Abstract Calcification of autologous pathological vessels and tissue engineering blood vessels (TEBVs) is a thorny problem in clinic. However, there is no effective and noninvasive treatment that is available against the calcification of TEBVs and autologous pathological vessels. Gli1^+ cells are progenitors of smooth muscle cells (SMCs) and can differentiate into osteoblast-like cells, leading to vascular calcification. Our results showed that the spatiotemporal distribution of Gli1^+ cells in TEBVs was positively correlated with the degree of TEBV calcification. An anticalcification approach was designed consisting of exosomes derived from mesenchymal stem cells delivering lncRNA-ANCR to construct the engineered exosome-Ancr/E7-EXO. The results showed that Ancr/E7-EXO effectively targeted Gli1^+ cells, promoting rapid SMC reconstruction and markedly inhibiting Gli1^+ cell differentiation into osteoblast-like cells. Moreover, Ancr/E7-EXO significantly inhibited vascular calcification caused by chronic kidney disease. Therefore, Ancr/E7-EXO reprogrammed Gli1^+ cells to prevent calcification of vascular graft and autologous pathological vessel, providing unique insights for an effective anticalcification. __________________________________________________________________ Engineered exosomes reprogrammed Gli1^+ cells to inhibit calcification in TEVBs and autologous vessels. INTRODUCTION The morbidity and mortality of cardiovascular diseases are increasing year by year worldwide ([50]1). Vascular calcification, as an independent risk factor of cardiovascular death, is an important predictor of cardiovascular disease incidence and mortality, which often leads to vascular wall sclerosis, lumen stenosis, hemodynamic damage, and, lastly, circulatory obstruction ([51]2). Vascular calcification is the most common complication and common pathological change of diabetes, chronic kidney disease (CKD), and atherosclerosis ([52]3), because the related metabolic and inflammatory factors of chronic disease can accelerate vascular calcification ([53]4). For the early vascular calcification in vivo, there is no effective reversal intervention in clinic ([54]5). With the aggravation of vascular calcification, patients often need to undergo vascular replacement, and tissue engineering blood vessels (TEBVs) are a promising biological vascular substitute ([55]6). However, when TEBVs are implanted into patients with underlying diseases such as diabetes, calcification is also easy to occur in grafts and leads to implantation failure ([56]7). For TEBVs, on the one hand, it is necessary to promote the rapid and normal reconstruction of SMCs to resist the occurrence of aneurysms ([57]8); on the other hand, it is necessary to inhibit calcification to maintain long-term homeostasis and vascular patency ([58]3, [59]9). At present, there is no noninvasive treatment for vascular calcification under the above pathological conditions. Therefore, exploring effective strategies to inhibit calcification is substantial to autologous vascular calcification caused by chronic diseases and vascular grafts. More and more studies have shown that endogenous vascular progenitor cells play a role in the occurrence and development of vascular remodeling diseases under pathological conditions ([60]3). They may serve as repositories of primitive progenitor cells and may be involved in vascular calcification and chondrogenic metaplasia in vessels ([61]10). Gli1^+ cells have mesenchymal stem cell (MSC) morphology and typical surface markers, have the ability of three-lineage differentiation, form an extensive perivascular network in tissue, and exist in adventitia of vessels ([62]11, [63]12). Studies have shown that Gli1^+ cells promote the formation of regenerative type H vessels for tissue regeneration and repair ([64]13). Recent studies have shown that Gli1^+ cells are the progenitors of vascular smooth muscle cells (VSMCs) and differentiate into highly differentiated SMCs in acute vascular injury, while, in chronic vascular injury diseases, Gli1^+ cells can differentiate into osteoblast-like cells, which may cause vascular calcification ([65]14). Therefore, targeting Gli1^+ cells and reprogramming in vivo may be an interesting way to treat vascular calcification. Exosomes are extracellular vesicles with a diameter of 30 to 100 nm that can carry RNA and affect cellular function by transmitting information between cells and regulating intercellular communication ([66]15). Exosomes derived from MSCs promote vascular repair after injury by secreting cytokines and inducing cell differentiation and are widely used in cardiovascular diseases ([67]16, [68]17). At the same time, exosomes are very promising nanovesicles in regenerative medicine by carrying drugs or nucleic acids for the regulation of tissue injury repair and regeneration, showing safety and effectiveness in vivo ([69]15). LncRNA-ANCR has shown the ability to inhibit osteoblast differentiation in previous studies. During the differentiation of human osteoblasts, the expression of lncRNA-ANCR decreased significantly, while its expression inhibits the differentiation of human osteoblasts by inhibiting Runt-related transcription factor2 (Runx2) expression through the recruitment of the enhancer EZH2 ([70]18). LncRNA-ANCR silencing can promote osteogenesis of postmenopausal osteoblasts both in vitro and in vivo ([71]19). Inspired by the potential of exosomes to load and deliver signal molecules, we hope to combine exosome therapy with long noncoding RNA (lncRNA) therapy through engineering method and regulate the differentiation trend of Gli1^+ cells by loading lncRNA-ANCR and targeting Gli1^+ cells. The surface modification of exosomes can achieve targeting in vivo. Previous studies have shown that E7 peptide has a special affinity for MSCs and can promote MSC homing ([72]20). Therefore, in this study, we designed and constructed Lamp2b-E7 plasmid, transfected it into human bone marrow MSCs (HuBM-MSCs), and introduced lncRNA-ANCR into HuBM-MSC–derived E7-EXO by electroporation to construct an engineering exosome-Ancr/E7-EXO. Through tail vein injection, Ancr/E7-EXO can recruit the site of vascular calcification in CKD rats and then inhibit the osteogenic differentiation of Gli1^+ cells. The modification of Ancr/E7-EXO on TEBVs by electrostatic self-assembly can regulate the differentiation of Gli1^+ cells into contractile SMCs in a high-glucose environment, promote the reconstruction of SMCs in TEBVs, and effectively inhibit calcification in vivo. The scheme of engineered exosome-Ancr/E7-EXO reprogramed Gli1^+ cells to prevent calcification of vascular grafts and autologous pathological vessels in vivo. Construction of the engineered exosomes was loaded with ANCR and surface-modified E7 peptide Ancr/E7-EXO ([73]Fig. 1A). Ancr/E7-EXO was modified to TEBVs by self-assembly, regulating the differentiation of Gli1^+ cells into contractile SMCs and inhibiting vascular grafts calcification in vivo ([74]Fig. 1B). Ancr/E7-EXO injected through the tail vein of CKD rats accumulated in the calcification sites due to Gli1^+ cells, reversed the osteogenic differentiation of Gli1^+ cells, and inhibited autologous vascular calcification ([75]Fig. 1C). Fig. 1. Scheme of engineered exosome-Ancr/E7-EXO reprogramming Gli1^+ cells to prevent calcification of vascular grafts and autologous pathological vessels in vivo. [76]Fig. 1. [77]Open in a new tab (A) Construction of the engineered exosomes loaded with ANCR and surface-modified E7 peptide Ancr/E7-EXO. (B) Ancr/E7-EXO was modified to TEBVs by self-assembly, regulating the differentiation of Gli1^+ cells into contractile SMCs and inhibiting vascular grafts calcification in vivo. (C) Ancr/E7-EXO injected through the tail vein of CKD rats accumulated in the calcification sites due to Gli1^+ cells, reversed the osteogenic differentiation of Gli1^+ cells, and inhibited autologous vascular calcification. RESULTS Tracing of the spatiotemporal distribution of Gli1^+ cells in TEBVs and autologous pathological vessels TEBVs are prone to calcification after implantation, which is accelerated in high-glucose conditions. Therefore, Alizarin red and von Kossa staining of TEBVs were performed at different times after TEBV implantation in normal and diabetic rats. The staining revealed that the calcified nodules were present in two groups, and a full-layer calcification appeared in diabetic rats, with severe lumen stenosis at the sixth weeks (fig. S1, A and B). These results suggested that TEBVs were prone to calcification in both normal and pathological conditions. Gli1^+ cells exist in the adventitia of artery and are considered to be the progenitor cells of VSMCs, but it is not clear whether they can dedifferentiate into osteoblasts under pathological conditions in TEBVs. Therefore, we tracked the spatial distribution of Gli1^+ cells in TEBVs at each time point after implantation. Gli1^+ cells in TEBVs at each time point after implantation increased with time, but the diabetes group had more Gli1^+ cells than normal group ([78]Fig. 2A). The total number of Gli1^+ cells in the diabetic group at 14 days was approximately 2.65-fold than that in the normal group ([79]Fig. 2B). The amount of Gli1^+ cells in different spatial locations still showed a similar trend ([80]Fig. 2, C to E). The number of Gli1^+ cells in the media of TEBVs in the diabetic group was 1.38 times of that in the normal group ([81]Fig. 2D). Gli1^+ cells in the intima of TEBVs in the diabetic group were about fourfold than that in the normal group. We also observed that there were more Gli1^+ and osteopontin (OPN) signal coexpression (fig. S2A) in the adventitia of TEBVs at the early stage of implantation, and the coexpression signal increased with the development of diabetes (fig. S2, B and C). Therefore, the above results showed the spatiotemporal distribution of Gli1^+ cells in TEBVs under normal physiological and pathological conditions, which was positively correlated with the degree of TEBV calcification, suggesting that they might be the main cellular source of calcification in TEBVs, as confirmed. In addition, the role of Gli1^+ cells was also evaluated in the model of chronic vascular calcification caused by CKD. The development of CKD due to the loss of kidney tissue was monitored by detecting the increased level of blood urea nitrogen (BUN) ([82]Fig. 2H). Fluorescent reflection imaging (FRI) showed that hydroxyapatite formation was detected in aortic and thoracic aorta ([83]Fig. 2F); the fluorescence intensity in the CKD group was 2.14 times higher than that in the sham group ([84]Fig. 2G). The cross-sectional images of the aorta showed that the amount of Gli1^+ cells in the CKD group was eightfold than that in the sham group ([85]Fig. 2, I and J). A positive correlation between the fluorescence intensity of hydroxyapatite and Gli1^+ cells was found. These results suggested that Gli1^+ cells might be the main cellular source of calcification in chronic vascular injury diseases in vivo. Fig. 2. Spatiotemporal distribution of Gli1^+ cells in TEBVs and CKD-induced calcified vessels. [86]Fig. 2. [87]Open in a new tab (A) Immunofluorescence staining in the cross-sectional area of TEBVs in each group after TEBV implantation [4′,6-diamidino-2-phenylindole (DAPI), blue; Gli1^+, red]. The lines divide the vascular adventitia (A), media (M), and intima (I). Scale bars, 100 μm. Higher magnification of the yellow line border. Scale bars, 20 μm. (B to E) Number of total Gli1^+ cells, as well as intima, media, and adventitia Gli1^+cells in TEBVs (n = 3). (F and G) Representative images and quantification of hydroxyapatite fluorescence intensity of the aortic segment in CKD and sham group (n = 3) at 13 weeks after subtotal nephrectomy (5/6Nx). (H) Blood urea nitrogen (BUN) stratified for the sham and CDK group. (I and J) Representative image and quantification of CKD-induced calcified vessels in the cross-sectional area of the aorta in rats by immunofluorescence staining (DAPI, blue; Gli1^+, red). Results are expressed as means ± SD by two-tailed Student’s t test. *P < 0.05, **P < 0.01, ***P <0.001, and ****P <0.0001. wk, weeks. Construction and characterization of Ancr/E7-EXO targeting Gli1^+ cells The exosomes from HuBM-MSCs were isolated by ultracentrifugation, called MSC-EXO, to obtain engineered exosomes with the ability to target Gli1^+ cells. They were characterized by typical saucer-like structure, surface markers, and particle size (fig. S3, A to C). MSC-EXO was modified with the following schematic modification ([88]Fig. 3A). First, the MSC affinity peptide-E7 was conjugated with lamp-2b to avoid being cut off and to increase its expression on the exosome membrane after E7 plasmid transfected into HuBM-MSCs. An amino acid sequence (MLKPLEQ) was also designed with the same amino acid as E7 but in a scrambled order, called M7 peptide, and used as a negative control for E7. After the E7 and M7 plasmid were transfected HuBM-MSCs, Western blot indicated that the expression of Lamp2b on the membrane of E7-EXO and M7-EXO and in HuBM-MSCs was increased ([89]Fig. 3C). Real-time polymerase chain reaction (PCR) also suggested that E7 plasmid was successfully transfected into HuBM-MSCs and expressed ([90]Fig. 3D). Scanning electron microscopy (SEM) image showed a typical oval structure of E7-EXO ([91]Fig. 3B). Next, MSC-EXO, E7-EXO, and M7-EXO were labeled with PKH67 and incubated with Gli1^+ cells at different times to determine the affinity of E7-EXO for Gli1^+ cells. The results revealed that E7-EXO was efficiently internalized into Gli1^+ cells (fig. S3, D and E). After carotid artery replacement, TEBVs come into direct contact with blood, which induces neutrophil recruitment and subsequently recruits proinflammatory macrophage infiltration. Although the E7-EXO has the affinity of Gli1^+ cells, it is impossible to predict its ability to deal with inflammatory cells, so we carried out coculture experiments ([92]Fig. 3I). The positive rate of E7-EXO phagocytosis by Gli1^+ cells was approximately 3.43 times higher than that of macrophages ([93]Fig. 3, I to K). In addition, we also cocultured Gli1^+ cells with endothelial cells (ECs), SMCs, and neutrophils (fig. S3, F to K). The above results demonstrated that E7-EXO had a specific affinity with Gli1^+ cells. The ANCR plasmid was loaded into E7-EXO through electroporation at 100 V and 50 μF, making the ANCR plasmid retained to the maximum extent. The E7-EXO after successful electroporation was called Ancr/E7-EXO. The loading efficacy was tested by quantitative PCR (qPCR), and the results showed that the Ancr/E7-EXO loaded 2.95 times more ANCR than MSC-EXO ([94]Fig. 3F). Approximately 1% of ANCR plasmids were loaded into Ancr/E7-EXO, meaning that each Ancr/E7-EXO loaded 100 ANCR plasmids. The SEM images and nanoparticle size of Ancr/E7-EXO were similar to those of MSC-EXO ([95]Fig. 3, E, G, and H). These results demonstrated that the electroporation did not change the physical properties and stability of E7-EXO, proving the successful construction of the Ancr/E7-EXO targeting Gli1^+ cells. Fig. 3. Synthesis and characterization of Ancr/E7-EXO. [96]Fig. 3. [97]Open in a new tab (A) Scheme representing Ancr/E7-EXO construction. (B) Scanning electron microscopy (SEM) image of E7-EXO. Scale bar, 100 nm. (C) Western blot analysis of CD63 and Lamp2b of transfected HuBM-MSCs and HuBM-MSC–derived exosomes. GAPDH, glyceraldehyde-3-phosphate dehydrogenase. (D) The quantitative expression of Lamp2b-E7 in transfected HuBM-MSCs was compared with that in HuBM-MSCs. (E) SEM images of Ancr/E7-EXO. Scale bar: 100 nm. (F) qPCR showing ANCR expression in the Ancr/E7-EXO. (G and H) Zeta potential (G) and particle size (H) of Ancr/E7-EXO and MSC-EXO. (I to K) Immunofluorescence staining and quantification of the phagocytic positive rate in the coculture experiment (n = 5). Results are expressed as means ± SD by two-tailed Student’s t test. **P < 0.01 and ****P < 0.0001. The promotion of Ancr/E7-EXO on the differentiation of Gli1^+ cells into contractile SMCs in high-glucose conditions On the basis of previous studies ([98]21), MSCs were isolated from rat bone marrow. The culture of MSCs revealed that they had osteogenic, adipogenic, and chondrogenic differentiation abilities (fig. S4A). Gli1^+ cell populations were positive for CD44, CD29, Sca1^+, and negative for CD34 and CD45 and were screened by flow cytometry from MSCs ([99]Fig. 4A and fig. S4B). We cultured the isolated Gli1^+ cells and carried out the follow-up experiments. Immunofluorescence staining showed that Gli1^+ protein was located in the cytoplasm and nucleus (fig. S4C). Further in vitro cell culture revealed that Gli1^+ cells were able to differentiate into three phenotypes of SMCs (fig. S4D). Besides, the number of Gli1^+ cells, which were extracted from the bone marrow of diabetic rats, increased in a time-dependent manner (fig. S4, E and F). The number of Gli1^+ cells in diabetic rats’ bone marrow at day 60 was 3.35 times higher than that in healthy rats. Wound-healing assay showed that Ancr/E7-EXO markedly increased Gli1^+ cell migration ([100]Fig. 4, B and C). The dose of 200 μg of Ancr/E7-EXO was best in promoting the differentiation of Gli1^+ cells into contractile SMCs and in inhibiting the synthetic SMCs induced by high glucose (fig. S5, A to C). It is generally believed that MYH11 and thrombospondin are both the differentiation markers of SMCs, while MYH11 represents the contractile phenotype of SMCs and thrombospondin represents the synthesis phenotype of SMCs ([101]22, [102]23). In high-glucose medium, both MSC-EXO and Ancr/E7-EXO increased the differentiation of Gli1^+ cells into contractile SMCs [Myosin heavy chain 11 (MYH11) and smooth muscle actin alpha (α-SMA)], but Ancr/E7-EXO showed a stronger ability obviously ([103]Fig. 4, D to F). The expression of MYH11 in the Ancr/E7-EXO group was 1.07 times higher than that in the MSC-EXO group ([104]Fig. 4E). The synthetic SMCs (thrombospondin) were observed in the high-glucose and MSC-EXO groups, while no thrombospondin was observed in the Ancr/E7-EXO group ([105]Fig. 4D). In addition, Ancr/E7-EXO notably reduced the expression of thrombospondin compared with the effect of MSC-EXO ([106]Fig. 4G). The qPCR results showed that the expression of MYH11 in the Ancr/E7-EXO group was 1.48 times higher than that in the high-glucose group, while the expression of thrombospondin decreased by 29% ([107]Fig. 4, H and J). Furthermore, the expression of MYH11 in the Ancr/E7-EXO group was 1.32 times higher than that in the MSC-EXO group, while the expression of thrombospondin decreased by 22% ([108]Fig. 4, H and J). The expression trend of α-SMA was similar to that of MYH11 ([109]Fig. 4I). Hence, Ancr/E7-EXO also had a remarkable effect on promoting Gli1^+ cell differentiation into contractile SMCs compared with the effect of MSC-EXO. Western blot was used to further clarify the effect of ANCR plasmid ([110]Fig. 4K). The transfection of ANCR plasmid into Gli1^+ cells did not affect the expression of Gli1^+, while α-SMA increased by 2.25 times and thrombospondin decreased by 45% ([111]Fig. 4, L to N). Therefore, the above results proved that Ancr/E7-EXO promoted the differentiation of Gli1^+ cells into contractile SMCs and inhibited the differentiation into synthetic SMCs in high-glucose conditions. Fig. 4. The effect of Ancr/E7-EXO on SMC phenotype. [112]Fig. 4. [113]Open in a new tab (A) Gli1^+ cells were isolated from rat bone marrow MSCs by fluorescence-activated cell sorting. (B and C) Ancr/E7-EXO–induced Gli1^+ cell migration was evaluated using wound-healing assay. Scale bar, 500 μm. (D) Immunofluorescence staining was used to assess the effect of 200 μg of Ancr/E7-EXO on the differentiation of Gli1^+ cells in high-glucose medium at day 7: MYH11 (red) and α-SMA (green) represented contractile SMCs, and thrombospondin (red) represented synthetic SMCs. (E to G) Quantification of the immunostaining intensity in each group (n = 3). (H to J) Phenotypic changes of Gli1^+ cells under different conditions quantitatively analyzed by qPCR. (K to N) Western blot analysis was used to detect the differentiation and quantification of ANCR transfected Gli1^+ cells after 72 hours. Results are expressed as means ± SD by two-tailed Student’s t test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. ns, not significant. The rescue of Ancr/E7-EXO on the differentiation of Gli1^+ cells into osteoblast-like cells in high-glucose conditions High-glucose environment will lead MSCs to differentiate into osteoblast-like cells and whether this process can be inhibited by Ancr/E7-EXO. Ancr/E7-EXO (200 μg) was added to Gli1^+ cells in high-glucose medium, the number of calcified nodules was reduced at day 7, and the result was more obvious at day 14 (fig. S5, D to G). The potential ability of Ancr/E7-EXO to reverse the osteogenic differentiation of Gli1^+ cells was further evaluated by dividing Gli1^+ cells in high-glucose mediums into the control group, the osteogenic induction medium group (OI), the osteogenic induction medium + MSC-EXO group (OI + MSC-EXO), and the osteogenic induction medium + Ancr/E7-EXO group (OI + Ancr/E7-EXO). The Alizarin red staining suggested that Ancr/E7-EXO significantly reduced the mineralized nodules compared with their number in the control and OI groups at day 7 (fig. S6A). Moreover, the number of calcium nodules in OI + Ancr/E7-EXO was 0.85 times that of OI + MSC-EXO, indicating that Ancr/E7-EXO inhibited the formation of calcium nodules more efficiently than MSC-EXO (fig. S6B). The fluorescence intensity of Runx2 and OPN in the OI + Ancr/E7-EXO group was significantly lower than that in the control group (fig. S6, C to E). In addition, qPCR also showed that the expression of bone morphogenetic protein-2 (BMP-2), Runx2, and OPN in the OI + Ancr/E7-EXO group was significantly lower than that in the control group (fig. S6, F to H). The osteogenic induction culture was carried out for 14 days to better prove the effect of Ancr/E7-EXO. Alizarin red staining showed that the number of calcified nodules in the control group was 37.11 times higher than that in OI + Ancr/E7-EXO group ([114]Fig. 5, A and B). The fluorescence intensity of OPN in the OI + Ancr/E7-EXO group was markedly lower than that in other groups, which was only 36% of that in the control group, and Runx2 was only 45% of that in the control group ([115]Fig. 5, C to E). In addition, the expression of osteoblast-like cell markers in each group demonstrated that the inhibitory effect of the OI + Ancr/E7-EXO group was markedly higher compared to that of the control group ([116]Fig. 5, F to H). qPCR showed that the expression of OPN in the OI + Ancr/E7-EXO group was only 26% of that in the high-glucose group, while the expression of Runx2 was 34%. Besides, the expression of OPN in the OI + MSC-EXO group was 1.33 times higher than that in the OI + Ancr/E7-EXO group, while Runx2 was 1.63 times higher. After 7 and 14 days of osteogenic induction, Ancr/E7-EXO exhibited excellent osteogenic inhibition in vitro. Fig. 5. Preventive effect of Ancr/E7-EXO on the differentiation of Gli1^+ cells into osteoblast-like cells. [117]Fig. 5. [118]Open in a new tab (A and B) Osteogenic differentiation of Gli1^+ cells in each group detected by Alizarin red staining at day 14. The number of calcified nodules (red) was calculated and quantified by ImageJ. Scale bar, 250 pixel (px). (C to E) Gli1^+ cells were labeled with anti-Runx2 (red) and anti-OPN (red) antibodies to distinguish the osteogenic differentiation of each group, and the fluorescence signals of each group were quantified (n = 5). (F to H) qPCR of the markers of the osteogenic differentiation of Gli1^+ cells at day 14 under different conditions. Results are expressed as means ± SD by two-tailed Student’s t test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. Phagocytosis of Ancr/E7-EXO by Gli1^+ cells in vivo and enrichment of Ancr/E7-EXO to calcified sites in CKD Ancr/E7-EXO–modified TEBVs were used to evaluate the inhibitory effect of Ancr/E7-EXO on vascular calcification in vivo. 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC)/N-hydroxysuccinimide (NHS) can covalently cross-link the exosomes on TEBVs by amide reaction, thus improving the fixation strength. Polyethyleneimine/Au Nanopraticle (PEI/AuNP) was used as a known method to modify exosomes on TEBVs ([119]24). By comparison, we hope to modify more exosomes on the media and adventitia of TEBVs (fig. S7A). According to the results, EDC/NHS self-assembly strategy was chosen to cross-link as much Ancr/E7-EXO as possible on TEBVs to construct the Ancr/E7-EXO–modified TEBVs ([120]Fig. 6A). Ancr/E7-EXO was labeled by PKH67, a large number of Ancr/E7-EXO cross-linking on the media and adventitia of TEBVs compared with that in the acellular vessels (without considering the autofluorescence of vessels) ([121]Fig. 6B). This result suggested that this simple self-assembly strategy successfully modified Ancr/E7-EXO on TEBVs. Then, the Ancr/E7-EXO on TEBVs was labeled with DiD and then implanted into rats. The colocalization of Gli1^+ cells and Ancr/E7-EXO in the adventitia of TEBVs was observed from weeks 1 to 3 ([122]Fig. 6C). It is suggested that, at early stage of implantation, Ancr/E7-EXO can be swallowed by Gli1^+ cells to play a role. In vivo fluorescence is used to observe the metabolism of Ancr/E7-EXO after implantation for a longer time, the TEBVs modified by Ancr/E7-EXO labeled with 1,1-dioctadecyl-3,3,3,3-tetramethylindotricarbocyaine iodide (DIR) fluorescence probe were implanted into the carotid artery of rats ([123]Fig. 6D). The signals of Ancr/E7-EXO retention were observed in the neck of the rats from weeks 1 to 6 after implantation ([124]Fig. 6, D and E), indicating that the Ancr/E7-EXO still remained in the TEBVs for 6 weeks in vivo, thus providing sufficient time for the regulation of Gli1^+ cell differentiation. Fig. 6. Modification of TEBVs with Ancr/E7-EXO and enrichment of Ancr/E7-EXO in autologous calcified blood vessels. [125]Fig. 6. [126]Open in a new tab (A) Scheme of Ancr/E7-EXO loaded into the media and adventitia of TEBVs by EDC-NHS, and implantation of modified TEBVs. (B) Ancr/E7-EXO was labeled with PKH67, and representative images of acellular vessel and Ancr/E7-EXO–modified TEBVs by EDC/NHS are shown. (C) Ancr/E7-EXO was labeled with DiD, and immunofluorescence staining in the cross-sectional TEBVs in each group from 1 to 3 weeks after implantation (DAPI, blue; Gli1^+ cells, green; Ancr/E7-EXO, red). Scale bars, 100 and 500 μm. (D and E) Bioluminescence images and quantification of Ancr/E7-EXO–modified TEBVs obtained at different times after TEBV transplantation (n = 4). (F) Scheme of CKD rats treated with Ancr/E7-EXO. (G and H) Ancr/M7-EXO and Ancr/E7-EXO were injected into the tail vein of CKD rats at week 13 from CKD development, and the bioluminescence images and quantification of aortic and thoracic aorta were compared (n = 3). Results are expressed as means ± SD by two-tailed Student’s t test. *P < 0.05. The ANCR/M7-EXO and Ancr/E7-EXO labeled with DIR were injected at week 13 after subtotal nephrectomy ([127]Fig. 6F). The fluorescence signal from the beginning of the aorta to the thoracic aorta in the Ancr/E7-EXO group was 1.58 times higher than that in the ANCR/M7-EXO group ([128]Fig. 6, G and H), indicating that Ancr/E7-EXO enriched to the calcified site in vivo and was phagocytosed by Gli1^+ cells because of its special affinity with Gli1^+ cells. The enhancement of Ancr/E7-EXO on long-term patency and normal mechanical properties of TEBVs in vivo We implanted TEBVs modified by Ancr/E7-EXO into Sprague-Dawley rats. Seven days after implantation, diabetic rats were established by injection of streptozotocin, and their blood glucose was continuously monitored for 60 days ([129]Fig. 7A and fig. S8G). According to the type of TEBVs and the presence or absence of diabetes, the rats were divided into four groups: group 1: normal rats with TEBVs (N-TEBVs); group 2: diabetic rats with TEBVs (D-TEBVs); group 3: normal rats with Ancr/E7-EXO–modified TEBVs (N-TEBVs-Ancr/E7-EXO); and group 4: diabetic rats with Ancr/E7-EXO–modified TEBVs (D-TEBVs-Ancr/E7-EXO). Groups 1 and 2 showed more severe intimal hyperplasia and lower blood flow velocity at 42 days after implantation (fig. S8, A, C, and E), while groups 3 and 4 showed a slighter intimal hyperplasia and collagen deposition (fig. S8, A and B). The immunofluorescence staining revealed that the expression of α-SMA in the media of groups 3 and 4 was significantly higher than that in the control group (fig. S8, D and F). The number of contractile SMCs in diabetic rats of group 4 was 17.13 times higher than that in group 2. These results revealed that Ancr/E7-EXO promoted the rapid remodeling of SMCs in TEBVs. Hematoxylin and eosin (H&E) staining performed at day 60 after implantation showed that vascular stenosis and occlusion occurred after TEBV implantation, while Ancr/E7-EXO–modified TEBVs maintained excellent cell regeneration and vascular patency in both normal and high-glucose environment ([130]Fig. 7B). Masson staining showed that Ancr/E7-EXO–modified TEBVs had less collagen fiber deposition in both normal and high-glucose environment ([131]Fig. 7C), indicating that Ancr/E7-EXO inhibited collagen fiber secretion. The intima-media thickness ratio revealed that Ancr/E7-EXO significantly reduced intimal hyperplasia of TEBVs in groups 3 and 4 ([132]Fig. 7E). The results of Doppler ultrasound showed that the Ancr/E7-EXO group maintained a more stable hemodynamics in either normal physiological or high-glucose condition ([133]Fig. 7D). The blood flow velocity in group 3 was stable at 81.20 ± 21.51 cm/s, while that in group 4 was stable at 70.27 ± 15.73 cm/s, which were much higher than those in groups 1 and 2 ([134]Fig. 7F). Micro–computed tomography (micro-CT) also showed that the TEBVs in groups 3 and 4 remained unobstructed and in excellent shape in normal and high-glucose conditions, while the control group had no vascular signal ([135]Fig. 7G). The above results suggested that Ancr/E7-EXO might represent a promising solution for an early and rapid reconstruction of VSMCs as well as long-term patency of TEBVs in both normal and high-glucose conditions. Fig. 7. Examination of long-term patency, normal morphology, and mechanical properties of TEBVs. [136]Fig. 7. [137]Open in a new tab (A) Diabetic rat model established by the injection of streptozotocin at day 7 after TEBV implantation. The change in blood glucose in rats was measured from days 11 to 14 and then measured once a week. SMC remodeling was detected at day 60. (B) Intimal hyperplasia and SMC remodeling were observed by H&E staining. (C) Collagen secretion by Masson trichrome staining. (D and F) Patency was determined with a portable ultrasound scanner (n = 5). (E) Intima-media thickness ratio was calculated to evaluate intimal hyperplasia (n = 5). (G) Overall patency of TEBVs in each group by micro–computed tomography (micro-CT). (H and I) Young’s Modulus of TEBV media in all groups measured by atomic force microscopy (AFM) (n = 5). (J and K) Images of the height and peak force error showing the arrangement of vascular fibers in each group (n = 5). Results are expressed as means ± SD by two-tailed Student’s t test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. Furthermore, the mechanical properties of the media of TEBVs were examined by atomic force microscopy (AFM) ([138]Fig. 7H). At day 60, the Young’s modulus of TEBVs and native blood vessels indicated an increase in the stiffness in groups 1 and 2, with a value of 3325 and 3729 kPa, respectively, compared with the values of the native blood vessels, while the stiffness in groups 3 and 4 was similar to that of the native vessels ([139]Fig. 7I). This result indicated that Ancr/E7-EXO could maintain the normal vascular stiffness of TEBVs. The above results suggested that, on the one hand, Ancr/E7-EXO inhibited the differentiation of Gli1^+ cells into synthesized SMCs and reduced collagen deposition; on the other hand, Ancr/E7-EXO inhibited the differentiation of Gli1^+ cells into osteoblast-like cells, thereby reducing the calcification and maintaining a normal vascular stiffness. The morphological characteristics ([140]Fig. 7J) and height characteristics ([141]Fig. 7K) showed that groups 3 and 4 had a regular fiber arrangement similar to that of the native vessels, while the matrix fiber arrangement of the control group was disordered. These results provided direct evidence that Ancr/E7-EXO maintained the normal vascular morphology and stiffness. The dual effects of Ancr/E7-EXO on promoting SMC remodeling and inhibiting calcification of TEBVs in vivo To evaluate the cell reconstruction after TEBV implantation, we quantified the cell composition of TEBVs in each group at day 42. In groups 1 and 2, we observed that osteoblast-like cells (OPN) contributed 92.5% and 86.3% of Gli1^+ cells, while only 13.6 and 10.7% of cells in groups 3 and 4 expressed OPN (fig. S9, A and C). These results indicated that Ancr/E7-EXO significantly inhibited Gli1^+ cells differentiated into osteoblasts. Similarly, the composition of SMCs was quantified. The proportion of contractile SMCs in group 3 was 63.5%, which was higher than that in group 1 (fig. S9B). However, there was no difference among all groups for the proportion of α-SMA (fig. S9D). The reason is that a large number of SMCs in groups 1 and 2 abnormally proliferate and migrate to the intima, leading to serious intima hyperplasia. Thus, the expression of contractile SMCs is abnormally increased. Furthermore, in the coimmunofluorescence staining performed at 60 days after implantation, it showed that few Gli1^+ cells and α-SMA were both located in groups 3 and 4, and the contractile SMCs in the media of groups 3 and 4 increased significantly compared with that in groups 1 and 2 ([142]Fig. 8A). The number of α-SMA positive cells in the media of groups 3 and 4 was 53.92 and 8.38 times higher than that of groups 1 and 2, respectively ([143]Fig. 8B); meanwhile, the number of thrombospondin positive cells decreased by 8.14 and 24.92% compared with groups 1 and 2, respectively ([144]Fig. 8C). OPN and Runx2 in TEBVs were stained to evaluate the inhibitory effect of Ancr/E7-EXO on vascular calcification. Double immunofluorescence staining revealed that more Gli1^+ cells and OPN were colocalized in groups 1 and 2, while groups 3 and 4 had almost no Gli1^+ cells and OPN signal ([145]Fig. 8, A and E). The Runx2 fluorescence intensity in groups 1 and 2 were 12.86 and 4.22 times higher than that of groups 3 and 4, respectively ([146]Fig. 8D). It indicated that Ancr/E7-EXO could significantly reduce the calcification of TEBVs, and this result was consistent with that of the vascular calcification found by Alizarin red and von Kossa staining ([147]Fig. 8, F and G). The above results suggested that Ancr/E7-EXO promoted vascular smooth muscle remodeling and inhibited the calcification after TEBV implantation, providing a promising therapeutic approach for the reconstruction of smooth muscle in TEBVs and the resistance to vascular calcification aggravated by basic diseases such as diabetes. Fig. 8. The regulation of Ancr/E7-EXO on SMC remodeling and calcification in TEBVs. [148]Fig. 8. [149]Open in a new tab (A) Immunofluorescence staining in the cross-sectional TEBVs in each group at 60 days after implantation (DAPI, blue; Gli1^+ cells, red; α-SMA, green; thrombospondin, green; Runx2, red; OPN, green). Scale bars, 20 μm. (B to E) Quantification of α-SMA, thrombospondin, Runx2, and OPN in the media of cross-sectional TEBVs (n = 3). (F and G) TEBV calcification in each group by Alizarin red and von Kossa staining. Results are expressed as means ± SD by two-tailed Student’s t test. *P < 0.05, **P < 0.01, and ***P < 0.001. The therapeutic effect of Ancr/E7-EXO on calcification of autologous vessels Ancr/E7-EXO also played the same role in vascular calcification caused by CKD. Increased levels of BUN were detected from weeks 1 to 12 ([150]Fig. 9A). According to FRI of near-infrared spectroscopy, calcium tracer showed that the positive signal of hydroxyapatite in the treatment group (5/6Nx 13 weeks + Ancr/E7-EXO) was 60.42% of that in the control group (5/6Nx 13 weeks), suggesting the degree of vascular calcification decreased in CKD rats after Ancr/E7-EXO treatment ([151]Fig. 9, B and C). The double immunofluorescence staining showed the presence of more Gli1^+ cells and contractile SMCs colocated in the media of the treatment group ([152]Fig. 9E). The number of Gli1^+ cells expressing α-SMA in the media was 110.4, which was 2.94 times higher than that in the control group ([153]Fig. 9F). Many Gli1^+ cells and osteoblast-like cells were colocated in the media of the control group, and the number of Gli1^+ cells expressing OPN in the media was 49.94, which was 6.39 times higher than that in the treatment group ([154]Fig. 9, E and G). The above results proved that Ancr/E7-EXO inhibited the osteogenic differentiation of Gli1^+ cells in autologous vessels and promoted the differentiation of Gli1^+ cells into contractile SMCs. Fig. 9. Protection of Ancr/E7-EXO against autologous vascular calcification in CKD. [155]Fig. 9. [156]Open in a new tab (A) BUN stratified for the sham and treatment group. (B and C) Representative images and quantification of hydroxyapatite fluorescence intensity in the sham, control, and treatment groups (n = 3). (D) Volcano plot between the vascular samples of the treatment and control groups. Red dots, genes significantly down-regulated in treatment group; yellow dots, genes significantly up-regulated in treatment group; blue dots, genes not differentially expressed. (E) Representative images of cross-sectional aorta in the sham, control, and treatment groups by immunofluorescence staining (DAPI, blue; Gli1^+ cells, red; α-SMA, green; OPN, green) (n = 4). (F and G) Quantification of Gli1^+ cells expressing α-SMA and OPN in the media of cross-sectional aorta (n = 4). (H) Heatmap of calcification-related gene expression. (I) Bubble map of the top 10 biological process terms in the Gene Ontology (GO) enrichment analysis. (J) Top 30 of Kyoto Encyclopedia of Genes and Genome (KEGG) pathways in the enrichment analysis. cAMP, cyclic adenosine 3′,5′-monophosphate; CoA, coenzyme A; IL-17, interleukin-17; MAPK, mitogen-activated protein kinase; TGF-β, transforming growth factor–β. (K) Gene set enrichment analysis of muscle contraction. Results are expressed as means ± SD by two-tailed Student’s t test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. wk, weeks. Furthermore, bioinformatic technique was used to analyze the differentially expressed genes in aorta and thoracic aorta induced by CKD between the Ancr/E7-EXO and control groups. We observed that the expression of Ptgs2 decreased, while the expression of Myocd, Nos3, Cd163, and Hspb7 increased ([157]Fig. 9D). Ptgs2, also known as COX-2, could induce the increased mRNA expression of osteogenic genes OPN and Runx2 and lead to calcification of aortic valve and aorta ([158]25, [159]26). Ptgs2 also played a pivotal role in the process of atherosclerosis, and its expression could be used as a biomarker of the degree of atherosclerosis ([160]27). In this study, the expression of Ptgs2 decreased after Ancr/E7-EXO treatment, indicating that Ancr/E7-EXO could inhibit vascular calcification–related genes. Myocd can regulate the expression of genes encoding SMC contractile proteins to maintain the contractile phenotype of SMCs ([161]28). In this study, Myocd was up-regulated, indicating that Ancr/E7-EXO could also maintain the contractile phenotype of SMCs. In addition, up-regulated expression of Nos3, Hspb7, and Cd163 indicated that Ancr/E7-EXO can play an active role in the maintenance and regulation of vascular immune microenvironment ([162]29–[163]31). At the same time, by comparing the expression changes of vascular calcification–related genes, the expression of calcification-related genes decreased significantly in the Ancr/E7-EXO group ([164]Fig. 9H). These results suggested that Ancr/E7-EXO can up-regulate the expression of contraction-related genes and down-regulate the expression of vascular calcification–related genes in SMCs. Moreover, Gene Ontology (GO) analyzed the most abundant biological processes, including myocardial contraction (P < 0.00001) ([165]Fig. 9I). Here, our sample tissues were aorta and thoracic aorta, and aortic smooth muscle and myocardium may have the same developmental origin, so we thought that enriched myocardial contraction is the SMC contraction. At the same time, by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, myocardial contractile signal pathway was detected in top 30 of the enrichment pathways (P < 0.02) ([166]Fig. 9J). In addition, after the gene set enrichment analysis of the overall expression forms of muscle contraction pathway, the related gene set was up-regulated in the Ancr/E7-EXO group ([167]Fig. 9K). These results suggested that Ancr/E7-EXO can promote vascular SMCs differentiation into contractile SMCs, thus maintaining vascular contractile function. DISCUSSION Vascular calcification is a chronic vascular disease, which is not only a common cause of vascular graft failure but also a common complication of diabetes, CKD, and other diseases. Previous studies on small diameter vascular grafts focused on the evaluation of the efficacy of TEBVs on neointimal hyperplasia and thrombosis in the early graft (≤3 months), which are early complications of grafts that are prone to vascular stenosis or blockage ([168]32). Calcification is an important risk factor for late graft failure in patients who underwent bioengineered cardiovascular grafts ([169]6) suffering from underlying diseases such as diabetes ([170]33), which is a problem rarely evaluated and managed. After TEBV implantation, on the one hand, the reconstruction of SMCs in vivo is difficult; on the other hand, TEBVs are more likely to calcify in combination with injury and pathological environment. Therefore, how to promote the normal reconstruction of the smooth muscle layer of TEBVs and inhibit calcification is still a technical challenge. Moreover, clinical study found that medial arterial calcification is detected in 17 to 42% of patients with type 2 diabetes mellitus, in 27 to 40% of patients with advanced CKD, and in up to 72% of patients with threatening chronic limb ischemia ([171]2). Severe vascular calcification can increase arterial hardness, can lead to loss of vascular function and even vascular rupture, and can lead to subsequent incidence and mortality of cardiovascular disease ([172]34). The incidence and degree of vascular calcification in patients with CKD are higher than those in the general population, and the incidence of vascular calcification increases with the decline of the renal function ([173]35). There is no noninvasive and effective solution for vascular calcification caused by TEBVs and CKD ([174]33). Endogenous vascular progenitor cells are an important source of cells for repair and regeneration after vascular injury, and their abnormal differentiation will also lead to abnormal repair after injury ([175]36). Gli1 is considered to be a specific marker of MSCs in vascular adventitia and has the differentiation ability of SMCs and osteoblast-like cells in the process of injury and repair ([176]14). As endogenous vascular progenitor cells, the fate and contribution of Gli1^+ cells after TEBV implantation are still unclear. Therefore, we evaluated the spatiotemporal distribution of Gli1^+ cells after TEBV implantation. The results showed that Gli1^+ cells continued to increase in both media and adventitia after TEBV implantation in normal and diabetic rats. Gli1^+ cells were significantly present at day 3 after TEBV implanted in diabetic rats. The Alizarin red and von Kossa staining of TEBVs revealed the formation of calcification foci as early as 2 weeks after implantation. These results indicated that the spatiotemporal distribution of Gli1^+ cells in TEBVs was positively correlated with the degree of calcification under normal physiological and pathological conditions. Through Gli1^+ costaining with OPN, more Gli1^+ cells had osteogenic phenotype in both normal and diabetic groups, indicating that Gli1^+ cells are also the key source of TEBV calcification, which is the first time to find that calcification of TEBVs is driven by Gli1^+ cells. Besides, we also localized Gli1^+ cells during autologous vascular calcification. After 13 weeks of induction of CKD in rats, we observed that Gli1^+ cells were mainly distributed in the media of blood vessels. Immunofluorescence staining showed that the presence of Gli1^+ cells was eight times higher than that of the control group, which was also consistent with the degree of aortic calcification shown by calcium tracer. Therefore, we also verified that the calcification of autologous blood vessels is caused by the differentiation of Gli1^+ cells into osteoblasts. Then, how to deal with TEBV and autologous vascular calcification caused by the differentiation of Gli1^+ into osteoblasts is the key target for us to solve the problem of vascular calcification. Effectively targeting and regulating the differentiation of Gli1^+ cells is the key goal of this study. Previous studies focused on the regulation of graft environment, such as modifying antioxidant substances ([177]32) or nitric oxide (NO)–releasing molecules on TEBVs ([178]9), achieving anticalcification through antioxidant effect, or using the immunomodulatory effect of MSC-EXO to prevent calcification ([179]37), but these studies did not eliminate the symptoms of vascular calcification from the source. Exosome is the key bridge of intercellular communication, which can be used for signal transmission such as RNA. Therefore, we designed Ancr/E7-EXO–engineered exosomes that can target Gli1^+ cells. The E7 peptide attached to the membrane surface has a special affinity for Gli1^+ cells, while the lncRNA-ANCR loaded inside can inhibit the differentiation of osteoblasts. We observed that the exosomes modified with E7 peptide could effectively target Gli1^+ cells. In the coculture of macrophages, neutrophils, and Gli1^+ cells, E7-EXO still had a high affinity for Gli1^+ cells. Therefore, Ancr/E7-EXO can avoid early phagocytosis of macrophages and provide time for Ancr/E7-EXO to reprogram Gli1^+ cells in vivo. After TEBV implantation, the rapid reconstruction of smooth muscle layer and the inhibition of calcification are the main issues of this study, which are also an important part of the functional remodeling of TEBVs in vivo and play an important role in maintaining vascular contraction and mechanical strength. However, the difficulties in SMC reconstruction and abnormal proliferation after reconstruction often lead to the occurrence of aneurysms and intimal hyperplasia. By modifying Ancr/E7-EXO on TEBVs, we verified its function in rat carotid artery replacement model. We observed that SMC regeneration appeared at the sixth week after operation, and there were abundant and orderly vascular smooth muscle layers in the media of TEBVs at the eighth week. At the same time, TEBVs modified with Ancr/E7-EXO can avoid intimal hyperplasia after implantation, in either normal or diabetic environment. The mechanism may be that Ancr/E7-EXO promotes the rapid and normal reconstruction of the smooth muscle layer, reduces the abnormal proliferation and migration of SMC, and thus inhibits the intimal hyperplasia of TEBVs. Therefore, Ancr/E7-EXO can promote the rapid and normal reconstruction of TEBV smooth muscle layer, avoid intimal hyperplasia, and open the way for the rapid host of TEBVs. We also evaluated the late calcification of the grafts. Calcification often leads to increased arterial stiffness and decreased vascular compliance ([180]38). In the control group, we found that the graft stiffness increased significantly at the eighth week. The stiffness of TEBVs modified by Ancr/E7-EXO is close to that of normal blood vessels. These results suggest that Gli1^+ cells can proliferate and differentiate into osteoblast-like cells during the repair of TEBV injury, which leads to the increase of graft stiffness, while Ancr/E7-EXO can inhibit the differentiation of Gli1^+ cells into osteoblast-like cells, thus making TEBV stiffness close to normal blood vessels. In addition, a large number of osteogenic signals were found in the TEBVs of the control group by immunofluorescence staining and flow cytometry. However, the osteoblast-like signal of Ancr/E7-EXO group was significantly suppressed, indicating that Ancr/E7-EXO could significantly inhibit the calcification of TEBVs. This may be due to the inhibitory effect of lncRNA-ANCR on osteoblast-like differentiation of Gli1^+ cells. Therefore, the Ancr/E7-EXO that we constructed can target and regulate Gli1^+ cells in vivo and overcome the problem of TEBV calcification from the source. It not only can promote the rapid reconstruction of SMCs but also can effectively inhibit the early intimal hyperplasia and long-term calcification of the graft and promote the normal reconstruction of the smooth muscle layer of TEBVs. Because the main cause of autologous vascular calcification is also the differentiation of Gli1^+ cells into osteoblast-like cells during chronic injury, the Ancr/E7-EXO that we designed has a special affinity for Gli1^+ cells. FRI showed that the exosome signal of aortic calcification site was higher than that of the control group, indicating that Ancr/E7-EXO was enriched in the calcification site. In addition, the autologous vessels treated with Ancr/E7-EXO had more phenotypes of SMCs and Gli1^+ cells, indicating that Ancr/E7-EXO promoted Gli1^+ cell differentiation into SMCs. In the control group, more osteogenic phenotypes were colocated with Gli1^+ cells, and the expression of OPN in autologous vessels treated with Ancr/E7-EXO decreased, indicating that Ancr/E7-EXO significantly inhibited the differentiation of Gli1^+ cells into osteogenic phenotypes. Calcium tracer imaging further showed that Ancr/E7-EXO inhibited autologous vascular calcification under pathological condition. By bulk RNA sequencing (RNA-seq), we observed a significant difference in calcification gene expression between the control group and the Ancr/E7-EXO treatment group. Ptgs2 could induce the increased expression of osteogenic genes OPN and Runx2 ([181]25, [182]26), in which the expression decreased after Ancr/E7-EXO treatment. In addition, the expression of genes encoding SMC contractile protein also increased after Ancr/E7-EXO treatment. The increased expression of genes related to the regulation and maintenance of vascular immune microenvironment may be the role of exosomes derived from MSCs ([183]37). These results further proved that Ancr/E7-EXO reprogrammed Gli1^+ cells in vivo, thus achieving the purpose of noninvasive treatment of autologous vascular calcification caused by chronic diseases. Because autologous vascular calcification has the same pathological mechanism in a variety of pathological conditions, the Ancr/E7-EXO designed in this study can also be used for autologous vascular calcification caused by other chronic diseases. For example, in vascular calcification caused by atherosclerosis and chronic diabetes, Ancr/E7-EXO also has an anticalcification effect. In conclusion, the Ancr/E7-EXO designed in this study can effectively capture and reprogram Gli1^+ cells by modifying TEBVs, thus accelerating the normal reconstruction of the smooth muscle layer of TEBVs and significantly inhibiting the calcification. Ancr/E7-EXO can also be infused intravenously, which can enrich in the calcified site and significantly inhibit autologous vascular calcification caused by CKD. This method provides a promising treatment strategy for vascular calcification and other cardiovascular graft calcification under complex pathological conditions, having a translational value in clinical application. MATERIALS AND METHODS Cell culture The HuBM-MSCs were purchased from Biospes and cultured in α–modified essential medium (α-MEM; HyClone), supplemented with 10% exosome-depleted fetal bovine serum (FBS) [Biological Industries (BI)] and 2% penicillin-streptomycin (Beyotime Biotechnology), and incubated in a humidified environment at 37°C under 5% CO[2]. The plasmid expressing E7-Lamp2b fusion protein was transfected into HuBM-MSCs using the Entranster-H4000 transfection reagent (Engreen). Gli1^+ cells were mechanically harvested from the femurs of Sprague-Dawley rats as previously described ([184]39). In brief, 100-g Sprague-Dawley rats were selected and euthanized through cervical dislocation. Bilateral femur and tibia were removed under sterile conditions and immersed in a phosphate-buffered saline (PBS) solution; the middle femur and tibia were cut into debris to release the bone marrow from the cavity. The cell culture medium containing 2% dual antibody α-MEM was aspirated with a 2-ml syringe, and bone marrow was repeatedly rinsed. The collected cell suspension was filtered through a 75-μm filter and centrifuged at 1000 rpm/min for 5 min. Then, the red blood cells were removed from collected cell suspension using red blood cell lysis buffer (Solarbio), the cell suspension was centrifuged at 1000 rpm/min for 5 min, and the cells were seeded in a flask containing α-MEM cell culture medium supplemented with 10% FBS (AusGeneX). The differentiation of purified Gli1^+ cells was obtained by culturing them until 60 to 70% or 90 to 100% confluence in 24-well plate, and α-MEM medium was replaced by osteogenic, chondrogenic, and adipogenic differentiation media (Cyagen). Alkaline phosphatase (Cyagen) staining and Oil red O (Sigma-Aldrich) staining were performed after 14 days of culture. Alcian blue (Cyagen) staining was performed after 21 days of chondrogenic induction. The smooth muscle cell differentiation was obtained by culturing Gli1^+ cells in α-MEM, containing 10% FBS, 2% penicillin-streptomycin, transforming growth factor–β (10 ng/ml; PeproTech), and platelet-derived growth factor B (5 ng/ml; PeproTech). The osteogenic differentiation was obtained by culturing Gli1^+ cells in osteogenic differentiation medium (Procell) supplemented with 30 mM glucose solution, and the culture medium was changed every 2 days; the cells at three to four passages were used for in vitro experiments. Cells cultured for 7 and 14 days were subjected to Alizarin red staining (Cyagen). Flow cytometry Rat Bone Marrow (RBM)-MSCs were subjected to an in vitro culture for three passages and selected to characterize the surface markers. A total of 10^6 cells were suspended in a volume of 400 μl of sterile PBS supplemented with 1% FBS. The cells were labeled with an antibody cocktails for 40 min on ice: Alexa Fluor 488–conjugated rat anti-mouse CD34 (1:100, BioLegend), allophycocyanin-CD45 (1:100, BioLegend), phycoerythrin (PE)–CD44 (1:100, eBioscience), PE-Cy7-CD29 (1:100, BioLegend), and fluorescein isothiocyanate (FITC)–Sca1^+ (1:100, Biorbyt). Cells were washed twice with cold PBS after incubation and then separated on a FACS Calibur flow cytometer (BD Biosciences). The data were analyzed using FlowJo V10 software (Tree Star Inc., Ashland, OR, USA). Flow sorting was performed when RBM-MSCs were subcultured to P3 passage. The cells were resuspended at a density of 10^6 in 2 ml of PBS containing 10 μl of PE-CD44 (1:100, BD Bioscience), PE-Cy7-CD29 (1:100, BioLegend), FITC-Sca1+ (1:100, Biorbyt), 10% FBS, and 2% dual antibody and incubated at 4°C for 30 min. The cells were washed twice with PBS and then sorted. MSC-EXO was analyzed on a Flow NanoAnalyzer (N30E, China) instrument. MSC-EXO pellets were resuspended in 30 μl of PBS. To a 30-μl MSC-EXO sample with a particle concentration of approximately 1.67×10^5 particles/ml, 20 μl of FITC-conjugated antibody against CD9, CD63, and CD81 was separately added. The mixture was incubated at 37°C for 30 min in the dark. MSC-EXO was washed twice with 1 ml of cold PBS by ultracentrifugation at 110,000g for 70 min at 4°C. The pellets were resuspended in 50 μl of cold PBS for NanoFCM analysis. TEBV dissociation and flow cytometry A single-cell suspension of TEBVs was prepared following the enzymatic digestion protocol described previously ([185]40). Briefly, the isolated TEBVs were finely cut and incubated in 1 ml of collagenase type II (20 mg/ml) for 1 hour at 37°C. The cell suspension was strained through a 40-μm filter and washed twice with PBS. The cells were resuspended at a density of 10^6 in 2 ml of PBS containing 10 μl of Alexa Fluor 488–conjugated rat anti-mouse GLI1 (1:50, Santa Cruz Biotechnology), PE–α-SMA (1:100, Novus Biologicals), Alexa Fluor 647–conjugated rat anti-mouse OPN (1:50, Santa Cruz Biotechnology), 10% FBS, and 2% dual antibody and incubated at 4°C for 30 min. The cells were washed twice with PBS and then analyzed. Exosome isolation The supernatant of E7 plasmid–transfected HuBM-MSCs subjected to 48 hours of culture was collected and centrifuged at 3000g for 10 min at 4°C. The supernatant was collected and filtered using a 0.22-μm filter (Biosharp) to remove the remaining cellular debris. Then, the supernatant was ultracentrifuged at 110,000g for 120 min at 4°C using an ultrahigh-speed centrifuge (CP100NX, HITACHI, Japan). The exosome pellets were resuspended in PBS, and the amount of exosomal protein was measured by the BCA Protein Assay kit (Beyotime). The exosomes were stored at 80°C. Loading of E7-EXO with plasmid A total of 25 μg of purified exosomes (BCA Protein Assay Kit, Beyotime) and 25 μg of ANCR plasmid were gently mixed in 100 μl of electroporation buffer to load E7-EXOs with ANCR plasmid. Electroporation was carried on an electroporation cuvette using a Gene Pulser II Electroporator (Bio-Rad, USA) at 100 V and 50 μF. After electroporation, the mixture was immediately placed at 37°C for 30 min to completely recover the membrane of the exosomes. The intact Ancr/E7-EXO membrane was confirmed by TEM, and the Ancr/E7-EXOs were washed twice with cold PBS and centrifuged at 100,000g for 70 min to remove the ANCR plasmid that was not incorporated into the exosomes. The efficiency of electroporation of the ANCR plasmid was detected by qPCR. Western blotting Total proteins were extracted from HuBM-MSCs or exosomes using radioimmunoprecipitation assay lysis buffer, and the concentration was determined by bicinchoninic acid (BCA) assay (Beyotime). Proteins were separated by electrophoresis and transferred to a polyvinylidene difluoride membrane. The membrane was incubated with the following primary antibodies: rat anti–Lamp-2b (1:400, Santa Cruz Biotechnology); rabbit anti-CD63 antibody (1:500, Bioss); rabbit anti–throbospondin 2 antibody (1:500, Bioss); mouse anti–α-SMA antibody (1:500, Bioss); and rabbit anti-GLI1 antibody (1:500, Bioss) at 4°C overnight, followed by 2 hours of incubation with the corresponding secondary antibody at room temperature. The blots were visualized by enhanced chemiluminescence (GelDoc 2000 Gel imaging system, Bio-Rad). Real-time qPCR and qPCR Total RNA was extracted from HuBM-MSCs using an miRNeasy Mini kit (217004, Qiagen) according to the manufacturer’s instructions. Complementary DNA (cDNA) was synthesized using the RevertAid First Strand cDNA Synthesis (K1622, Thermo Fisher Scientific). cDNA was amplified in a real-time qPCR thermocycler (CFX96, BIO-RAD) using the SYBR Green qPCR Master (4943914001-SR, Roche). Cel-miR-39 was used as control, and the relative gene expression was calculated using the 2^−ΔΔCT method. The primers used for reverse transcriptase (RT)–PCR were as follows: rat DANCR, (forward) 5′-ACATTTAATGACCCCGCTTG-3′ and (reverse) 5′-GATTCCAGGACACCCAGAAA-3′; rLamp2b-E7, (forward) 5′-TTGGCTAATGGCTCAGCTTT-3′ and (reverse) 5′-ATGGGCACAAGGAAGTTGTC-3′; and cel-miR-39, (forward) 5′-agcccgTCACCTGGTGTAAATC-3′ and (reverse) 5′-CAGTGCAGGGTCCGAGGTAT-3′. In qPCR for osteogenic inhibition, Gli1^+ cells were dissolved in TRIzol reagent, and total RNA was isolated by a TRIpure total RNA rapid extraction kit. cDNA was synthesized using a TSK302S (RT6 cDNA Synthesis Kit version 2) and amplified in a RT-qPCR thermocycler (FQD-96A, BORI Hangzhou, China) using the 2× T5 Fast qPCR Mix (SYBR Green I). The relative gene expression was normalized to the expression of actin. The primers used for RT-PCR were as follows: MYH11, (forward) 5′-GGCCATGAGTGACAGAGTCC-3′ and (reverse) 5′-TTGCCGTGCACTCTCATTCT-3′; α-SMA, (forward) 5′-CAGCTATGTGGGGGACGAAG-3′ and (reverse) 5′-TCCGTTAGCAAGGTCGGATG-3′; thrombospondin 2, (forward) 5′-CCCAAGGGGACCACACAAAT-3′ and (reverse) 5′-TAGTCATCGTCCCGGTCAGT-3′; OPN, (forward) 5′-CCGAGGTGATAGCTTGGCTT-3′ and (reverse) 5′-GACTCATGGCTGGTCTTCCC-3′; BMP-2, (forward) 5′-GCCAAACACAAACAGCGGAA-3′ and (reverse) 5′-GGTGATCAGCCAGGGGAAAA-3′; Runx2, (forward) 5′-ATGGCCGGGAATGATGAGAA-3′ and (reverse) 5′-GGGGAGGATTTGTGAAGACCG-3′; and actin, (forward) 5′-AGATCAAGATCATTGCTCCTCCT-3′ and (reverse) 5′-ACGCAGCTCAGTAACAGTCC-3′. Gli1^+ cell endocytosis of E7-EXOs Gli1^+ cells were prestained with PKH67 (green), and SMCs, ECs, neutrophils, and macrophages were stained with CellMasTM Deep Red plasma membrane stain (Life Technology) (blue). Gli1^+ cells and SMCs, ECs, neutrophils, and macrophages were separately cultured in 24-well plate at a density of 2 × 10^5 cells. Meanwhile, a cocktail containing both Gli1^+ cells and SMCs, ECs, neutrophils, and macrophages (cell density of 1 × 10^5, respectively) was seeded together in 24-well plates. E7-EXO and M7-EXO were fluorescently labeled with PKH26 (red) membrane dye (Bestbio). After labeling, the E7-EXO and M7-EXO were washed with 50 ml of PBS, collected by ultracentrifugation, and resuspended in PBS. After cell adherent, 2 μg of E7-EXO or M7-EXO was added into 2 × 10^5 recipient cells and incubated for 24 hours. Unphagocytic exosomes were washed with PBS, and the nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI) after fixation with 4% paraformaldehyde. Olympus laser scanning confocal microscope (Olympus SpinSR, Japan) was used to observe the phagocytosis of exosomes by Gli1^+ cells and macrophages. Vascular cell preparation and Ancr/E7-EXO–modified TEBVs The carotid artery of Sprague-Dawley rats was isolated, and the blood was rinsed away with PBS. The vessels were digested with 20% trypsin at 4°C for 2 hours and then washed twice with PBS to obtain the vascular matrix. The prepared vascular matrix was first incubated with collagen solution (4 mg ml^−1) for 24 hours, washed twice with PBS, and then incubated with 0.1 M EDC, 0.7 M NHS, and 20 μl of Ancr/E7-EXO (25 μg/μl) at 4°C for 48 hours to obtain Ancr/E7-EXO–modified TEBVs. Implantation of TEBVs in Sprague-Dawley rats and establishment of CKD rat model All animal surgeries and experiments were approved by the Animal Ethics Committee of the Military Medical University. Male Sprague-Dawley rats (230 to 250 g) were treated with an intraperitoneal injection of 1% sodium pentobarbital (40 mg/kg). The rats were anesthetized, and the two ends of one side of the common carotid artery were removed by forceps, cut in the middle, pulled out by a self-made cuff, and flipped. Both ends of the prepared TEBVs [1 cm long, 1 mm diameter, soaked in heparin sodium (50 U/ml)] were anastomosed to the end of the carotid artery. The vascular suture was ligated at the cuff and the vascular clamp was released. The diabetic rats were treated with an intraperitoneal injection of streptozotocin on day 7 after the operation to establish the diabetic rat model, which was performed according to the literature ([186]41). A two-step subtotal nephrectomy (5/6Nx) was performed according to the literature ([187]14), and a right single nephrectomy was performed 1 week after the left subtotal nephrectomy. Rats in the sham operation group were fed with normal diet. Rats in the Ancr/E7 group were treated with Ancr/E7-EXO (10 mg/kg) through the tail vein every 5 days. The control group and the sham group were treated with the same volume of normal saline for 13 weeks. Calcification was accelerated by feeding the rats with a high-phosphorus diet (1.2% Pi) throughout the study. The CKD model was established successfully by increasing the serum urea nitrogen level. Rats were euthanized at 13 weeks after surgery. Assay of Gli1^+ phagocytosis of Ancr/E7-EXO in vivo Ancr/E7-EXO was fluorescently labeled with 1,1'-Diocatadecyl-3,3,3',3'-Tetramethylindodicarbocyanine,4-Chlorobenze nesulfonate Salt (DiD) membrane dye (Engineering For Life), and, then, Ancr/E7-EXOs were washed twice with 10 ml of PBS by ultracentrifugation at 100,000g for 70 min at 4°C. Then, Ancr/E7-EXO was modified to vascular matrix in accordance with the above method. The Ancr/E7-EXO–modified TEBVs were implanted into rats. On days 7, 14, and 21, rats were anesthetized, and, then, the TEBVs were harvested and postfixed with 4% paraformaldehyde for 3 hours at 4°C and then placed in 30% sucrose overnight. TEBVs were embedded by Optimal Cutting Temperature Compound (OCT) and then sectioned. Assay of Ancr/E7-EXO stability in vivo Ancr/E7-EXO was fluorescently labeled with DiR membrane dye probe (Abcam), and the unbound probe was washed twice with 10 ml of PBS by ultracentrifugation at 100,000g for 70 min at 4°C. Then, Ancr/E7-EXO stained by probe was modified to vascular matrix in accordance with the above method. The Ancr/E7-EXO–modified TEBVs were implanted into rats. Rats were anesthetized at various time points to receive skin preparation for FRI (NEWTON 7.0 FT-100. VILBER). Filter parameters were set to 782-nm emission and 750-nm excitation. Histological analysis Harvested TEBVs were rinsed with saline and cut into two sections. One section of TEBVs was directly embedded in a cryogenic agent, cut into 40-μm-thick sections, and used to measure the Young’s modulus by AFM. The other section of TEBVs was immediately fixed in 4% paraformaldehyde, embedded in paraffin, and cut into 5-μm-thick sections. Paraffin sections were stained with H&E, Masson’s trichrome, von Kossa, and Alizarin red. Images were observed under a bright-field microscope (Olympus VS200, Japan). Immunofluorescence staining Cells were fixed in 4% paraformaldehyde for 15 min, and the paraffin sections of TEBVs were dewaxed. After washing twice with PBS, 0.05% Triton X-100 (Solarbio, 30 min, room temperature) and 10% goat serum (Solarbio, 40 min, 37°C) in PBS were used for permeation and blocking, respectively. The samples were then incubated with antibodies at 4°C overnight. The primary antibodies were as follows: mouse anti-Gli (1:200, Novus Biologicals), mouse anti-MYH11 (1:200, Abcam), mouse anti–α-SMA (1:300, Boster), rabbit anti-thrombospondin (1:200, Abcam), rabbit anti-Runx2 (1:200, Bioss), and mouse anti-OPN (1:200, Bioss). The secondary antibodies were as follows: Alexa Fluor 488–, Alexa Fluor 568–, and Alexa Fluor 647–conjugated antibodies (Invitrogen). The nuclei were stained with DAPI (Biosharp) for 5 min at room temperature. Confocal microscopy images were taken and analyzed by the Olympus laser scanning confocal microscope (Olympus SpinSR, Japan) and cellSens software, respectively. Macroscopic fluorescence reflection imaging Aortic calcification was detected by a bisphosphonate derivative near-infrared fluorescent imaging agent (Osteosense 750 EX, PerkinElmer, Boston, MA, USA), which was injected into the rats through the tail vein 24 hours before imaging. The rats were euthanized, and the aorta was perfused with normal saline. After dissection, FRI was used to map the near-infrared fluorescence signal using OsteoSense 750 EX fluorescent imaging agent (excitation/emission: 745/800 nm). Ultrasound and micro-CT detection of TEBVs in vivo The rats were anesthetized with 1% sodium pentobarbital, and the neck skin was prepared. The neck of the rats was smeared with ultrasonic gel. The color Doppler ultrasound (Esaote) frequency was 13.0 MHz, the depth was 2.5 cm, the mechanical index was M10.4, and the gray scale was 46. After the detection of several consecutive cardiac cycles, the Doppler mode was selected to obtain the common carotid artery ultrasound spectrum. The images were saved, and the carotid blood flow velocity of the grafted TEBVs was measured. After anesthesia, the thoracic cavity of the rats was opened, and the angiography agent iohexol (300 mg/ml; Sigma-Aldrich) was injected from the left ventricle. The patency of TEBVs in each group was characterized by CT images using Bruker micro-CT of the carotid artery in small animals. AFM assay An AFM (JPK NanoWizard NanoOptics, Germany) was used to measure vessel stiffness and to scan vessel media. The optical microscopy mirror (Eclipse-Ti-S, Nikon, Japan) combined with AFM was able to control the tip and sample positioning. All measurements were performed with the same soft cantilever (MLCT Bio, Bruker; nominal spring constant, 0.01 N/m). Four points were measured for each blood vessel. A total of 50 to 60 points were measured, and the average Young’s modulus was calculated. AFM force mapping of the media was performed in the range of 30 μm. Bulk RNA-seq analysis of aorta and thoracic aorta of CKD rats Harvested aorta and thoracic aorta of CKD rats were rinsed with precooled PBS and immediately flash-frozen in liquid nitrogen. The raw RNA-seq data were collected by Berry Genomics Corporation, Beijing, China. R package DESeq2 v1.28.1 was used to perform differential gene expression analysis of bulk RNA-seq data. The resulting P values were adjusted using the Benjamini and Hochberg’s approach for controlling the false discovery rate. Genes with |log[2] (fold change)| > 1 and q value < 0.05 were assigned as differentially expressed. GO and KEGG enrichment analysis of differentially expressed gene sets were implemented by the top G ([188]www.bioconductor.org/packages/release/bioc/html/topGO.html) and KOBAS ([189]42) package, respectively. Statistical analysis Statistical analysis was performed using GraphPad Prism 8.0 (San Diego, CA, USA) software. All experimental results were presented as means ± SD. Two groups were compared using two-tailed Student’s t test. A value of P < 0.05 was considered statistically significant. Acknowledgments