Abstract The tissue environment governs vascular remodeling, a key determinant of collateral circulation (CC) in ischemic disease, yet the mechanisms driving CC in adults remain unclear. Plasma profiling from patients with peripheral artery disease (PAD) and ischemic murine muscle revealed dysregulated lipid metabolism, including elevated APOA1 binding protein (AIBP), with levels positively correlating with PAD severity. Myeloid cells enriched at CC sites increased AIBP expression postischemia. Genetic deletion of AIBP expanded CXCR4⁺ capillary endothelial cells (CECs) with stemlike and proliferative properties that remodeled into functional collaterals, a process blocked by CXCR4 inhibition. Mechanistically, AIBP bound the endocytic receptor LRP2 to promote endothelial uptake of high-density lipoprotein (HDL)–associated miR-223, a repressor of CXCR4. Disruption of this AIBP–LRP2–HDL–miR-223 axis restored CXCR4 and rescued CC growth. These findings define a two-phase mechanism in which stemlike CECs first expand and then transition to arterial fates, establishing a therapeutic strategy for revascularization in ischemic vascular disease. __________________________________________________________________ Targeting the AIBP–LRP2–miR-223 axis reprograms endothelial cells to augment collateral circulation in ischemic disease. INTRODUCTION Ischemic vascular disease, such as peripheral artery disease (PAD), is a prevalent and debilitating condition that causes tissue ischemia in the lower extremities due to vascular narrowing or occlusion ([58]1). Although stenting is a common treatment approach, its long-term effectiveness is often limited by in-stent restenosis and diffuse vascular disease, underscoring the need for alternative strategies to improve blood flow ([59]2). Collateral circulation (CC), defined by the formation of anastomotic vessels that bypass occluded arteries, serves as a critical compensatory mechanism to restore tissue perfusion ([60]3–[61]5). The development of robust CC is associated with better clinical outcomes ([62]6), making it a promising therapeutic approach. However, the mechanisms underlying collateral vessel formation remain poorly understood, and no treatments are currently available to enhance CC. Traditional models of CC have primarily focused on arteriogenesis, the enlargement of preexisting collateral arteries ([63]7), and arterialization, the transition of capillary endothelial cells (CECs) into arterial endothelial cells (AECs) ([64]8). Although these processes are well documented, the capacity of CECs to form direct artery-to-artery connections has been recognized in a prior study ([65]8), yet the underlying mechanisms and physiological relevance of this phenomenon remain largely unknown. Further investigation into this alternative route of vascular remodeling is needed to fully elucidate its contribution to CC and potential for therapeutic targeting. Emerging evidence suggests that chemokine receptor type 4 (CXCR4) signaling plays a pivotal role in arterial development and CC formation ([66]9). Studies have demonstrated that CXCR4 regulates arteriogenesis in both developmental and pathological contexts ([67]10). For example, CXCR4 is critical for the reassembly of CXCR4^+ AECs during neonatal heart regeneration ([68]11) and guides the migration of endothelial cells (ECs) from venous to arterial vasculature during zebrafish development ([69]5, [70]12). Despite these advances, the molecular mechanisms driving CC formation in the adult lower extremities remain largely uncharted. We hypothesize that the extracellular microenvironment, shaped by tissue ischemia, plays an integral role in orchestrating CC. Ischemic injury and subsequent immune cell infiltration trigger cellular responses that reshape the secretome—a complex repertoire of proteins and signaling molecules—which can either facilitate or impede vascular remodeling ([71]13). To identify key regulators within the secretome, we performed unbiased proteomic analyses of plasma from patients with PAD and healthy controls, as well as muscle tissue from a murine model of femoral artery ligation (FAL), an established model to induce CC in the hindlimbs ([72]14). These analyses highlighted lipid metabolism as a critical pathway, with APOA1 binding protein (AIBP) (aka NAXE) emerging as a central player linking lipid metabolism to vascular remodeling. Our findings uncover a previously unappreciated role for AIBP, derived from local myeloid cells, in regulating proximal CC formation. Contrary to reported models that emphasize CXCR4^+ AEC–mediated reassembly ([73]11), we show that AIBP depletion promotes the expansion of procollateral CXCR4^+ CECs exhibiting a stemness signature. A subset of these stemlike CECs commits to an arterial fate and contributes to functional CC. This identified mechanism not only challenges existing paradigms but also underscores the therapeutic potential of targeting the AIBP pathway to enhance CC and improve clinical outcomes in PAD and other ischemic conditions. RESULTS Unbiased profiling of secretory proteins highlights alterations in lipid metabolism We hypothesized that the tissue environment, shaped by secretory components, orchestrates vascular remodeling in ischemic disease. To identify secreted proteins altered in PAD, we performed a proteomic analysis of patient plasma ([74]Fig. 1A). We detected a total of 1337 proteins in both healthy individuals and patients with PAD (table S1). Among these, 664 (fold change ≥ 1.2) were increased and 329 (fold change ≤ 0.83) were reduced in PAD plasma versus normal plasma ([75]Fig. 1B). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the up-regulated proteins revealed enrichment in cholesterol metabolism ([76]Fig. 1C). Reanalysis of RNA sequencing (RNA-seq) data from a prior study ([77]15) yielded consistent findings (fig. S1, A and B). Among the proteins identified within the 10 most significantly enriched pathways linked to lipid metabolism (table S2), AIBP, a key regulator of vascular function ([78]16–[79]18), was markedly up-regulated in the plasma of patients with PAD ([80]Fig. 1D). This plasma increase was mirrored by higher AIBP levels in ischemic muscle tissue from patients with PAD ([81]Fig. 1, E and F). In contrast, other lipid metabolism–related proteins such as FABP5 and APOC4 showed no corresponding change in ischemic tissue (fig. S2, L and M). The concordance between plasma and tissue AIBP levels points to a potential direct role in ischemic tissue, prompting us to focus on AIBP in subsequent investigations. Subsequent analysis of AIBP protein expression in an independent cohort of patients with PAD (basic characteristics in table S3) confirmed a significant increase compared to healthy controls ([82]Fig. 1G). Notably, AIBP levels exhibited a negative correlation with the ankle-brachial index (ABI), where higher ABI values (<1.4) typically indicate better outcome of PAD ([83]Fig. 1H). Consistent with this, AIBP levels were reduced in the patients with PAD with longer painless walking distance (>50 m) ([84]Fig. 1I). Fig. 1. Unbiased proteomic analysis identifies lipid metabolism and AIBP association with PAD. [85]Fig. 1. [86]Open in a new tab (A) Experimental design of proteomic profiling of plasma derived from healthy individuals and patients with PAD. (B) Volcano plot showing significantly up-regulated (red) and down-regulated (blue) proteins in PAD versus healthy controls. The representative lipid related proteins are indicated. (C) KEGG pathway enrichment analysis of up-regulated proteins (≥1.5-fold change). (D) Heatmap displaying significantly up-regulated proteins (≥1.5-fold) associated with vascular function in patients with PAD (n = 3 healthy; n = 3 PAD). (E and F) Representative Western blot analysis (E) and quantification (F) of AIBP protein in less ischemic versus ischemic muscles from patients with PAD (n = 8). (G) ELISA-based quantification of plasma AIBP levels in healthy individuals (n = 20) and patients with PAD (n = 48). (H) Pearson correlation between plasma AIBP levels and ABI, with correlation coefficient (R) calculated using GraphPad Prism 10. The R value reflects the strength and direction of the relationship. (I) ELISA analysis of plasma AIBP levels in patients with PAD stratified by pain-free walking distance. (J) Plasma AIBP levels in patients with PAD with collateral category 1 (n = 20) versus category 2 (n = 17), assessed by ELISA. (K) Time-course analysis of plasma AIBP levels in B6 mice following FAL (n = 3 per time point; each sample pooled from plasma of three mice). Means ± SEM; *P < 0.05; **P < 0.01; n.s., not significant. We further evaluated collateral vessel formation in patients with PAD using computed tomography angiography (CTA) (fig. S1C), as previously reported ([87]19) (table S4). As expected, AIBP levels were lower in patients with more collaterals (category 2) compared with those with fewer collaterals (category 1) ([88]Fig. 1J). To investigate whether AIBP levels differ by sex, we performed enzyme-linked immunosorbent assay (ELISA) and found similarly elevated AIBP levels in both male and female patients with PAD compared with their respective controls (fig. S1, D and E). The negative correlation between AIBP and ABI (fig. S1, F and G), as well as the association between lower AIBP and improved walking distance (fig. S1, H and I), was consistently observed in both sexes. To determine whether these pathways are associated with CC, we subjected C57BL/6J (B6) mice to FAL. The adductor muscles, which harbor abundant collateral vessels, were harvested from the ischemic and control limbs for label-free proteomic analysis (fig. S2A). Our data revealed 849 up-regulated proteins (fold change > 1.5) in FAL muscles compared to controls (table S5), with 140 of the 1730 proteins being uniquely detected in the FAL group (fig. S2B). Among the total differentially expressed proteins, 16.6% were classified as secretory proteins according to curated human secretome database (fig. S2C, top). In support of our hypothesis that extracellular environment dictates CC, we found a profound increase in secreted proteins in the ischemic adductors versus the control adductors (50.2%; fig. S2C, bottom). Consistent with findings in humans, KEGG pathway analysis of these up-regulated proteins revealed enrichment in lipid metabolism pathways, including cholesterol metabolism and peroxisome proliferator–activated receptor (PPAR) signaling (fig. S2D). GO term analysis further identified enrichment in secretion-related categories such as extracellular region/component and secretory granule (fig. S2E). Twenty-eight proteins within the lipid metabolism pathway exhibited >1.2-fold increase (fig. S2F). Notably, plasma AIBP levels were also elevated in mice following FAL ([89]Fig. 1K). This ELISA-based method was validated using AIBP knockout mice, in which AIBP was undetectable (fig. S2G). This up-regulation of AIBP occurs predominantly at the translational or posttranscriptional level rather than the transcriptional level, as evidenced by increased protein levels detected by Western blot without corresponding changes in mRNA levels observed by quantitative polymerase chain reaction (qPCR) analysis (fig. S2, H to K). In agreement with the results from human PAD tissue analysis, FABP5 and APOC4 expression remained unchanged in the ischemic hindlimb tissues of mice (fig. S2, N and O). Together, our data from both patients with PAD and FAL mouse model indicate that the lipid metabolism pathway, in particular AIBP expression, is significantly increased in ischemic tissue, with AIBP levels mirroring the disease severity. Extracellular AIBP is derived from leukocytes post-FAL and its up-regulation by hypoxia A prior study assessed AIBP secretion using cell surface AIBP as a surrogate marker ([90]20). To identify the tissue source of extracellular AIBP, we isolated ischemic adductor muscles post-FAL and prepared a single-cell suspension for flow cytometry analysis. We surveyed eight cell types within the ischemic tissues, including fibro-adipogenic progenitors (FAPs), ECs, vascular smooth muscle cells (VSMCs), muscle stem cells (MUSCs), pericytes, skeletal muscle cells, macrophages, neutrophils, and monocytes. Compared to corresponding cell types in unoperated controls, a substantial elevation of cell surface AIBP was observed in monocytes (1.5-fold increase) and neutrophils (5.6-fold increase) following FAL ([91]Fig. 2, A to D). Given that hypoxia is a key feature of ischemic injury, we examined its effect on AIBP expression in THP-1, a human monocyte cell line. Western blot analysis revealed a time-dependent increase in AIBP expression under hypoxic conditions ([92]Fig. 2E), which was corroborated by flow cytometry ([93]Fig. 2, F and G) and ELISA of culture media ([94]Fig. 2H). The sequential up-regulation of intracellular, cell surface, and secreted AIBP suggests an expected route of AIBP extracellular transport. These findings were replicated in murine J774A.1 monocyte cell line (fig. S2, P and Q), further supporting a myeloid origin for AIBP under ischemic conditions. Fig. 2. AIBP originates from leukocytes following FAL, and its expression is up-regulated by hypoxia. [95]Fig. 2. [96]Open in a new tab (A) Representative flow cytometry analysis of AIBP expression in various cell types isolated from adductors on day 7 post-FAL, including Sca-1^+CD31^−CD45^− FAPs, CD31^+CD45^− ECs, SMA^+CD31^−CD45^− VSMCs, VCAM1^+CD31^−CD45^− MUSCs, Desmin^+CD31^−CD45^− cells (skeletal muscles, SKM), and NG2^+CD146^+ pericytes. (B) Representative flow cytometry analysis of AIBP expression in F4/80^+CD31^−CD45^+ macrophages, Ly6G^+CD11b^+CD31^−CD45^+ cells (neutrophils), and Ly6c^+CD11b^+CD31^−CD45^+ cells (monocytes) isolated from mice adductors on day 7 post-FAL. (C) Quantification of AIBP^+ cells across the populations shown in (A) and (B). Data represent n = 3 independent experiments, with each sample consisting of adductors pooled from five mice. (D) Representative flow cytometry analysis of AIBP expression in Apoa1bp^−/− monocytes (left) and AIBP fluorescence minus one (FMO) control in wild-type monocytes (right). Cell viability was assessed using the Zombie Aqua Fixable Viability Kit. n = 3, with each adductors pooled from five mice. (E) Immunoblotting analysis (top) and quantification (bottom) of AIBP expression in THP-1 following hypoxia (1% O[2]) treatment for the indicated durations. n = 7 repeats. h, hours. (F and G) Representative flow cytometry analysis (F) and quantification (G) of AIBP expression in THP1 cells exposed to hypoxia. n = 6 independent experiments. IgG, immunoglobulin G; FITC, fluorescein isothiocyanate; a.u., arbitrary units. (H) ELISA of AIBP concentration in the culture medium of THP1 cells exposed to hypoxia. n = 3 independent experiments. Means ± SEM; *P < 0.05; **P < 0.01; ****P < 0.0001; n.s., not significant. To assess the functional relevance of extracellular AIBP in CC, B6 mice were administered intravenous recombinant AIBP or control bovine serum albumin (BSA) following FAL. We performed micro–computed tomography (micro-CT) analysis to quantify the hindlimb arterial network in the ligated adductors (fig. S3A). We found that AIBP treatment markedly reduced arterial interconnections, as evidenced by decreased vascular volume and impaired growth of collateral vessels (fig. S3, B and C). Immunofluorescence staining of α–smooth muscle actin (α-SMA) further validated these findings (fig. S3, D and E). Collateral artery growth requires EC proliferation. To analyze this, we performed Ki67 immunostaining on the ischemic adductors. Compared to BSA, AIBP delivery suppressed the endothelial proliferation in the ischemic muscle (fig. S3, F and G). Laser Doppler imaging revealed ~70% recovery of arterial flow in the BSA-treated mice by day 21, whereas flow recovery was significantly impaired in the AIBP-injected mice (fig. S3, H and I). Consequently, mice receiving AIBP exhibited greater gastrocnemius necrosis (fig. S3, J and K) and worse mobility (fig. S3, L and M), and this was analyzed using (i) Tarlov score that assesses motor function and (ii) modified ischemia score that evaluates the severity of ischemia (table S6) ([97]21). These findings indicate that elevated extracellular AIBP limits collateral growth and exacerbates vascular and functional deficits following ischemic injury. Identification of AIBP-regulated stemlike CXCR4^+ CEC cluster following FAL To elucidate how AIBP depletion promotes CC, we performed single-cell RNA sequencing (scRNA-seq) on unsorted single-cell suspensions from Apoa1bp^−/− and control adductors at day 7 post-FAL (fig. S4A), an approach that minimized tissue processing–induced transcriptome change. After excluding low-quality cells on the basis of mitochondrial gene content (fig. S4, B and C), we retained 12,391 control and 8359 Apoa1bp^−/− cells. ECs constituted ~3.6% of total cells, consistent with normal muscle vasculature. T-distributed Stochastic Neighbor Embedding (t-SNE) analysis also identified fibroblasts and immune cells (fig. S4D). Differentially expressed gene (DEG) analysis [Log[10](Fc) > 0.25] revealed that Apoa1bp deficiency up-regulated Icam1, Icam2, and Vcam1 (fig. S4, E and F), suggesting an enhanced immune response ([98]22). Apoa1bp^−/− ECs also showed increased expression of arterial markers ([99]23) Gja4, Gja5, and Gjc1 (fig. S4, G and H). NOTCH1 is a well-established regulator of arterial specification. Although Notch1 expression was reduced in Apoa1bp^−/− ECs, expression of its canonical downstream targets—Hes1, Hey1, and Hey2—remained largely unchanged (fig. S4, I and J), implying a NOTCH1 signaling–independent mechanism. We next focused on EC subpopulations. T-SNE analysis identified four distinct EC clusters ([100]Fig. 3A), each defined by unique marker genes ([101]Fig. 3B). Two clusters were CECs (CEC1 and CEC2), distinguished by Plvap ([102]22) in CEC1 and Cxcl10/Cd52 ([103]24) CEC2, with the latter also expressing proliferation markers Ki67 and Pcna. Cluster 3 comprised AECs bearing arterial markers Sema7a and Efnb2 ([104]24), whereas cluster 4 included venous ECs (VECs) enriched in Nr2f2 (CoupTFII) (fig. S4K). Fig. 3. scRNA-seq analysis reveals the crucial role of AIBP in collateral vessel growth. [105]Fig. 3. [106]Open in a new tab (A) Unbiased clustering of 944 ECs isolated from adductor muscles of control and AIBP knockout mice using scRNA-seq and t-SNE dimensionality reduction. Four transcriptionally distinct EC subpopulations were identified: AECs, VECs, CECs, and LECs. (B) Heatmap displaying the top 50 marker genes for each EC cluster. (C) t-SNE visualization of individual ECs. Cyan depicts control ECs, and yellow marks show Apoa1bp^−/− ECs. (D) Quantification of the CEC2 subpopulation in control versus Apoa1bp^−/− ischemic adductors. (E) Violin plots depicting Cxcr4 mRNA expression across EC clusters. (F) Stemness-associated gene list in CEC2 based on our ChatGPT analysis. (G) Upstream regulator analysis in CEC2 using Ingenuity Pathway Analysis. FDR, false discovery rate. (H) Box plot showing stemness and proliferation scores across EC clusters. (I) Stemness analysis of the CEC2 cluster performed using CytoTRACE. (J) Bubble plot depicting enriched metabolic pathways in CEC2. (K) Pseudotime trajectory analysis of AEC, CEC1, CEC2, LEC, and VEC clusters to infer lineage relationships. (L) CellChat analysis indicating strong communication between macrophages/neutrophils and the CEC2 cluster. (M) Probability of cell-cell interactions between macrophages/neutrophils and EC clusters (AECs, CEC1, CEC2, and VECs) predicted by CellChat. (N) Ligand-receptor interaction analysis between macrophages/neutrophils and specific EC subsets. Means ± SEM; ****P < 0.001. Apoa1bp deficiency doubled the proportion of CEC2 ([107]Fig. 3, C and D), which expressed the stemness marker Cxcr4 ([108]Fig. 3E). Cell identity analysis using ChatGPT-based algorithms ([109]25) identified 14 stemness-associated genes enriched in CEC2 ([110]Fig. 3F). Upstream regulator analysis highlighted MYC as the top driver ([111]Fig. 3G), consistent with its known role in driving stem cell proliferation and metabolic reprogramming. CEC2 also exhibited a proliferative signature ([112]Fig. 3H). Cell fate assessment revealed that both CECs and VECs exhibited more stemlike characteristics compared to AECs or lymphatic endothelial cells (LECs) (fig. S4L). Intriguingly, mechanistic target of rapamycin (mTOR) signaling was suppressed as rapamycin and Torin1 emerged among the top regulators. Given mTOR’s role in restraining autophagy and promoting anabolic metabolism, its inhibition may facilitate autophagy-driven repair and support a metabolically adaptive, proregenerative EC phenotype in the ischemic settings ([113]26–[114]28). In addition, p53 also emerged as an upstream regulator, consistent with its role in stress-induced angiogenesis ([115]29) and mTOR adaptation ([116]30). AIBP knockout significantly elevated the stemness score ([117]Fig. 3I) and cell cycle score (fig. S4M). In line with this, flow cytometry analysis revealed a significant increase in c-Kit^+CXCR4^+ ECs in AIBP-deficient mice post-FAL (fig. S4N). Genes related to lipid metabolism were also up-regulated in the CEC2 cluster ([118]Fig. 3J), paralleling our proteomic findings. Pseudotime analysis suggested a differentiation trajectory from CEC2 to CEC1 and ultimately to AECs, with a minor contribution to VECs ([119]Fig. 3K and fig. S4O), suggesting that these stemlike capillary cells transition toward arterial fates during CC. Along this trajectory, lipid metabolism genes remained dynamically regulated (fig. S4P), implying that CEC2 exhibits proliferative, proregenerative, and metabolic adaptations to form functional CC. Cell-cell cross-talk plays a critical role in CC. CellChat analysis revealed preferential cross-talk between immune cells—particularly macrophages and neutrophils—and the CEC2 subpopulation, surpassing their interactions with other endothelial subtypes ([120]Fig. 3, L and M). In line with a prior study ([121]11), our receptor-ligand analysis confirmed the interaction between macrophage-derived CXCL12 and CXCR4 in AECs. Furthermore, we identified migration inhibitory factor (MIF) as the key ligand from macrophages binding to CXCR4 in CEC2 ([122]Fig. 3N), consistent with MIF’s role in regulating endothelial progenitor cell function and neovascularization ([123]31). These findings are consistent with the inflammatory signature associated with CEC2 as lipopolysaccharide was identified as one of the upstream regulators ([124]Fig. 3G). Overall, these data suggest that AIBP-regulated stemlike CECs play a pivotal role in driving CC through mechanisms that involve immune cross-talk, metabolic rewiring, and mTOR suppression. Apoa1bp depletion increases formation of small-sized collateral vessels in the ischemic hindlimbs We created Apoa1bp knockout mice to investigate the functional role of AIBP ablation in CC (fig. S5, A and B). To determine whether AIBP governs formation of preexisting collateral vessels, we performed micro-CT analysis to quantify arterial number and diameter ([125]Fig. 4, A and B) and conducted SMA immunostaining in the adductor muscles (fig. S5, C and D) of unoperated control and Apoa1bp^−/− adult mice. Both groups exhibited comparable collateral vessel profiles, consistent with the quiescent nature of vasculature in healthy adults, which limits the detectability of active collateral formation. In contrast, vascular remodeling is more prominent in the early postnatal period ([126]32). We therefore expanded our investigation to postnatal day 6 (P6) mice at the well-established anatomical locations. Specifically, we analyzed interconnected vessels between the proximal caudal femoral artery (PCFA) and the saphenous artery (SA) in the adductor muscles and still found a similar number of collateral vessels (fig. S5, E and F). These findings indicate that AIBP loss does not affect collateral vessel development under baseline, nonischemic conditions in either adult or early postnatal mice. Fig. 4. AIBP deficiency impairs hindlimb collateral vessel formation. [127]Fig. 4. [128]Open in a new tab (A and B) Representative micro-CT images (A) and quantification (B) of vessels <100 μm in diameter within the region of interest (ROI) from nonischemic limbs of Apoa1bp^−/− and control littermates. 3D rendering images are shown. n = 3 mice per group. Scale bar, 1 mm. (C and D) Representative micro-CT images (C) and quantification (D) of vessel volumes across various size ranges within the ROI from ischemic limbs on day 21 post-FAL. Yellow arrowheads indicate collateral arteries. n = 5 mice per group. Scale bar, 0.5 mm. (E and F) Representative confocal images (E) and quantification (F) of CD31^+ (green) and SMA^+ (red) arteries in adductors from Apoa1bp^−/− and control mice on day 21 post-FAL. n ≥ 5 mice per group. Scale bars, 20 μm. DAPI, 4′,6-diamidino-2-phenylindole. (G and H) Representative confocal images (G) and quantification (H) of CD31^+ (red) and Ki67^+ (green) cells in ischemic adductors. Arrowheads indicate proliferating ECs (CD31^+Ki67^+). n ≥ 5 mice per group. Scale bars, 20 μm. (I and J) Tarlov scores (I) and modified ischemic scores (J) of Apoa1bp^−/− mice and control littermates at the indicated time points following FAL. Scoring details are provided in table S6. n = 5 at each time point. Data are shown as means ± SEM. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; n.s., not significant. However, by day 21 after FAL, AIBP knockout muscle demonstrated a significant increase in the volume of small-diameter collateral arteries (lumen: 20 to 100 μm), as evidenced by micro-CT imaging ([129]Fig. 4, C and D), whereas larger vessels (>100 μm) remained unchanged between the two groups. This increase in small collaterals was accompanied by greater arteriole density (SMA^+) and enhanced EC proliferation (Ki67^+) in the ischemic adductors of Apoa1bp^−/− mice ([130]Fig. 4, E to H). Correspondingly, AIBP knockout mice manifested clinical improvements, as indicated by improved modified ischemia scores and enhanced Tarlov scores ([131]Fig. 4, I and J). Although preexisting collaterals in the adductor muscles can undergo enlargement and remodeling to support flow recovery, we applied a canonical flow–elicited arteriogenesis model ([132]33) (fig. S5G) and observed no enlargement in the diameter and the volume of collaterals (fig. S5, H to J) and no increase in blood perfusion (fig. S5, K and L) as well as Tarlov and modified ischemia scores in Apoa1bp^−/− mice (fig. S5, M and N). These effects of Apoa1bp deficiency on collateral formation were also reproduced in female mice (fig. S6, A and B), which showed increased collateral number but not areas, as indicated by immunostaining of adductor cross sections (fig. S6, C and D). To confirm that extracellular AIBP regulates collateral vessel formation, we delivered intravenous recombinant AIBP to the Apoa1bp^−/− mice. This treatment abolished the enhanced perfusion recovery in Apoa1bp^−/− mice (fig. S7, A and B), reduced arteriole density (fig. S7, C and D), and suppressed EC proliferation (fig. S7, E and F). These results collectively suggest that extracellular AIBP limits collateral artery formation. Loss of AIBP primers the expansion and arterial transition of stemlike CEC2 To analyze the tissue distribution of CEC2 post-FAL, we performed CXCR4 immunostaining in combination with detection of endomucin (EMCN) ([133]34), a capillary marker protein. We found that CXCR4 is predominantly enriched in CECs at day 7 postinjury, with a progressive decline by day 21 (fig. S8, A and B). Costaining for CXCR4 and SMA revealed a gradual increase in CXCR4^+ AECs over time, accompanied by rising SMA^+ arteriole density. Apoa1bp deficiency led to a marked increase in CXCR4^+ CECs at day 7, a difference that diminished by day 21 (fig. S8, A and B). In contrast, Apoa1bp^−/− mice exhibited increased CXCR4^+ AECs at day 21, with no significant difference at day 7 (fig. S8, C and D). We further investigated the EC subpopulations using flow cytometry analysis. Our flow cytometry gating is shown in fig. S8 (E and F). Stem cell antigen-1 (Sca-1), a marker of arterioles ([134]34), enabled distinction between CECs and AECs. On day 7, there were more CXCR4^+EMCN^+CD31^+ CECs and fewer CXCR4^+Sca-1^+CD31^+ arteriolar ECs, a pattern that reversed by day 21 in B6 mice (fig. S8G), indicating a dynamic shift in EC subpopulations over time. Accordingly, AIBP knockout increased the number of CXCR4^+ CECs at day 7 and arteriolar ECs at day 21 (fig. S8, H to J). Collectively, our data support a model where the stemlike CXCR4^+ CECs proliferate and convert to mature AECs, a process repressed by AIBP. To define the vascular identity of CXCR4^+ ECs in situ, we performed whole-mount immunostaining of the entire thigh muscle. Confocal imaging revealed that CXCR4 expression was primarily localized to the capillary network on day 7 post-FAL, with a transition toward mature arteries by day 21 in wild-type B6 mice (fig. S8K). As expected, Apoa1bp^−/− mice exhibited a significant up-regulation of CECs on day 7 and an increase in CXCR4^+ arterioles by day 21 post-FAL (fig. S8, L and M). Furthermore, we observed an increased number of proliferating CXCR4^+ CECs in AIBP knockout mice (fig. S8, N and O). In sum, these data suggest that AIBP absence augments CC formation following ischemic injury. To determine whether the stemlike CECs augments CC, CXCR4^+ CECs were isolated from the ischemic hindlimbs of donor mice 7 days post-FAL and subsequently transplanted intravenously into recipient mice immediately following FAL. Assessment by micro-CT and immunofluorescence revealed a significantly increased density of collateral vessels in recipients of CXCR4^+ CECs relative to those receiving CXCR4^− CECs (fig. S9, A to D). Furthermore, laser Doppler imaging demonstrated accelerated restoration of hindlimb blood flow‌ in the CXCR4^+ CEC cohort (fig. S9, E and F). Concomitant with these improvements, both Tarlov locomotor scores and modified ischemia scores exhibited significant enhancement (fig. S9, G and H). Collectively, these findings demonstrate‌ that CXCR4^+ CECs promote neovascularization and functional recovery following ischemic injury. CXCR4 signaling mediates the Apoa1bp deficiency effect on collateral formation CXCR4 drives artery reassembly by promoting EC migration ([135]11). To determine the effect of AIBP on CXCR4 signaling in vivo, we delivered AMD3100, a specific inhibitor of CXCR4 ([136]35), into control and Apoa1bp^−/− mice. The resulting phenotypes mirrored those observed in mice receiving recombinant AIBP. Specifically, AMD3100 administration abolished the enhanced perfusion recovery seen in the Apoa1bp^−/− mice following FAL (fig. S10, A and B). Furthermore, AMD3100 treatment impaired collateral formation in Apoa1bp^−/− mice, as evidenced by the lack of CC with vessel diameter less than 100 μm (fig. S10, C and D), a reduction of SMA^+ arteries (fig. S10, E and F), and a decrease in EC proliferation (fig. S10, G and H). Mice treated with AMD3100 also demonstrated weakened physical activities compared to either control or AIBP null mice receiving saline (fig. S10, I and J). In vitro, AIBP and high-density lipoprotein (HDL) in combination, but neither alone, inhibited endothelial proliferation and migration (fig. S10, K to N). To further evaluate CXCR4 dependency, we knocked down CXCR4 in human umbilical cord endothelial cells (HUVECs) using CXCR4 small interfering RNA (siRNA) (fig. S10O). Cotreatment with AMD3100 and CXCR4 siRNA did not produce additional inhibition of EC proliferation compared with either alone (fig. S10P; treatments 4 and 7 versus 8), suggesting that AMD3100 and siRNA converge on the same CXCR4-mediated pathway. Together, these results suggest that AIBP regulates endothelial behavior and ischemia-induced CC through CXCR4-dependent signaling. AIBP down-regulates CXCR4 via enhancing endothelial uptake of HDL-associated miR-223 Among known regulators of CXCR4 expression ([137]36, [138]37), miR-223 is notable for its association with HDL ([139]38). We found that miR-223 predominantly resides on HDL rather than non-HDL components (fig. S11A). In line with this, miR-223 abundance positively correlates with HDL levels (fig. S11B). Because our previous works established that AIBP enhances HDL binding to ECs ([140]16, [141]17), we hypothesized that AIBP facilitated cellular miR-223 delivery through HDL. To test this, we used both HUVECs and human microvascular endothelial cells (HMVECs), which align with the CEC subpopulation identified in our scRNA-seq analysis. Cells were incubated with Cy3-labeled HDL and Fam-labeled miR-223 in the presence or absence of AIBP. As expected, AIBP increased HDL uptake ([142]Fig. 5, A and B, and fig. S11, C and D) and significantly elevated intracellular miR-223 levels at 6 and 16 hours ([143]Fig. 5C). In contrast, free miR-223 alone failed to enter ECs, even in the presence of AIBP (fig. S11, E and F), indicating that HDL association is essential for intracellular miR-223 delivery. Notably, AIBP selectively increased mature miR-223 without elevating pre–miR-223 ([144]Fig. 5D), and transcriptional inhibition with actinomycin D had no effect on mature miR-223 levels ([145]Fig. 5D and fig. S11G), suggesting that the rise stems from uptake rather than de novo synthesis. Furthermore, RNase treatment of HDL reduced the miR-223 content ([146]Fig. 5E and fig. S11H), reinforcing the role of HDL as the delivery vehicle. Fig. 5. HDL–miR-223 mediates AIBP effects on ECs. [147]Fig. 5. [148]Open in a new tab (A and B) Representative confocal images (A) and quantification (B) of HDL uptake in HUVECs. n = 6 independent repeats. Each data point represents the average fluorescence area for each coverslip, calculated by summing the total fluorescence from all cells in randomly selected fields on a coverslip and dividing by the total cell number. Scale bar, 10 μm. (C) Quantification of miR-223 abundance. HUVECs were treated with AIBP, HDL, or their combination for the indicated durations. n = 3 independent repeats. (D and E) Quantification of mature miR-223 and precursor miR-223 (pre–miR-223) levels following coincubation with AIBP and HDL, in the presence or absence of actinomycin D (ACTD; 5 μg/ml) and RNase (20 μg/ml) for 2 hours (E). n = 3 independent experiments. (F) qPCR analysis of CXCR4 mRNA levels in HUVECs transfected with either miR-negative control (NC) or miR-223 (left) and anti–miR-NC or anti–miR-223 (right). n = 3 independent experiments. (G and H) Western blot analysis of CXCR4 (G) and quantification (H). HUVECs were transfected with miR-223 for the indicated times. n = 3 independent repeats. FC, fold change. (I) Immunoblotting of CXCR4 (top) and quantification (bottom). HUVECs were transfected with control or miR-223 or coincubated with HDL and AIBP followed by control or anti–miR-223 transfection. n = 3 independent experiments. (J) qPCR analysis of miR-223 levels. CXCR4^+CD31^+CD45^− ECs were sorted from ischemic control or Apoa1bp^−/− adductors on day 7 after FAL. The sorted cells were lysed using the miRNeasy Micro Kit to extract total RNA. n = 3 independent repeats. Each sample pooled from four mice. Data are expressed as means ± SEM. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; n.s., not significant; n.d., not determined. To determine whether miR-223 regulates CXCR4 expression in ECs, we performed gain-of-function and loss-of-function studies. Overexpression of miR-223 down-regulated CXCR4, whereas its inhibition up-regulated CXCR4 ([149]Fig. 5F). A brief 6-hour transfection of miR-223 alone significantly decreased CXCR4 protein levels ([150]Fig. 5, G and H). Similarly, incubating HUVECs with AIBP and HDL for 16 hours reduced CXCR4 expression (lanes 1 versus 2 in [151]Fig. 5I), and this effect was reversed by anti–miR-223 (lanes 3 versus 4 in [152]Fig. 5I). In vivo, CXCR4^+ ECs from AIBP knockout adductors exhibited reduced miR-223 levels ([153]Fig. 5J). The data validate endothelial CXCR4 as the direct target of miR-223 both in vitro and in vivo. To investigate the uptake mechanism, we blocked clathrin-dependent endocytosis with chlorpromazine (CPZ) ([154]39). CPZ lowered AIBP-mediated uptake of HDL and miR-223, as evidenced by their reduced fluorescence intensity (fig. S12, A and B), decreased intracellular mature miR-223 levels, and restored CXCR4 mRNA and protein level in HUVECs (lanes 3 versus 5 in fig. S12, C and D). In addition to clathrin, caveolae also mediates endocytosis of cell surface proteins, which can be disrupted by methyl-β-cyclodextrin (MβCD) ([155]40). In contrast to CPZ, MβCD treatment had no effect on miR-223 or CXCR4 levels in HUVECs treated with AIBP and HDL (lanes 3 versus 7 in fig. S12, C and D). CPZ alone recapitulated MβCD and CPZ coadministration effect (lanes 5 versus 8 in fig. S12, C and D). These data suggest that clathrin-mediated endocytosis is critical for AIBP-driven HDL–miR-223 internalization. Because CXCR4 localizes to lipid rafts ([156]41, [157]42), we examined whether AIBP-mediated raft disruption ([158]16, [159]43–[160]45) lowers CXCR4 levels. Treatment with AIBP and HDL for 16 hours reduced CXCR4 levels in lipid rafts (lanes 1 versus 4 in fig. S13, A and B), but this effect was reversed when HDL was pretreated with RNase (lanes 4 versus 5), suggesting that miR-223 underlies CXCR4 reduction. In contrast, shorter (4 hours) AIBP-HDL incubation also disrupted rafts, but eliminating miR-223 did not rescue CXCR4 levels (lanes 2 versus 3 in fig. S13, C and D). We further assessed lipid raft levels using fluorescently labeled CTB, which detects core raft lipid-GM1 gangliosides. Our data show that miR-223 levels did not change lipid raft abundance (fig. S13, E and F), when compared with MβCD treatment (fig. S13G). These data suggest that AIBP-driven lipid raft disruption and miR-223–mediated CXCR4 suppression are distinct processes, with the latter depending on clathrin-mediated uptake of HDL-bound miR-223. Lipoprotein receptor–related protein-2 is the endothelial receptor that mediates AIBP function in CC To elucidate the mechanism by which AIBP regulates CC, we used TurboID proximity labeling to identify AIBP-interacting proteins (fig. S14, A and B). Flow cytometry analysis confirmed that AIBP was significantly enriched on the EC surface after transfection with AIBP-TurboID (fig. S14, C and D). Following AIBP-TurboID overexpression and subsequent optimization (fig. S14, E and F), biotinylated proteins were isolated and analyzed by mass spectrometry. A total of 684 membrane-associated proteins were detected, and KEGG pathway analysis revealed enrichment in pathways related to endocytosis and fatty acid metabolism (fig. S14G). We chose to focus on lipoprotein receptor–related protein-2 (LRP2) (fig. S14H), given its established role in lipoprotein metabolism ([161]46). We validated the binding of AIBP to LRP2 using coimmunoprecipitation (Co-IP) ([162]Fig. 6A). To further confirm this interaction in situ, we performed proximity ligation assay (PLA), which detects protein-protein interactions via DNA probe-conjugated antibodies, producing fluorescent puncta when in close proximity. As expected, HMVECs incubated with recombinant AIBP exhibited significantly more PLA puncta compared to BSA-treated cells ([163]Fig. 6, B and C). Consistently, immunofluorescence staining showed colocalization of AIBP with LRP2 on the endothelial plasma membrane following recombinant AIBP treatment ([164]Fig. 6D). As anticipated, LRP2 deficiency attenuated AIBP-mediated disruption of lipid rafts (fig. S14, I to K, lanes 9 versus 10). Together, these data identify LRP2 as the endothelial receptor that binds AIBP. Fig. 6. LRP2 mediates AIBP binding and function. [165]Fig. 6. [166]Open in a new tab (A) Co-IP of AIBP-LRP2 interaction in HMVECs treated with recombinant AIBP. Lysates were immunoprecipitated with anti-LRP2 antibody and analyzed by Western blot as indicated. (B) Representative PLA confocal images showing AIBP-LRP2 binding. Scale bar, 10 μm. (C) Quantification of PLA puncta from (B); n = 3 mice. (D) Confocal images of AIBP (red), LRP2 (green), and CD31 (white) in HMVECs. n = 3 mice. Scale bar, 10 μm. (E) qPCR of Lrp2 mRNA in CD31^+CD45^− ECs from adductors of mice transduced with AAV9-shCtrl or AAV9-shLrp2. (F and G) Confocal images (F) and quantification (G) of LRP2 (green) and CD31 (red) in adductors following EC-specific AAV9-shCtrl or AAV9-shLrp2. n = 5 mice. Scale bar, 20 μm. (H and I) Micro-CT images (H) and vessel volume quantification (I) in ischemic limbs on day 21 post-FAL. Yellow arrowheads: collateral arteries. n = 5. Scale bar, 0.5 mm. (J and K) Flow cytometry (J) and quantification (K) of CXCR4^+EMCN^+ ECs in ischemic adductors. n = 3 samples (five mice pooled per sample). (L and M) qPCR of miR-223 (L) and Cxcr4 (M) in CD31^+CD45^− ECs from ischemic adductors (day 7 post-FAL). n = 8 to 12 mice per group. (N) Model: AIBP binds endothelial LRP2, triggering clathrin-mediated endocytosis and uptake of HDL–miR-223, leading to CXCR4 suppression. Means ± SEM; *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. LRP2 mediates the AIBP-facilitated HDL–miR-223–dependent down-regulation of CXCR4 Next, we assessed whether LRP2 mediates the effects of AIBP on HDL–miR-223 uptake. We knocked down LRP2 in HMVECs (fig. S15A) and assessed HDL-Cy3 uptake in the presence of AIBP. Our data showed that LRP2 knockdown significantly reduced cellular HDL uptake (fig. S15, B and C) and intracellular miR-223 levels (fig. S15, D and E). Notably, LRP2 deficiency rescued CXCR4 protein expression following incubation with AIBP and HDL (lanes 4 versus 5 in fig. S15, F and G), indicating that LRP2 is required for AIBP-HDL–induced CXCR4 suppression. To examine whether LRP2 mediates AIBP’s effects on CC, we generated an endothelial-specific adeno-associated virus (AAV)9–short hairpin RNA (shRNA) construct targeting endothelial LRP2 (AAV9-shLrp2), in which the Cdh5 promoter drives shLrp2 expression within an miR30 backbone ([167]47). Following intravenous delivery into B6 mice, AAV9-shLrp2, compared to control AAV9-shRNA, elicited EC-specific Lrp2 knockdown, as evidenced by qPCR analysis of Lrp2 in ECs isolated by fluorescence-activated cell sorting (FACS) ([168]Fig. 6E) and reduced vascular LRP2 protein in ischemic adductors ([169]Fig. 6, F and G), as confirmed by confocal microscopy analysis. Micro-CT analysis showed that endothelial LRP2 knockdown abrogated AIBP-mediated suppression of CC formation (lumen < 100 μm) ([170]Fig. 6, H and I). Consistent with this, Lrp2 knockdown restored the density of SMA^+ arterioles (fig. S16, A and B) and number of Ki67^+ ECs in the ischemic adductors following recombinant AIBP delivery (fig. S16, C and D). Accordingly, mice receiving AAV9-shLrp2 exhibited improved blood perfusion recovery following FAL (fig. S16, E and F). Flow cytometry demonstrated that AAV9-shLrp2 attenuated the AIBP-induced reduction of CXCR4^+ CECs (CXCR4^+EMCN^+CD31^+CD45^−) at day 7 post-FAL, resulting in increased CXCR4^+ CEC abundance ([171]Fig. 6, J and K), a finding corroborated by immunostaining (fig. S16, G and H). By contrast, no difference in the number of CXCR4^+ AECs (CXCR4^+Sca-1^+CD31^+CD45^− cells) was detected (fig. S16, I and J). Last, the AIBP-induced increase in miR-223 and reduction in CXCR4 levels were abolished by LRP2 knockdown but remained intact in control mice ([172]Fig. 6, L and M). Collectively, these data suggest that AIBP binds to LRP2 to promote the uptake of HDL-associated miR-223, leading to the down-regulation of CXCR4 in CECs ([173]Fig. 6N). Neutralization of extracellular AIBP by MM02 promotes collateral vessel formation The above data led us to investigate the impact of neutralizing extracellular AIBP on collateral formation. To this end, we generated a panel of murine anti-AIBP monoclonal antibodies (mAbs). The antibodies #2, #5, and #6 selectively recognized endogenous AIBP in wild-type but not AIBP-deficient muscle tissue (fig. S17A). Among these, MM02 uniquely disrupted AIBP binding to HUVECs (fig. S17B) and most effectively reversed AIBP-mediated suppression of HUVEC proliferation (fig. S17C), HDL uptake, and miR-223 delivery (fig. S17, D to F). Furthermore, preincubation with MM02 abolished the AIBP-mediated reduction of CXCR4 mRNA and protein in HMVECs compared to the control antibody (fig. S17, G to I). These findings established a functional role for extracellular AIBP in limiting CC formation. We next evaluated the therapeutic effect of MM02 on CC. Administration of MM02, but not a control antibody, significantly increased collateral artery volume by day 21 following FAL ([174]Fig. 7, A and B). Immunohistochemical analyses revealed a higher density of SMA^+ arterioles in MM02-treated mice ([175]Fig. 7, C and D), accompanied by a marked increase in Ki67^+ ECs by day 7 post-FAL ([176]Fig. 7, E and F). This proliferative response was paralleled by increased CXCR4^+ CECs in MM02-treated adductors at day 7 (fig. S18, A and B) and a subsequent rise in CXCR4^+ AECs observed by day 21 (fig. S18, C and D), when compared with control antibody-treated mice. Fig. 7. Systemic delivery of AIBP mAb MM02 enhances collateral vessel formation post-FAL. [177]Fig. 7. [178]Open in a new tab (A) Representative Micro-CT images showing collateral arteries (yellow arrowheads) in B6 mice treated with control or MM02. Scale bar, 1 mm. (B) Quantification of total vascular volume (≤100 μm in diameter) in the ROI. n = 5 mice. (C and D) Confocal images (C) and quantification (D) of CD31^+SMA^+ vessels in ischemic adductors on day 21 post-FAL. n = 11 to 13 mice. Scale bar, 50 μm. (E and F) Confocal images (E) and quantification (F) of Ki67^+CD31^+ proliferating ECs at day 7 post-FAL after control or MM02 treatment. Arrowheads indicate double-positive cells. n = 5 to 8 mice. Scale bar, 20 μm. (G) qPCR of miR-223 in CXCR4^+CD31^+CD45^− ECs from adductors on day 7 post-FAL. n = 3 (each pooled from four mice). (H and I) Representative laser Doppler images (H) and perfusion ratios between injured and control limbs (I) on day 21 post-FAL. n = 7 mice. (J and K) Tarlov (J) and ischemic scores (K) at indicated time points post-FAL. n = 7 mice. (L and M) Representative hematoxylin and eosin (H&E) images (L) and necrotic area quantification (M) in ischemic adductors on day 21 post-FAL. n = 6 to 10 mice. Means ± SEM; *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; n.s., not significant. Furthermore, analysis of CXCR4^+ ECs isolated on day 7 revealed a significant reduction in miR-223 levels in the MM02-treated adductors ([179]Fig. 7G). MM02 treatment accelerated perfusion recovery ([180]Fig. 7, H and I), alleviated tissue ischemia and impaired mobility ([181]Fig. 7, J and K), and mitigated necrosis ([182]Fig. 7, L and M). Consistent with its extracellular mechanism of action, MM02 treatment significantly reduced plasma AIBP levels on day 7 and day 14 post-FAL (fig. S18E). These results suggest that neutralizing extracellular AIBP promotes collateral vessel development and functional recovery following ischemic injury. Previous studies have established that capillary remodeling mediates CC formation via fluid shear stress-dependent mechanisms, including the up-regulation of shear-responsive genes. In AIBP-deficient mice subjected to FAL, we observed a marked up-regulation of the shear-responsive gene Klf2 at postoperative day 14 (fig. S19, A and B), implicating shear stress as a predominant driver of collateral vessel maturation. Our above work demonstrated that Apoa1bp deficiency promotes expansion of the CXCR4^+ stemlike CEC subpopulation in the early stage of ischemia. To assess whether the enhanced collateral formation observed in AIBP-deficient mice at later stages depends on sustained shear stress, we established a model of complete iliac artery ligation (IAL) (fig. S19C), which eliminates distal hindlimb perfusion and thereby abolishes flow-mediated cues. Notably, although AIBP knockout mice exhibited an expanded CXCR4^+ CEC population on postoperative day 7 (fig. S19, D and E), neither AIBP-deficient nor control mice formed mature collateral arteries by day 21 (fig. S19, F and G). Together, these findings suggest that persistent fluid shear stress is essential for collateral artery maturation in AIBP-deficient mice during the later phase of ischemia. DISCUSSION In this study, we revealed the lipid metabolism pathway as a critical regulator of CC and identified previously unknown procollateral CEC type typical of stemness. All previously reported EC subtypes that contribute to arteriogenesis exhibit stemlike features, including both CECs and VECs. In contrast, LECs—primarily derived from VECs—show low stemness, comparable to AECs. Through complementary loss-of-function and gain-of-function approaches, we demonstrate that AIBP—a lipid metabolism protein up-regulated in the skeletal muscle of patients with PAD—acts as a negative regulator of CC formation. In the ischemic hindlimb, AIBP depletion enhances CC by promoting the expansion of CXCR4^+ CECs rather than opening preexisting collaterals. Our results support a two-phase model of CC formation (fig. S20): Phase 1—sprouting phase: an initial stage during which CXCR4^+ stemlike CEC2 subtype sprouts and establish interarterial angiogenic connections. This phase is enhanced by Apoa1bp deficiency. Phase 2—remodeling phase: a subsequent stage characterized by the progressive transition of interarterial capillaries into functional collateral arteries, involving the up-regulation of flow-responsive genes. During this process, the CEC2 subpopulation is converted to CEC1 and then to AECs. This trajectory is mechanistically distinct from the reassembly of fate-committed AECs previously proposed in the literature ([183]11). Notably, AIBP’s function is preferentially confined to the first phase as AIBP deficiency does not enhance CC in the classical arteriogenesis model. Therefore, the procollateralization effect of AIBP deficiency manifests specifically in the FAL model ([184]17), which uniquely couples angiogenesis with arteriogenesis, but is absent in models of classical arteriogenesis that rely preferentially on flow-induced remodeling ([185]33). Our observations align with reports supporting a role for CECs in arterial regeneration during ischemic tissue repair ([186]48). Although VEGF gene therapy has shown limited efficacy in clinical trials and is no longer actively pursued as a therapeutic strategy, our study identifies an alternative and potentially more targeted approach. By uncovering the AIBP–miR-223–CXCR4 regulatory axis that restricts capillary-to-artery transition, we propose a mechanism-driven strategy that may offer greater precision in enhancing collateral growth, particularly in patients with impaired arteriogenesis. We show that AIBP suppresses the sprouting phase of CC by facilitating HDL-mediated delivery of miR-223 via LRP2, which down-regulates CXCR4 expression and restricts CEC2 expansion. From an evolutionary perspective, AIBP-driven suppression of CC may serve to limit excessive vascular remodeling, which could otherwise predispose tissues to aberrant vessel formation and instability ([187]49–[188]52). Neutralization of AIBP using a mAb enhanced collateral network formation and accelerated perfusion recovery. Given that the antibody targets extracellular AIBP, it is expected to exert its effects without disrupting AIBP’s intracellular functions ([189]53, [190]54). In this study, we identified neutrophils and monocytes as the predominant sources of AIBP during the early phase of ischemic injury, and F4/80^+CD11b^+ macrophages also express AIBP, albeit at lower levels compared to neutrophils and monocytes. The temporally distinct expression patterns among myeloid subsets highlight the dynamic regulation of AIBP in response to ischemic stress and underscore its potential multifaceted role across different stages of the immune response. Our study reveals a moonlighting role for HDL as an miR-223 carrier in CC, distinct from its canonical role in lipid transport. This noncanonical function is amplified by myeloid cell–derived AIBP localized at CC sites ([191]55, [192]56), reinforcing the emerging view that myeloid-derived factors are critical orchestrators of extracellular signaling networks during CC. These myeloid cells likely coordinate CC establishment by modulating the extracellular environment. Although traditionally viewed as “good cholesterol” ([193]57, [194]58) emerging evidence suggests that HDL exhibits functional heterogeneity and may exert context-dependent effects on vascular biology. Our findings indicate that the HDL–miR-223 axis, particularly in the presence of elevated AIBP, may impair collateral vessel formation by facilitating miR-223 uptake into ECs. These results highlight the complex and potentially dualistic role of HDL in vascular remodeling and underscore the need for further investigation into HDL subtype-specific functions. In vascular development, arterialization is a distinct process that relies on NOTCH1-dependent cell cycle inhibition ([195]59), enabling the transition of proliferative ECs into a quiescent arterial fate. During the later stages of arterial growth and remodeling, CECs and VECs serve as a cellular reservoir ([196]22), contributing additional ECs to the expanding arterial network. We observed a similar transition in CC, where proliferative CEC2 give rise to less proliferative CEC1, consistent with the requirement for cell cycle inhibition during capillary arterialization. Our prior work showed that AIBP up-regulates the NOTCH1 pathway in the developing retinal vasculature ([197]17). In this study, AIBP depletion led to reduced Notch1 expression in ECs of ischemic tissue, although expression of canonical downstream targets (Hes1 and Hey1/2) remained unchanged, suggesting a context-dependent modulation of NOTCH signaling. Of note, bioinformatic analysis nominated transforming growth factor–β1 (TGF-β1) as a top upstream regulator of CEC2 gene expression. TGF-β1 may enhance NOTCH activity by up-regulating Ephrin-B2, NOTCH4, and HEY family members or by remodeling the extracellular matrix to facilitate arterial specification ([198]60–[199]62). Collectively, our findings define a two-stage model of collateral vessel formation that relies on the dynamic expansion and fate transition of CXCR4^+ CECs. Our studies reveal extracellular AIBP as a key repressor of this process and highlight its potential as a therapeutic target to augment revascularization in ischemic disease. Limitation Although APOC4 and FABP5 were also identified in our initial screening, their functional roles in CC were not examined in depth; future work using different ischemic stages, genetic models, and assessments of cross-organ communication, expanded cohorts, extended time points, and genetic models is needed to clarify their potential contributions. Although MM02 demonstrates therapeutic potential, its clinical translation may be constrained by the requirement for repeated antibody administration. To overcome this, future efforts will explore Fc engineering, sustained-release platforms, and site-specific delivery strategies to enhance pharmacokinetics and patient adherence. In terms of outcome assessment, although the modified ischemia score offers clinical practicality, it remains semiquantitative and may not fully differentiate true tissue regeneration from superficial improvements due to necrotic tissue retraction or desiccation. Our data support a potential fate transition of stemlike CECs into AECs; however, this conclusion remains inferential in the absence of direct lineage-tracing evidence. Future studies incorporating genetic fate-mapping strategies specific to these endothelial subpopulations are necessary to rigorously define this trajectory. Although direct artery-to-artery collateral formation has been observed in preclinical models, current clinical imaging modalities lack the spatial resolution and molecular specificity to confirm this mechanism in patients. Emerging technologies—including single-cell transcriptomics, spatially resolved profiling, and AI-assisted imaging—may enable precise characterization of collateral vessel formation in human tissues. These innovations are poised to bridge the translational gap between mechanistic discovery and clinical application. MATERIALS AND METHODS Mass spectrometric analysis of the TurboID proteins Peptides were loaded onto a capillary and analyzed using the timsTOF Pro (Bruker Daltonics) mass spectrometer. The electrospray voltage was set to 1.60 kV. Precursors and fragments were detected with a scan range of 100 to 1700 m/z (mass/charge ratio) in the time-of-flight (TOF) detector. The timsTOF Pro operated in PASEF mode, selecting precursors with charge states of 0 to 5 for fragmentation, with 10 PASEF-MS/MS scans acquired per cycle. Dynamic exclusion was set at 30 s ([200]63). Bioinformatics analysis Functional enrichment. KEGG pathway enrichment analysis was performed using the KEGG database. Fisher’s exact test was applied to analyze KEGG pathway enrichment significance for differentially expressed proteins (using identified proteins as the background), with a significance threshold of P < 0.05 ([201]64). Bulk RNA-seq analysis We analyzed the DEGs in the public bulk RNA-seq dataset ([202]15) using functional enrichment analysis as above described in the Bioinformatics analysis section. Single-cell RNA-seq Sample preparation We performed FAL surgery on four Apoa1bp^−/− mice and four control littermates. Ischemic adductor muscles were collected and stored in tissue storage solution (Miltenyi, 130-100-008) for subsequent RNA isolation and library preparation. Quality control and library preparation scRNA-seq was conducted by GENE DENOVO (China). Ischemic adductor muscles were digested, and the resulting single-cell suspension was loaded into the 10X Genomics GemCode Single-cell instrument to generate Gel Bead-In-Emulsion (GEMs). cDNA libraries were prepared using Chromium Next GEM Single Cell 3′ Reagent Kits v3.1. Upon GEM dissolution, barcoded cDNAs were reverse transcribed from polyadenylated mRNA. Silane magnetic beads were used to purify the cDNA, which was then amplified for library construction. The final library was sequenced using Illumina paired-end sequencing, with the 16–base pair (bp) 10x Barcode and 10-bp UMI encoded in Read 1 and cDNA fragments sequenced in Read 2. Sample index sequences were incorporated into the i7 index read. EC cluster identification and marker gene analysis Raw BCL files were converted to FASTQ files, aligned, and quantified using 10X Genomics Cell Ranger software (v.3.1.0). Gene matrices from individual samples were imported into Scanpy (v.1.9.1) for downstream analysis. The integrated expression matrix was scaled, and dimensional reduction was performed using principal components analysis (PCA). The number of principal components used was determined by examining their contribution to the total variance, using a screen test to identify the most influential components. Seurat was used for graph-based clustering, with distances between cells calculated on the basis of identified principal components. A shared nearest-neighbor (SNN) graph was constructed on the basis of Euclidean distances in PCA space, refining edge weights using the Jaccard distance. Clusters were identified using the Louvain method to maximize modularity. Gene expression within each cluster was compared to all other cells using the Wilcoxon rank sum test. Up-regulated genes were defined by a 1.28-fold increase, expression in more than 25% of cluster cells, and P value < 0.05. Genes were considered functionally related if involved in similar biological processes. Heatmaps were generated from cluster-averaged gene expression, row-wise z-score scaled, and visualized using the heatmaply R package (v.0.15.2). Cell cycle pseudotime analysis Cell pseudotime trajectories were analyzed using Monocle 3, reducing data to one dimension. The cell fate trajectory was visualized in a treelike structure, including branches and tips, with parameters sigma = 0.001, lambda = NULL, param.gamma = 10, and tol = 0.001. The resulting cell fate trajectory was visualized in reduced dimensional space and represented as a treelike structure, including tips and branches, to reflect the progression of cell states ([203]65). Pathway and transcription factor activity analysis Gene set variation analysis (GSVA) was used to convert the gene-by-cell matrix into a gene set–by–cell matrix. Transcription factor activity was analyzed using the SCENIC package (v.0.1.5), applying the 20,000 motif database for RcisTarget and GRNboost analysis. A normalized expression matrix of 14,917 filtered genes served as the input. Western blot analysis Western blotting was performed as previously described ([204]66). In brief, the adductor muscles were homogenized in protein lysis buffer (Sangon Biotech, catalog no. C500028-0010, China) on ice. HUVECs and HMVECs were lysed in Beyotime lysis buffer (catalog no. P0013B, China) with protease inhibitors. Protein concentration was measured using a BCA Assay Kit (Beyotime, catalog no. P0012, China). SDS–polyacrylamide gel electrophoresis (PAGE) and Western blotting were carried out according to standard protocols. PVDF (polyvinylidene difluoride) membranes were blocked in 5% skim milk in TBST [1X tris-buffered saline (TBS) with 0.05% Tween 20] and incubated with primary antibodies against AIBP (1:1000, homemade), CXCR4 (1:1000, Abcam, catalog no. 124824), glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (1:10,000, Abcam, catalog no.181602), CAV-1 (1:5000, Abcam), FLOT1 (1:200, sc-74566, Santa Cruz Bio), FLOT2 (1:1000, PTM-5369, PTM BIO), LRP2 (1:1000, A24690, ABclonal), and anti-rabbit (1:10,000, Abcam, catalog no. 6721) or anti-mouse (1:10,000, Abcam, catalog no.6789) secondary HRP (horseradish peroxidase) antibodies. Signals were detected using the SuperSignal West Pico PLUS Chemiluminescent Substrate (Thermo Fisher Scientific). AIBP binding assay The AIBP binding assay was performed as previously elaborated ([205]17). Recombinant human AIBP was first conjugated to Cy3–N-hydroxysuccinimide ester according to the manufacturer’s instructions (APEXBIO A8107, USA). The Cy3-conjugated AIBP was incubated with HUVECs on ice for 4 hours. The cells were subsequently washed twice with ice-cold phosphate-buffered saline (PBS), fixed with 4% paraformaldehyde (PFA) for 10 min, and then washed twice with ice-cold PBS. The fluorescence intensity was measured using a microplate reader (Biotek ELx800), and data were expressed as relative intensity per well. HDL isolation HDL and HDL3 were isolated as previously described ([206]67). Fresh plasma from healthy donors was obtained from Xiangya Hospital of Central South University. Plasma was adjusted to a density of 1.063 kg/liter using KBr and subjected to ultracentrifugation at 35,000 rpm for 12 hours at 4°C. Crude HDL was isolated and further purified by adjusting to densities of 1.125 and 1.21 kg/liter with KBr, followed by ultracentrifugation. The HDL3 fraction was collected and dialyzed against PBS. Purity was confirmed with Sudan black staining and agarose gel electrophoresis. The purity of HDL and HDL3 was verified with Coomassie blue staining and agarose gel electrophoresis ([207]67). Caveolar fraction isolation Caveolar fractions were isolated through a detergent-free, discontinuous gradient ultracentrifugation method as described before ([208]68). In brief, HUVECs were washed twice with ice-cold PBS and cells were scraped from the two D150 plates in 0.5 M sodium carbonate buffer (pH 11.0) containing protease inhibitor cocktail, homogenized, and sonicated on ice. Following protein quantification, protein extracts were mixed with MBS buffer [25 mM MES and 0.15 M NaCl (pH 6.5)] with 45% sucrose (w/v) in the ultracentrifugation tubes, and overlaid with 35 and 5% sucrose. After ultracentrifugation at 35,000 rpm for 20 hours at 4°C in an SW-41 rotor (Beckman), a discontinuous sucrose density gradient of 5 to 45% was generated. Eleven 1-ml fractions were collected from the top to the bottom. An equal amount of each faction was analyzed by SDS-PAGE using CXCR4 (Abcam, catalog no.124824) and Cav-1 (Abcam) antibodies. TurboID mediated biotinylation Turbo-ID assay was performed as previously described ([209]69). A pLenti-AIBP-TurboID-HA plasmid was prepared for lentiviral packaging. HUVECs in the logarithmic growth phase were cultured in 10-cm dishes. When the cell density reached 70 to 80%, the viral concentrate carrying the AIBP-TurboID-HA gene was added. After 24 hours of viral infection, the medium was replaced with a fresh medium. Over the next 7 days, the medium was changed every 2 days using a G418-containing medium (300 to 500 ng/ml) for selection, and cells were monitored daily. Cell lysates were collected for immunoblotting analysis to confirm successful transformation. Cells stably expressing AIBP-TurboID-HA were designated AIBP-HUVECs. For biotin labeling, AIBP-HUVECs were seeded into 6-well plates. Once they reached approximately 80% confluence, the medium was removed, and cells were washed twice with PBS and incubated with biotin-labeling buffer containing 500 μM biotin and 200 μM adenosine triphosphate (ATP). The medium was then replaced with complete medium containing the same biotin-labeling buffer. Cells were collected at the following time points: 0 and 10 min and 1, 6, and 12 hours. Protein lysates were analyzed via Western blot using a streptavidin-peroxidase antibody. For further experiments, AIBP-HUVECs were cultured in T75 flasks and divided into two groups: group I: cells cultured without additional treatment and group II: cells cultured in Dulbecco’s modified Eagle’s medium supplemented with 500 μM biotin and 200 μM ATP for 12 hours. Cells were collected, washed with cold PBS, and lysed in cold lysis buffer [8 M urea, 150 mM NaCl, 20 mM tris-HCl (pH 7.4), and 1% protease inhibitor cocktail] using cell scrapers. Lysates were sonicated and cleared by centrifugation (15,000g, 10 min, 4°C). For biotin pull-down, streptavidin magnetic beads were prewashed with radioimmunoprecipitation assay (RIPA) buffer (1 ml, 2 min) and the prepared protein lysates were incubated with the beads overnight at 4°C. Beads were washed twice with RIPA buffer (1 ml, 2 min), once with 1 M KCl (1 ml, 2 min), once with 0.1 M Na[2]CO[3] (1 ml, 10 s), once with 2 M urea in 10 mM tris-HCl (pH 8.0) (1 ml, 10 s), and twice more with RIPA buffer. Beads were not allowed to sit in Na[2]CO[3] or urea for extended periods to prevent aggregation. After the final wash, the beads were transferred to fresh tubes, washed twice with PBS, and stored at −20°C. Beads were then sent to OE Biotech Co. Ltd. (Shanghai, China) for mass spectrometry analysis. Coimmunoprecipitation Co-IP assay was done as previously described ([210]70). Briefly, HUVECs were incubated with AIBP (200 ng/ml) or BSA for 2 hours on ice. Cells were then lysed in the lysis buffer containing 1% Triton X-100, 1% NP-40, 0.5% sodium deoxycholate, 150 mM NaCl, 10 mM Tris (pH 7.4), 1 mM EDTA, 50 mM NaF, and a protease inhibitor cocktail. The LRP2 antibody was cross-linked using an IP kit (Thermo Fisher Scientific, Cat: 88805) following the manufacturer’s protocol. The prepared protein lysates were incubated with the LRP2 antibody-bound protein A/G resin overnight at 4°C. After three washes with lysis buffer, the bead pellets were incubated with sample loading buffer and prepared for Western blot analysis to detect bound AIBP. FAL model The surgery of FAL was performed in 9-month-old control or AIBP knockout mice as previously reported ([211]14) with minor changes. Briefly, surgical procedures were performed in mice under anesthesia and sterile conditions. Mice were injected with analgesic buprenorphine SR (0.15 mg/kg). Anesthesia was induced with 3% isoflurane and maintained with 1% isoflurane. Unilateral hindlimb ischemia was established by double ligation of the left femoral artery at both the proximal (near the inguinal ligament) and distal (just above the bifurcation of the saphenous and popliteal arteries) sites. The artery was transected between the two ligatures to ensure complete occlusion. All animal procedures adhered to the guidelines of National Institutes of Health for the care and use of laboratory animals and received approval from the Institutional Animal Care and Use Committee at Xiangya Hospital, Central South China University. AAV9-ShLRP2cdh5 construction and administration rAAV9 were constructed by the vendor General Biology, and AAV9-Cdh5-Mir30-m-shRNA (Ctrl) and AAV9-Cdh5-Mir30-m-shRNA (LRP2) were used ([212]71). This design enables endothelium-specific LRP2 knockdown. AAV9-ShLRP2cdh5 [1.0 × 10^11 vector genomes (vg)] or ShCtrl-AAV9cdh5 (1.0 × 10^11 vg) was injected via the tail vein. Two weeks later, ECs of the recipient mice were FACS sorted, and qPCR was performed to validate the Lrp2 knockdown efficiency. Micro-CT angiography Micro-CT analysis of the hindlimb vasculature was performed as described previously with modifications ([213]72). Briefly, mice were injected with heparin (2000 U/kg), 5 min before euthanasia. After confirming cessation of cardiac activity, mice underwent cardiac perfusion with vasodilation solution [Dulbecco’s phosphate-buffered saline (DPBS) containing 100 μM adenosine, 10 μM sodium nitroprusside, and heparin (20 U/ml)] to remove blood from the circulatory system. Mice were subsequently perfused with 4% PFA followed by an injection of 0.8-ml Microfil (MV-122, Flow Tech, Carver, MA) in the descending aorta. The specimens were stored in 4% PFA and kept at 4°C overnight to allow polymerization. Samples were scanned using a Bruker Micro-CT SkyScan 1176 system (Kontich, Belgium). Scan settings are as follows: voxel size, 6.534165 μm; medium resolution, 58 kV; 431 μA; 0.5-mm Al filter; and integration time, 1000 ms. Density measurements were calibrated to the manufacturer’s calcium hydroxyapatite (phantom). Analysis was performed using the manufacturer’s evaluation software. Reconstruction was carried out by NRecon (version 1.7.4.2). 3D images were obtained from contoured 2D images by methods on the basis of distance transformation of the grayscale original images (CT vox; version 3.3.0). 3D and 2D analyses were performed using the software CT Analyzer (version 1.18.8.0). 3D structural parameters, including vessel volume and vessel numbers, were analyzed. Laser Doppler measurement of blood flow Blood flow was measured by scanning both rear paws with an LDI analyzer (Moor Infrared Laser Doppler Imager Instrument, Wilmington, Delaware) at designated time points: before and after the surgical procedure (days −1, 0, 3, 7, 14, and 21). During the procedure, animals were kept under 1% isoflurane anesthesia on a heating pad. The level of perfusion in both the ischemic and nonischemic hindlimbs was quantified using the mean pixel value within the region of interest, and the relative changes in hindlimb blood flow were expressed as the ratio of the left (ischemic) versus right (normal) laser Doppler–detected blood perfusion ([214]73). Functional scoring Mice were examined preoperatively, immediately postsurgery following recovery from anesthesia, and at 1 and 2 weeks postoperation. Functional grading was performed according to the Tarlov scale and a modified ischemic scale designed to detect milder ischemia. Details of the functional and ischemic scoring criteria were elaborated in table S6 ([215]21). Statistical analysis Statistical analyses for [216]Figs. 1 to [217]7 were performed using GraphPad Prism 10, and results are presented as means ± SEM. Significance was determined using appropriate tests on the basis of data structure: unpaired two-tailed Student’s t tests with Welch’s correction ([218]Figs. 1, E, H, and K; [219]3I; [220]4, B, F, and H; [221]5F; [222]6, C, E, and G; and [223]7, D, F, G, and M), one-way analysis of variance (ANOVA) followed by Tukey’s ([224]Figs. 1G and [225]6, I and K to M), Dunnett’s ([226]Fig. 2, E and G to J), or Sidak’s post hoc tests ([227]Fig. 5, B, H, and I), and two-way ANOVA followed by Tukey’s ([228]Fig. 2C), Bonferroni’s ([229]Fig. 4, D, I, and J), or Sidak’s post hoc tests ([230]Fig. 7, I to K). Nonparametric tests with Dunn’s post hoc analysis were used for multiple comparisons ([231]Fig. 1I), and the two-stage step-up method of Benjamini, Krieger, and Yekutieli was used to control the false discovery rate in [232]Fig. 5 (C and J). Statistical significance is denoted as *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, with “n.s.” indicating no significance and “nd” indicating not determined. Other detailed experimental protocols and the source of reagents used in this study are available in the Supplementary Materials. Use of artificial intelligence (ChatGPT) We used ChatGPT (GPT-4, Open AI) for the capillary stemness score analysis. We used the following prompts in our analysis: “You are an expert in vascular biology and single-cell analysis. Score ‘capillary endothelial cell stemness’ for each gene I provide. Use a stemness score (1–10), with 10 being the highest and 1 the lowest.” Acknowledgments