed by Bonferroni’s test was used. Representative images were chosen to represent the group mean most accurately of all available data. P values less than 0.05 were considered statistically significant. Statistical analyses were performed using GraphPad Prism 10.0 (GraphPad Software) and SPSS version 27.0. Extended Data Extended Data Fig. 1 |. The gene expression values of IGFs and IGFBPs. Extended Data Fig. 1 | [286]Open in a new tab a, The mRNA expression counts of IGFs and IGFBPs in UF- and DF-treated HUVECs. (GEO accession No. [287]GSE20739, n = 3). b, A heatmap showing the differentially expressed genes (DEGs) between the two groups. Upregulated genes are labelled in red and downregulated genes are shown in blue. c, The mRNA expression of IGFBP1-7 in HUVECs under static conditions (n = 3). Statistical analysis was performed by two-tailed Welch’s t test for a, by one-way ANOVA followed by the Tukey’s test for c. Extended Data Fig. 2 |. Construction and genotype identification of IGFBP6 global knockout mice. Extended Data Fig. 2 | [288]Open in a new tab a, Identification of genotypes of Igfbp6^−/− mice. b, The LDLR protein level in liver tissues of mice after PCSK9 injection as detected by Western blot. Liver tissues from male C57BL/6 J and Ldlr^−/− mice were used as controls. c, Analysis of ALT, AST, TG, TC, HDL-C, and LDL-C levels in male WT and Igfbp6^−/− mice (n = 13–15). Statistical analysis was performed by two-tailed Student’s t test, Mann–Whitney U test and Welch’s t test for c. Extended Data Fig. 3 |. Construction of EC-specific IGFBP6 knockout mice, genotype identification and serum biochemistry. Extended Data Fig. 3 | [289]Open in a new tab a, Schematic view of the generation of Igfbp6^ECKO mice. b, Identification of genotypes of Igfbp6^ECKO mice. c, The mRNA expression of Igfbps in the aortic intima of male Igfbp6^ECKO mice and control mice (n = 5). d, Analysis of body weight, blood glucose, ALT, AST, TG, TC, HDL-C, and LDL-C levels (n = 7–11). Statistical analysis was performed by two-tailed Student’s t test and Mann–Whitney U test for c, d, and by Welch’s t test for d (Male: LDL-C). Extended Data Fig. 4 |. Validation and serum biochemistry of EC-specific IGFBP6 overexpression mice. Extended Data Fig. 4 | [290]Open in a new tab a, En face immunofluorescence staining IGFBP6 (red), VE-Cadherin (green), and DAPI (blue) in mouse aorta, showing increased IGFBP6 expression in the aortic endothelium of male AAV9-EC-Igfbp6 mice (n = 3). Scale bar: 50 μm. b, The mRNA expression of Igfbps in the aortic intima of male AAV9-EC-Igfbp6 mice and control mice (n = 5). c, Analysis of body weight, blood glucose, ALT, AST, TG, TC, HDL-C, and LDL-C levels in mouse serum (n = 10). Statistical analysis was performed by two-tailed Mann–Whitney U test and by Student’s t test for b, c, and by Welch’s t test for c (ALT). Extended Data Fig. 5 |. IGFBP6 suppresses inflammation in HAECs and RNA-sequencing of IGFBP6 overexpressed ECs indicates the anti-inflammatory effect of IGFBP6. Extended Data Fig. 5 | [291]Open in a new tab a and b, HAECs were transfected with control si-NC or si-IGFBP6 for 48 h and then treated with TNF-α for 6 h. The expression of IGFBP6, VCAM-1, ICAM-1 was determined by qRT-PCR (a, n = 3) and Western blot (b, n = 4). c and d, HAECs were transfected with Ad-NC or Ad-IGFBP6 for 24 h and then treated with TNF-α for 6 h. The expression of Flag-IGFBP6, VCAM-1, ICAM-1, SELE and CCL2 was determined by qRT-PCR (c) and Western blot (d) (n = 3). e and f, HAECs were treated as indicated and THP-1 monocyte adhesion assay was performed and the number of adherent monocytes were quantified (n = 3). Scale bar: 50 μm. g, HUVECs were transfected with Ad-NC or Ad-IGFBP6 for 24 h and then treated with TNF-α for 6 h. The treated cells were analyzed by transcriptome sequencing (n = 3). h, Bubble diagram of the KEGG pathway enrichment analysis of DEGs. i, Heatmap diagram showing the DEGs between the two groups. Upregulated genes are labelled in red and downregulated genes are shown in blue. Scale bar: 50 μm. Statistical analysis was performed by two-tailed paired t test for a (left panel), b, d, by two-way ANOVA (repeated measures) followed by the Bonferroni’s test for a (right panel), c, by Student’s t test for e, f. Extended Data Fig. 6 |. KLF4 up-regulates the expression of IGFBP6 and MVP depletion promotes inflammation in HUVECs. Extended Data Fig. 6 | [292]Open in a new tab a and b, HUVECs were treated with Ad-NC or Ad-KLF4 for 24 h, and the expression of KLF4, NOS3 and IGFBP6 was determined by qRT-PCR (a, n = 5), and ELISA (b, n = 3). c, HUVECs were transfected with si-NC or si-MVP for 48 h and then treated with TNF-α for 6 h. The expression of MVP, ICAM-1and SELE was determined by qRT-PCR (n = 3). d, HUVECs were treated as indicated and THP-1 monocyte adhesion assay was performed and the number of adherent monocytes were quantified (n = 3). Scale bar: 50 μm. Statistical analysis was performed by two-tailed paired t test for a, by Student’s t test for b, by two-way ANOVA (repeated measures) followed by the Bonferroni’s test for c, and by two-way ANOVA followed by the Tukey’s test for d. Extended Data Fig. 7 |. The anti-inflammatory effect of IGFBP6 depends on the binding site of IGFBP6 to MVP. Extended Data Fig. 7 | [293]Open in a new tab a, In HUVECs, Co-IP experiments were performed on MVP and ASK1 (n = 3). b, HUVECs were treated with adenovirus to overexpress IGFBP6. After 42 h of treatment, TNF-α was added for 6 h. After that, flag-tagged IGFBP6 was pulled down and ASK1 (n = 3) was detected. c, Representative Co-IP and Western blot assays revealed dimerization of ASK1 in HEK293T cells transfected with indicated plasmid vectors (n = 3). d, Molecular docking was performed using Alphafold3 to predict the binding sites of IGFBP6 and MVP. e, Representative Co-IP and Western blot assays showing the binding domains of IGFBP6 to MVP in HEK293T cells (n = 3). f, HUVECs were treated with IGFBP6 and mutated IGFBP6 adenovirus for 42 h, followed by TNF-α for 6 h. The expression levels of Flag, VCAM-1, ICAM-1 were detected by Western blot (n = 3). Extended Data Fig. 8 |. Secreted IGFBP6 and MVP do not affect inflammation in HUVECs. Extended Data Fig. 8 | [294]Open in a new tab a and b, HUVECs were treated with adenoviruses (Ad-MVP and Ad-IGFBP6) for 7 days, during which the supernatant was continuously collected. Ultrafiltration tubes were used to concentrate the protein in the supernatant, and flag (a, n = 3) or HA (b, n = 3) antibodies were used for Co-IP, respectively, and finally verified by Western blot. c, HUVECs were treated with different concentrations of IGFBP6 recombinant protein for 42 h, and then treated with TNF-α for 6 h, and indicated proteins were detected by Western blot (n = 3). d, HUVECs were treated with different concentrations of IGFBP6 recombinant protein for 3 h, and then treated with TNF-α for 10 mins, and indicated proteins were detected by Western blot (n = 3). e, 80% of confluent HUVECs were treated with Ad-NC or Ad-IGFBP6 in the presence of TNF. After that, whole cell lysate was collected for Western blot (left 4 lanes, control). In parallel experiments, the treated cells were washed with PBS and new complete medium was supplemented. Then, conditioned media were collected to treat HUVECs before lysate was collected for Western blot (right 4 lanes) (n = 3). f, HUVECs were treated with different concentrations of MVP recombinant protein for 3 h, and then treated with TNF-α for 10 mins, and indicated proteins were detected by Western blot (n = 3). Extended Data Fig. 9 |. Graphical Abstract. Extended Data Fig. 9 | [295]Open in a new tab IGFBP6 serves as a novel endothelial homeostasis-associated molecule which is mechanoresponsive and confers anti-inflammation and atheroprotection via binding with intracellular MVP and suppresses JNK and p65 phosphorylation. Extended Data Fig. 10 |. Immunostaining of murine aortic roots and human coronary arteries with isotype control IgG. Extended Data Fig. 10 | [296]Open in a new tab a and b, Representative images of immunofluorescence staining with rat IgG, rabbit IgG or mouse IgG for mouse and human plaque tissues (n = 3). Scale bar = 50 μm. Supplementary Material Supplemental Data [297]NIHMS2048285-supplement-Supplemental_Data.pdf^ (298.5KB, pdf) Acknowledgements