Abstract
Objective:
Reelin and its receptor Apoer2 play a prominent role in endothelial cell dysfunction by promoting leukocyte–endothelial cell adhesion, an important component of the inflammatory process underlying atherosclerosis. We therefore hypothesized that pharmacological depletion of circulating Reelin represents a novel therapeutic strategy to impede the progression of atherosclerosis.
Approach and Results:
In vitro studies demonstrated that human plasma induced monocyte adhesion to endothelial cells, while Reelin-depleted plasma had no effect on monocyte adhesion. Signaling analysis revealed that Reelin activated Dab2, PI3K, Akt and NF-κB cascade to promote the expression of adhesion markers (E-selectin, ICAM-1, and VCAM-1). Intravital microscopy confirmed decreased leukocyte-endothelial adhesion in mice treated with Reelin antisense oligonucleotide (ASO). In vascular smooth muscle cells, Reelin induced Stat3 phosphorylation to promote cell proliferation, which is another hallmark of atherosclerotic plaque progression. To investigate if Reelin pharmaceutical depletion protects against atherosclerosis, low-density lipoprotein receptor–deficient (Ldlr−/−) mice fed with high cholesterol diet were treated with either Reelin ASO or neutralizing antibody (CR-50) to systemically deplete circulating Reelin. In both treatments, atherosclerotic plaque progression was markedly attenuated. These in vivo results suggest that Reelin depletion decreases vascular adhesion and inhibits the recruitment of monocytes and consequently prevents plaque progression.
Conclusions:
These findings suggest that Reelin inhibition may provide a novel therapeutic approach to counteract leukocyte or monocyte adhesion as well as extravasation and inhibit the progression of atherosclerosis. This strategy may also be relevant for other diseases that involve leukocyte or monocyte extravasation as a central pathological mechanism, such as multiple sclerosis or arthritis.
Keywords: Atherosclerosis, chronic inflammation, leukocyte, monocyte, endothelial permeability, endothelial adhesion, E-Selectin, ICAM-1, VCAM-1, Reelin, antibody
Graphical Abstract

INTRODUCTION
Endothelial dysfunction has been associated with the inflammatory process underlying the development of atherosclerosis 1. In endothelial cells (EC), the Apolipoprotein E receptor-2 (Apoer2 or Lrp8), a low-density lipoprotein receptor (LDLR) family member, is involved in inflammatory processes such as nitric oxide synthesis and regulation of endothelial cell surface expression of leukocyte adhesion molecules 1–4. Systemic Apoer2 deficiency has been associated with increased susceptibility to atherosclerosis in mouse models 5,6, and in human genomic studies 7–9. However, Apoer2 deletion in mice has also been demonstrated to have anti-thrombotic effects in the endothelium by reducing leukocyte-endothelial cell adhesion in response to antiphospholipid antibodies 10,11. We sought to better understand the cell-specific effects of Apoer2 in atherosclerosis by focusing on the effect of the Apoer2 ligand Reelin 4 in endothelial cells.
Originally described in the brain, Reelin guides neurons to their correct positions in the developing brain and modulates synaptic function in the adult brain 12–16. Outside the nervous system, we have previously shown that circulating Reelin promotes vascular adhesion and leukocyte infiltration by elevating the basal state of vascular immune surveillance 4,17. In the atherosclerosis-prone LDLR-deficient (Ldlr−/−) mouse model, Reelin was deleted ubiquitously or only in the liver, thus preventing its release into the circulation. In these two models, Reelin deletion reduced leukocyte-endothelial adhesion as well as the expression of the pro-inflammatory markers vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1). This was accompanied by decreased atherosclerosis progression and macrophage content in atherosclerotic lesions. It was confirmed in human endothelial cell (EC) cultures that Reelin increased monocyte adhesion as well as the expression of ICAM1, VCAM1, and E-selectin. Interestingly, Reelin action was mediated by its receptor Apoer2, followed by upregulation of nuclear factor κB (NF-κB) activity.
Together, these previous findings demonstrate that the Reelin-Apoer2 signaling cascade regulates leukocyte adhesion on the endothelium, thus promoting chronic inflammation and atherosclerosis progression. To lay the foundation for future therapeutic interventions, we tested whether reducing circulating Reelin, using antisense oligonucleotide (ASO) or neutralizing antibodies, would limit atherosclerotic plaque progression in the atherosclerosis-prone Ldlr−/− mouse model.
MATERIALS AND METHODS
Authors will make their data, analytic methods, and study materials available to other researchers upon request.
Cell culture.
Commercially available human aortic endothelial cells (HAEC; ATCC; PCS-100-011) were cultured in EBM-2 Medium (Lonza; CC-3156) supplemented with EGM-2 (Lonza; CC-4176) on petri-diches coated with gelatin. Commercially available human aortic smooth muscle cells (HASMC; ATCC; PCS-100-012) were cultured in SmGM-2 BulletKit medium (Lonza; CC-3182) for smooth muscle cells as previously described 18–20. The monocyte cell line U937 (human histiocytic lymphoma; CRL-1593; ATCC) was grown in RPMI 1640 medium (Sigma-Aldrich) containing 10% FBS. For in vitro experiments, all cells were used between passages 3 and 8.
Protein expression:
Cells were serum starved (1% fetal bovine serum (FBS)) overnight and then stimulated as described in the figure legends with doses tested in previous studies 4,17,18. Then, cells were washed and protein harvested in RIPA buffer.
shRNA transduction:
shRNA transduction was performed as previously described 18. shRNA lentivirus against Dab2 was purchase from Origen (TL313573).
Monocyte adhesion assay:
HAEC were seeded and grown in 6-well plate to form a confluent monolayer. Cells were serum starved (1% FBS) overnight and then stimulated as described in the figure legends, with the following components for 24 hours: human plasma (dilution 1/50 in starvation medium), human plasma depleted for Reelin with CR-50 immunoprecipitation (Dr. Katsuhiko Mikoshiba originally provided the CR-50 hybridoma). Then the cells were washed with PBS and incubated with human monocyte (U937) for 45min at 37°C with gentle rocking. Finally, the cells were washed and fixed with PFA 4%. The number of adherent monocytes was manually counted in 5 random areas per well, with at least six wells per condition.
Cx3cr1-GFP mice and Intravital microscopy for quantification of monocyte–endothelial cell adhesion.
Cx3cr1-GFP mice (B6.129P-Cx3cr1tm1Litt/J) were purchased from The Jackson Laboratory (Stock No. 005582). These mice express EGFP in monocytes, dendritic cells, NK cells, and brain microglia under the control of the endogenous Cx3cr1 locus. Cx3cr1-GFP monocytes downregulate GFP expression upon differentiation into macrophages.
For intra-vital microscopy, 4-week-old Cx3cr1-GFP males and females received one injection (25mg/kg, intra-peritoneally) of control or anti-Reelin ASO. Live imaging was then performed one week later.
After anesthesia with Ketamine/xylazine, the mesentery of 5-week-old Cx3cr1-GFP mice was exposed for observation and recording of images of GFP-monocyte adhesion and rolling using a Regita digital camera (QImaging) attached to a Nikon Eclipse Ti microscope with NIS Element image-capturing software. Per mice, videos on 6 different marginal mesenteric veins were recorded. The velocity and quantity of monocyte rolling were measured by ImageJ (wrmtrck plugin) on each video for 10 seconds. The rolling index was calculated as monocyte number / velocity for each record. For each 10-second-video, all the frames were stacked into one final picture and the intensity was measured to visually reflect and measure the monocyte exposition to the vascular wall.
After recording, the mice were sacrificed, blood was collected for white blood cell count and plasma analysis, the thoracic aorta was dissected out, the adventitia was removed and the media-intima was snap frozen for further protein analysis.
Cx3cr1-GFP; Ldlr−/− mice and atherosclerosis model.
Cx3cr1-GFP line was crossed with Ldlr−/− 21 to obtain the Cx3cr1-GFP; Ldlr−/− mice. To evaluate atherosclerosis severity, 8-week-old male and female mice were placed on an atherogenic high-cholesterol diet containing 21% (w/w) milk fat, 1.25% (w/w) cholesterol, and 0.5% (w/w) cholic acid (TD 02028, Harlan Laboratories), for 16 weeks.
Mice euthanized by anesthetic overdose and hearts were perfused with phosphate-buffered saline (PBS) and 4% paraformaldehyde, and hearts and the entire aorta were collected. For en face analysis, entire aortas from the heart extending 5 to 10 mm beyond the bifurcation of the iliac arteries were removed and dissected free of adjoining tissues, opened, and stained with Oil Red O. Lesion extent was evaluated by morphometry of scanned images using ImageJ software.
Western blot.
Cell lysates or tissue pieces were prepared by adding protease and phosphatase inhibitor cocktail in RIPA buffer and centrifuging for 10 minutes at 12000 rpm to removed debris. From the lysates, protein concentrations were determined by the Lowry protein assay (500-0113, 500-0114, 500-0115; Bio-Rad). Equal amounts of protein were loaded into each lane of a 4-12% Tris gel (BioRad) and subjected to electrophoresis. After blotting, nitrocellulose-membranes (BioRad) were blocked for 1h (milkpowder 5% in TBS/tween 0.1-0.2%) and incubated overnight at 4°C with primary antibodies (pPI3K, Abcam, ab12135; PI3K, Upstate, 05-212; pAkt, Biosource, 44-622Z; Akt, Cell Signaling, 9272S; pIκBα, Abcam, ab12135; pP65, Cell Signaling, 3036S; P65, Cell Signaling, 4764S; pSTAT3, Cell Signaling, 9131; STAT3, Cell Signaling, 9139; E-Selectin, Santa Cruz, sc-137054; ICAM-1, R&D Systems, AF796; G10 anti-Reelin, made in house; Reelin, Millipore, MAB5366; GAPDH, Sigma-Aldrich, G8795), all diluted at 0,2 μg/ml. Binding of secondary HRP-antibodies were visualized by ECL or ECL plus chemiluminescent (Amersham). After densitometric analyses with ImageJ, optical density values were expressed as arbitrary units and normalized for protein loading to the total form of a protein (for the phosphorylated proteins) or to a housekeeping protein as indicated on the figures, as described in the figure legends.
RT-qPCR.
Total RNA was extracted according to the TRIzol protocol (TRIzol, Life Technologies). RNA concentrations were measured spectrophotometrically (Nanopdrop 2000c; Thermo Scientific). For RNA, first strand cDNA was synthesized using SuperScript III enzyme according to the manufacturer’s instructions (11752; Life Technologies). Quantitative PCR reaction was then performed with TaqMan probes (E-selectin, IDT, Hs.PT.58.1165629; ICAM-1, IDT, Hs.PT.58.4746364; VCAM-1, IDT, Hs.PT.58.20405152, B2M, IDT, 58v.18759587). For each sample, multiplexed reaction was performed, for the target gene (with FAM reporter) and the housekeepers B2M or RPS9 (with VIC reporter). Quantitative PCR reaction was performed with TaqManTM probes using the TaqManTM Universal Master Mix II (4440046; Life Technologies) according to the manufacturer’s instructions. Relative quantity (RQ) values were calculated using the ViiA7 software V1.2.2 (Applied Biosystems).
Statistics.
For cell culture, each condition was tested at least in duplicate (unless specified differently), and all experiments were repeated at least 3 times at different passages. For animals, the “n” values are specified in each legend. The software GraphPad Prism was used to run all the statistical analysis. Values from multiple experiments are expressed as mean±SEM. Normality was tested using the Kolmogorov-Smirnov test. Statistical significance was determined for multiple comparisons using one-way analysis of variance (ANOVA) followed by Turkey multiple comparison (for normal distribution) or Kruskal-Wallis followed by Dunn multiple comparison (for non-normal distribution) test. Student’s t-test (for normal distribution) or Mann Whitney (for non-normal distribution) were used for comparisons of two groups. Equal variance was tested. p<0.05 was considered significant.
RESULTS
Reelin signaling in HAEC promotes monocyte adhesion
In neurons, Reelin binds to Apoer2 to induce phosphorylation of the intracellular adapter protein Disabled-1 (Dab1), which then drives activation of PI3K/Akt, followed by IKK and IκBα and NF-κB 22,23. We hypothesized that a similar signaling cascade exists in HAEC, but through Dab2 instead of Dab1 due to the predominance of this variant in endothelial cells.
Stimulation of HAEC with Reelin resulted in first increased phosphorylation of PI3K, Akt and IκBα, followed in time by NF-κB (Figure 1A). In addition, Reelin promoted the mRNA expression of adhesion molecules E-selectin, ICAM-1 and VCAM-1 which was inhibited by the depletion of Dab2 using an shRNA (Figure 1B). These results confirmed in HAEC that Reelin triggers a phosphorylation cascade similar to that observed in neurons that involves Dab2, PI3K, Akt and NF-κB to promote the expression of adhesion markers.
Figure 1. Reelin signaling in HAEC promotes monocyte adhesion.

(A) In a time-course experiment, HAEC were starved and stimulated with Reelin (20nM) for the indicated time to look at phosphorylation cascade (PI3K, Akt, IκBα, NFκB) by western blot. Representative blots are shown (n=6, ANOVA / Kruskal-Wallis). (B) In expression experiment, HAEC were transduced using shRNA negative control (NC) or targeting Dab2 mRNA (Dab2). Then, cells were starved and stimulated with Reelin (20nM) for 24 hours to look at mRNA expression of DAB2 and adhesion markers (E-selectin, ICAM-1, VCAM-1) by RT-qPCR (n=3, ANOVA). (C) Human plasma was depleted by immunoprecipitation using protein G beads with mouse IgG (control antibody) or CR-50 (anti-Reelin antibody), to obtain “normal” (=N) and “Reelin-depleted” (=D) plasma respectively, and a representative blot is shown. In adhesion functional assay, HAEC were starved and stimulated with PBS as control or human plasma (“normal”=N or “Reelin-depleted”=D) for 1 or 24 h. After washing, the cells were incubated with human monocytes (U937) for 45 min, washed and fixed. Adherent monocytes appearing as small white round cells were counted, and representative pictures are shown (n=6, Kruskal-Wallis). *p<0.05 and **p<0.01.
Next, we investigated the role of Reelin in monocyte adhesion to HAEC in an in vitro system. Human plasma was immunoprecipitated with protein G beads bound to either mouse IgG or CR-50, a mouse anti-Reelin antibody, to obtain “normal” and “Reelin-depleted” plasma, respectively (Figure 1C). HAEC were then incubated with human plasma followed by exposure to human monocytes. Treatment with human “normal” plasma induced monocyte adhesion on endothelial cells at 1 and 24 hours, while the “Reelin-depleted” plasma induced significantly less monocyte adhesion and was similar in effect to untreated HAEC.
Together, these experiments demonstrate in HAEC that Reelin-Apoer2 activates the PI3K / Akt / NF-κB signaling pathway, promotes the expression of adhesion molecules and consequently induces adhesion of monocytes to endothelial cells.
Reelin promotes HASMC proliferation via Stat3
In contrast to canonical (NF-κB) signaling recruited in HAEC, we have also observed a strong phosphorylation of Stat3 induced by Reelin in HASMC. After treatment of HASMC with Reelin, Stat3 phosphorylation peaked at 2 hours, while total Stat3 expression was not affected (Figure 2A). Stat3 is known to regulate cell proliferation in many cell types including SMC. We therefore investigated the role of Reelin/Stat3 on HASMC proliferation using a crystal violet staining assay (Figure 2B). As depicted in Figure 2B, Reelin-induced HASMC proliferation was inhibited by both CR-50 and the Stat3 inhibitor (BP-1-102). These data suggest that reelin, through Stat3 activation, is mitogenic in HASMCs.
Figure 2. Reelin promotes HASMC proliferation via Stat3.

(A) Phosphorylated and total protein expression of Stat3 was evaluated by western blot in HASMC (n≥6, ANOVA). (B) Proliferation was assessed by cell count on HASMC fixed and stained with crystal violet. Cells were starved overnight and incubated as indicated with Reelin (20nM), CR50 (100nM) and Stat3 inhibitor (BP-1-102; 5μM) for 72h. Scale bar = 200μm and representative pictures are shown (n≥6, ANOVA / Kruskal-Wallis). *p<0.05 and **p<0.01.
Anti-Reelin Treatment decreases endothelial adhesion of monocytes
We have previously demonstrated that genetic depletion of Reelin on an Ldlr−/− background impedes monocyte rolling on the vessel wall 4. To explore in vivo the potential of pharmaceutical Reelin depletion on monocyte rolling, we tested anti-Reelin ASO treatment in intravital microscopy. A Cx3cr1-GFP mouse model with genetically GFP-labelled monocytes was used. Seven days after a single ASO intraperitoneal injection, the number of rolling monocytes attached to the endothelial surface was greatly reduced in the setting of reelin depletion, and rolling velocity was increased compared to control (Figure 3A). This resulted in decreased monocyte interaction with the vascular wall as shown by decreased rolling index and cumulative intensity (illustrated by representative cumulative pictures; Figure 3A). We did not observe any significant differences in the monocyte adhesion or velocity between male and female mice. White blood cell number and the vessel area measured by intravital microscopy was similar in both ASO groups.
Figure 3. Anti-Reelin ASO inhibits adhesion of monocytes to endothelial cells.

(A) 4-week-old Cx3cr1-GFP male and female mice were injected intraperitoneally with control (n=10) or anti-Reelin ASO (n=10) at 25mg/kg. Intravital microscopy was performed 7 days after injections to record numbers and speed of rolling monocytes on the marginal mesenteric vessels. 6 different vessels / mouse were recorded for 10 sec and analyzed; cumulative pictures represent individual images (100 frames/second) integrated over a 10-second period and stacked together; rolling index = monocyte number / velocity; t-test / Mann Whitney. (B,C) After intravital microscopy, immunoblotting was performed on plasma and aorta protein extracts. *p<0.05 and **p<0.01 (t-test/Mann Whitney).
We confirmed that Reelin was effectively depleted in the plasma of Anti-Reelin ASO treated mice (Figure 3B) and that this correlated with decreased expression of rolling (E-selectin) and adhesion (ICAM1) markers in the aorta (Figure 3C). This is consistent with our previously published findings showing that Reelin increases E-selectin and ICAM-1 expression in endothelial cells 4,17. These data support Reelin as a therapeutic target for the inhibition of monocyte adhesion and recruitment, with potential impact for perivascular inflammation.
Anti-Reelin Treatment decreases plaque progression in atherosclerosis
Interaction of Reelin with Apoer2 24–29, and Apoer2 itself play a central role in atherosclerosis development, inducing vascular cell adhesion molecule expression and leukocyte–endothelial cell adhesion 1–3. Therefore, we hypothesized that pharmacological Reelin blockade would dampen atherosclerosis progression. To test this hypothesis, Cx3cr1-GFP; Ldlr−/− mice were treated with ASO targeting Reelin mRNA or CR-50 antibody targeting Reelin protein, both effectively decreasing circulating Reelin (Figure 4A,D) and initiated with the high-cholesterol feeding.
Figure 4. Anti-Reelin interventions attenuate atherosclerotic lesion development in Ldlr−/− mice.

8-week-old Ldlr−/− male and female mice were fed with high-cholesterol diet for 16 weeks. Littermates were randomly distributed in four groups and treated with 25mg/kg control ASO (n=11) versus anti-Reelin ASO (n=12) (A-C), or with 100 μg control IgG (n=11) versus anti-Reelin antibody (CR-50; n=11) (D-F). (A,D) 16 weeks after starting the high-cholesterol feeding, plasma Reelin was measured by western blot (Mann Whitney). (B,E) Representative en face photomicrographs and average percent of aortic lesion area stained with Oil Red O are presented (t-test). (C,F) Effect of 16 weeks of high cholesterol diet on the average body weight, fasting plasma cholesterol and triglyceride concentrations are presented. *p<0.05 and **p<0.01 (t-test / Mann Whitney).
After high-cholesterol feeding for 16 weeks beginning at 8 weeks of age, aortas were excised and stained with Oil Red O to visualize atherosclerotic plaques. En face analyses revealed a decrease in atherosclerotic lesion area in aortas from mice treated with Anti-Reelin ASO or antibody compared to control mice treated with control ASO or IgG respectively (Figure 4B,E). Notably, control versus Reelin-depleted mice (either with ASO or CR-50) had similar body weights, and plasma cholesterol and triglyceride concentrations (Figure 4C,F), consistent with our previous results in the systemic and liver-specific Reelin deficient mouse models 4. Taken together, our results show that Reelin depletion by both ASO and CR-50 greatly diminished atherosclerotic lesion formation, independent of plasma lipoprotein levels.
DISCUSSION
In this study, we demonstrated that reduction in circulating Reelin either by ASO or neutralizing antibody CR-50 markedly attenuated atherosclerotic plaque progression in Ldlr−/− mice fed with high cholesterol diet. In vivo, ASO treatment effectively decreased leukocyte-endothelial adhesion as shown by intravital microscopy, suggesting that by decreasing vascular adhesion, anti-Reelin treatment inhibits the recruitment of monocytes and consequently prevents plaque progression. Using human plasma in vitro to study Reelin mechanism, we demonstrated that untreated plasma induced monocyte adhesion to HAEC, while Reelin-depleted plasma had no effect on adhesion. In addition, signaling analysis in HAEC revealed that Reelin activated a Dab2, PI3K, Akt and NF-κB cascade to promote the expression of adhesion markers (E-selectin, ICAM-1, and VCAM-1). Finally, in HASMC, Reelin induced Stat3 phosphorylation to promote cell proliferation, which is a hallmark of plaque progression.
These results are in accordance with and expand our previous studies on Reelin contribution in the development of chronic inflammatory diseases 4,17. We have previously shown in the atherosclerosis-prone Ldlr−/− mouse model 4, and in a mouse experimental autoimmune encephalomyelitis model for multiple sclerosis 17, that Reelin deletion protected against monocyte recruitment and infiltration. Specially, we studied atherosclerosis-prone Ldlr−/− mice in which we deleted Reelin either ubiquitously or only in the liver, thus preventing the production of circulating Reelin 4. In both types of Reelin-deficient mice, atherosclerosis progression was markedly attenuated, and macrophage content and endothelial cell staining for VCAM-1 and ICAM-1 were reduced at the sites of atherosclerotic lesions. In cultured human endothelial cells, Reelin enhanced monocyte adhesion and increased ICAM1, VCAM1, and E-selectin expression by increasing NF-κB activity in an Apoer2-dependent manner. These findings demonstrate that genetical depletion of circulating Reelin protects from atherosclerosis by decreasing vascular inflammation. Here, we show similar protection with two pharmacological anti-Reelin treatments, the ASO and the antibody CR-50 administrated by intraperitoneal injection. While both result in a drastic diminution of Reelin concentration in plasma, they have different modes of action. ASO abolishes Reelin synthesis, which is abundantly produced by stellate cells in the liver 30–33, by entering into cells and degrading Reelin mRNA. On the other hand, CR-50 binds to circulating Reelin protein and thus, clears it most likely through classical immunocomplex degradation.
The atherosclerosis plaque characterization was performed in our previous study 4. Briefly, we investigated the presence of macrophages in atherosclerotic plaques by immunostaining. Morphometric quantification of the lesions revealed a reduction of Mac-3–positive macrophages in Reelin KO mice compared to Ldlr−/− control mice 4. To determine the impact of Reelin genetical deletion on macrophage accumulation in the lesions, we measured VCAM-1 and ICAM-1 abundance in plaques by immunostaining. Both were reduced in endothelium overlying plaques in Reelin KO mice. These results suggest that Reelin promotes macrophage foam cell accumulation in atherosclerotic lesions, and that this is likely driven by increased adhesion molecule expression.
In addition, here we detail the phosphorylation cascade in HAEC, by showing the activation of Dab2, PI3K and Akt, in addition to NF-κB, as previously demonstrated 4. This cascade highlighted in HAEC is in accordance with previous studies in neurons and confirms phosphorylation of similar mediators 34–36. Of note, unlike in neurons, in HAEC, we have not observed Erk phosphorylation under Reelin stimulation. Another difference concerns the Dab protein adaptor for Apoer2. In the nervous system, Reelin binds to Apoer2 and to very low-density lipoprotein receptor (Vldlr), leading to phosphorylation of the intracellular adapter protein Dab1 by Src-family tyrosine kinases 27–29,37, which then activates PI3K/Akt 28,38, thereby controlling various cellular functions, including neuronal positioning, synaptic plasticity, and memory formation 39. In endothelial cells, instead of Dab1 we focused on Dab2, which is believed to be more relevant as an Apoer2 adaptor protein in endothelial cells, whereas Dab1 is predominant in neurons 40.
Finally, we report a previously unknown effect of Reelin on vascular SMC (VSMC) proliferation. Apoer2 deficiency in SMCs has been associated with cell cycle arrest and phenotypic shift from a proliferative to a secretory phenotype 6. It has been shown that Apoer2 deficiency inhibits the formation of the PP2A-C – CDC20 complex, which prevents activation of the APC/C-CDC20 complex, which is required for progression through the cell cycle. Here, we confirmed that Apoer2-mediated signaling induced VSMC proliferation, however through a different mechanism involving Reelin-induced phosphorylation of Stat3. Consequently, Reelin depletion was also associated with decrease in cell proliferation. This mechanism is especially relevant for atherosclerosis since VSMC phenotypic switch (from contractile to proliferative synthetic cells) and aberrant proliferation with intimal invasion promotes plaque formation 41,42. However, the detailed pathway activated by Reelin in VSMC remains unclear. Indeed, the Reelin receptor Apoer2 is abundantly expressed in the vascular wall cells, including platelets, monocytes/macrophages, endothelial cells, and SMC 43. It has been described in multiple myeloma that Reelin activates Stat3 via integrin α5β1 (specially via the subunit β1) 44,45, integrin which is also expressed on VSMC membrane 46. Interestingly, direct Reelin interaction with integrins could also explain how Reelin promotes short term (1 hour) monocyte/endothelial cell adhesion without de novo synthesis of additional adhesion molecules (Figure 1C). The novel, Stat3-mediated signaling observed in VSMC would require a dedicated study to investigate the recruitment of integrins.
In conclusion, we have exploited a function of Reelin in the regulation of leukocyte-endothelial adhesion to devise a novel strategy for combating inflammation and plaque progression in atherosclerosis. Despite the broad clinical use of statins to lower LDL cholesterol, atherosclerosis is still an important cause of cardiovascular morbidity and mortality. This underlines the need for adjunctive non-lipid-lowering therapies against the progression of atherosclerotic disease. Reelin depletion decreases atherosclerosis progression independently of lipid levels, and therefore supports targeting the inflammatory side of atherosclerosis, which could be an adjunct to the widely accepted lipid-lowering therapies. Beyond cardiovascular diseases, anti-Reelin strategies would be expected to be similarly effective for the treatment of other chronic inflammatory syndromes that depend upon excessive leukocyte extravasation such as multiple sclerosis, arthritis or Crohn’s disease.
Supplementary Material
Figure 5. Therapeutic Reelin depletion decreases endothelial adhesion of leukocytes and protects against atherosclerosis.

This mechanistic model incorporates the findings described in this article using pharmacological depletion of Reelin, our previous article using genetical depletion of Reelin 4, and the literature to date as discussed. In vascular endothelial cells, Reelin promotes the expression of vascular adhesion proteins, thereby increasing monocyte adhesion / extravasation, inflammation and consequently plaque progression. In vascular smooth muscle cells, Reelin promotes cell proliferation via Stat3 phosphorylation. Therapeutic Reelin depletion prevents this inflammatory cascade and thus atherosclerosis. We surmise that these mechanisms are conserved in humans.
HIGHLIGHTS SECTION.
Pharmacological Reelin depletion (either by ASO or neutralizing antibody) decreases plaque progression in atherosclerosis mouse model.
Reelin via Apoer2 activates Dab2, PI3K, Akt and NF-κB cascade to promote the expression of adhesion markers (E-selectin, ICAM-1, and VCAM-1) in vascular endothelial cells and therefore, Reelin promotes monocyte/endothelial cell adhesion.
Identification of a non-canonical Reelin pathway via Stat3 activation to promote vascular smooth muscle cell proliferation.
ACKNOWLEDGEMENTS
We thank Anna Middleton, Tamara Terrones, and Alexander Brennan for their technical assistance.
SOURCES OF FUNDING
L.C. was supported by postdoctoral fellowship grants from DFG (CA 1303/1-1). P.W.S. and C.M. were supported by grants from NIH (R01-HL131597 and R01-HL109604 respectively). J.H. was supported by grants from the NHLBI (R37 HL063762), NIA (RF AG053391), the NINDS and NIA (R01 NS093382), BrightFocus A2016396S, the Bluefield Project to Cure FTD and a Harrington Scholar Innovator Award (2019).
DISCLOSURES
L.C., M.Z.K. and J.H. are shareholders of Reelin Therapeutics, Inc.
L.C. and J.H. are co-inventors of a patent related to anti-Reelin strategies (application number 15/763,047 and publication number 20180273637).
Abbreviations
- EC
endothelial cells
- Apoer2 or Lrp8
Apolipoprotein E receptor-2
- LDLR
low-density lipoprotein receptor
- VCAM-1
vascular cell adhesion molecule-1
- ICAM-1
intercellular adhesion molecule-1
- NF-κB
nuclear factor κB
- ASO
antisense oligonucleotide
- HAEC
human aortic endothelial cells
- FBS
fetal bovine serum
- PBS
phosphate-buffered saline
- Dab1
Disabled-1
REFERENCES
- 1.Sacre SM, Stannard AK, Owen JS. Apolipoprotein E (apoE) isoforms differentially induce nitric oxide production in endothelial cells. FEBS Lett. 2003;540:181–187. [DOI] [PubMed] [Google Scholar]
- 2.Stannard AK, Riddell DR, Sacre SM, Tagalakis AD, Langer C, von Eckardstein A, Cullen P, Athanasopoulos T, Dickson G, Owen JS. Cell-derived apolipoprotein E (ApoE) particles inhibit vascular cell adhesion molecule-1 (VCAM-1) expression in human endothelial cells. J Biol Chem. 2001;276:46011–46016. doi: 10.1074/jbc.M104812200 [DOI] [PubMed] [Google Scholar]
- 3.Ramesh S, Morrell CN, Tarango C, Thomas GD, Yuhanna IS, Girardi G, Herz J, Urbanus RT, de Groot PG, Thorpe PE, Salmon JE, Shaul PW, Mineo C. Antiphospholipid antibodies promote leukocyte-endothelial cell adhesion and thrombosis in mice by antagonizing eNOS via β2GPI and apoER2. J Clin Invest. 2011;121:120–131. doi: 10.1172/JCI39828 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ding Y, Huang L, Xian X, Yuhanna IS, Wasser CR, Frotscher M, Mineo C, Shaul PW, Herz J. Loss of Reelin protects against atherosclerosis by reducing leukocyte-endothelial cell adhesion and lesion macrophage accumulation. Science Signaling. 2016;9:ra29–ra29. doi: 10.1126/scisignal.aad5578 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Waltmann MD, Basford JE, Konaniah ES, Weintraub NL, Hui DY. Apolipoprotein E receptor-2 deficiency enhances macrophage susceptibility to lipid accumulation and cell death to augment atherosclerotic plaque progression and necrosis. Biochim Biophys Acta. 2014;1842:1395–1405. doi: 10.1016/j.bbadis.2014.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Komaravolu RK, Waltmann MD, Konaniah E, Jaeschke A, Hui DY. ApoER2 (Apolipoprotein E Receptor-2) Deficiency Accelerates Smooth Muscle Cell Senescence via Cytokinesis Impairment and Promotes Fibrotic Neointima After Vascular Injury. Arterioscler Thromb Vasc Biol. 2019;39:2132–2144. doi: 10.1161/ATVBAHA.119.313194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wongsurawat T, Woo CC, Giannakakis A, Lin XY, Cheow ESH, Lee CN, Richards M, Sze SK, Nookaew I, Kuznetsov VA, Sorokin V. Transcriptome alterations of vascular smooth muscle cells in aortic wall of myocardial infarction patients. Data Brief. 2018;17:1112–1135. doi: 10.1016/j.dib.2018.01.108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wang Q, Rao S, Shen G-Q, Li L, Moliterno DJ, Newby LK, Rogers WJ, Cannata R, Zirzow E, Elston RC, Topol EJ. Premature myocardial infarction novel susceptibility locus on chromosome 1P34-36 identified by genomewide linkage analysis. Am J Hum Genet. 2004;74:262–271. doi: 10.1086/381560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Asif M, Bhat S, Nizamuddin S, Mustak MS. TG haplotype in the LRP8 is associated with myocardial infarction in south Indian population. Gene. 2018;642:225–229. doi: 10.1016/j.gene.2017.10.037 [DOI] [PubMed] [Google Scholar]
- 10.Ramesh S, Morrell CN, Tarango C, Thomas GD, Yuhanna IS, Girardi G, Herz J, Urbanus RT, de Groot PG, Thorpe PE, Salmon JE, Shaul PW, Mineo C. Antiphospholipid antibodies promote leukocyte-endothelial cell adhesion and thrombosis in mice by antagonizing eNOS via β2GPI and apoER2. J Clin Invest. 2011;121:120–131. doi: 10.1172/JCI39828 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Romay-Penabad Z, Aguilar-Valenzuela R, Urbanus RT, Derksen RHWM, Pennings MTT, Papalardo E, Shilagard T, Vargas G, Hwang Y, de Groot PG, Pierangeli SS. Apolipoprotein E receptor 2 is involved in the thrombotic complications in a murine model of the antiphospholipid syndrome. Blood. 2011;117:1408–1414. doi: 10.1182/blood-2010-07-299099 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Weeber EJ, Beffert U, Jones C, Christian JM, Forster E, Sweatt JD, Herz J. Reelin and ApoE receptors cooperate to enhance hippocampal synaptic plasticity and learning. J Biol Chem. 2002;277:39944–39952. doi: 10.1074/jbc.M205147200 [DOI] [PubMed] [Google Scholar]
- 13.Chen Y, Beffert U, Ertunc M, Tang T-S, Kavalali ET, Bezprozvanny I, Herz J. Reelin modulates NMDA receptor activity in cortical neurons. J Neurosci. 2005;25:8209–8216. doi: 10.1523/JNEUROSCI.1951-05.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Beffert U, Weeber EJ, Durudas A, Qiu S, Masiulis I, Sweatt JD, Li W-P, Adelmann G, Frotscher M, Hammer RE, Herz J. Modulation of synaptic plasticity and memory by Reelin involves differential splicing of the lipoprotein receptor Apoer2. Neuron. 2005;47:567–579. doi: 10.1016/j.neuron.2005.07.007 [DOI] [PubMed] [Google Scholar]
- 15.Herz J, Chen Y. Reelin, lipoprotein receptors and synaptic plasticity. Nat Rev Neurosci. 2006;7:850–859. doi: 10.1038/nrn2009 [DOI] [PubMed] [Google Scholar]
- 16.Förster E, Bock HH, Herz J, Chai X, Frotscher M, Zhao S. Emerging topics in Reelin function. Eur J Neurosci. 2010;31:1511–1518. doi: 10.1111/j.1460-9568.2010.07222.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Calvier L, Demuth G, Manouchehri N, Wong C, Sacharidou A, Mineo C, Shaul PW, Monson NL, Kounnas MZ, Stüve O, Herz J. Reelin depletion protects against autoimmune encephalomyelitis by decreasing vascular adhesion of leukocytes. Sci Transl Med. 2020;12:eaay7675. doi: 10.1126/scitranslmed.aay7675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Calvier L, Chouvarine P, Legchenko E, Hoffmann N, Geldner J, Borchert P, Jonigk D, Mozes MM, Hansmann G. PPARγ Links BMP2 and TGFβ1 Pathways in Vascular Smooth Muscle Cells, Regulating Cell Proliferation and Glucose Metabolism. Cell Metab. 2017;25:1118–1134.e7. doi: 10.1016/j.cmet.2017.03.011 [DOI] [PubMed] [Google Scholar]
- 19.Calvier L, Boucher P, Herz J, Hansmann G. LRP1 Deficiency in Vascular SMC Leads to Pulmonary Arterial Hypertension That Is Reversed by PPARγ Activation. Circ Res. 2019;124:1778–1785. doi: 10.1161/CIRCRESAHA.119.315088 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Calvier L, Chouvarine P, Legchenko E, Kokeny G, Mozes MM, Hansmann G. Chronic TGF-β1 Signaling in Pulmonary Arterial Hypertension Induces Sustained Canonical Smad3 Pathways in Vascular Smooth Muscle Cells. Am J Respir Cell Mol Biol. 2019;61:121–123. doi: 10.1165/rcmb.2018-0275LE [DOI] [PubMed] [Google Scholar]
- 21.Ishibashi S, Brown MS, Goldstein JL, Gerard RD, Hammer RE, Herz J. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J Clin Invest. 1993;92:883–893. doi: 10.1172/JCI116663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Jossin Y, Gui L, Goffinet AM. Processing of Reelin by embryonic neurons is important for function in tissue but not in dissociated cultured neurons. J Neurosci. 2007;27:4243–4252. doi: 10.1523/JNEUROSCI.0023-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Yasui N, Nogi T, Kitao T, Nakano Y, Hattori M, Takagi J. Structure of a receptor-binding fragment of reelin and mutational analysis reveal a recognition mechanism similar to endocytic receptors. Proc Natl Acad Sci USA. 2007;104:9988–9993. doi: 10.1073/pnas.0700438104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Trommsdorff M, Gotthardt M, Hiesberger T, Shelton J, Stockinger W, Nimpf J, Hammer RE, Richardson JA, Herz J. Reeler/Disabled-like disruption of neuronal migration in knockout mice lacking the VLDL receptor and ApoE receptor 2. Cell. 1999;97:689–701. [DOI] [PubMed] [Google Scholar]
- 25.Hiesberger T, Trommsdorff M, Howell BW, Goffinet A, Mumby MC, Cooper JA, Herz J. Direct binding of Reelin to VLDL receptor and ApoE receptor 2 induces tyrosine phosphorylation of disabled-1 and modulates tau phosphorylation. Neuron. 1999;24:481–489. [DOI] [PubMed] [Google Scholar]
- 26.D’Arcangelo G, Homayouni R, Keshvara L, Rice DS, Sheldon M, Curran T. Reelin is a ligand for lipoprotein receptors. Neuron. 1999;24:471–479. [DOI] [PubMed] [Google Scholar]
- 27.Arnaud L, Ballif BA, Förster E, Cooper JA. Fyn tyrosine kinase is a critical regulator of disabled-1 during brain development. Curr Biol. 2003;13:9–17. [DOI] [PubMed] [Google Scholar]
- 28.Bock HH, Herz J. Reelin activates SRC family tyrosine kinases in neurons. Curr Biol. 2003;13:18–26. [DOI] [PubMed] [Google Scholar]
- 29.Kuo G, Arnaud L, Kronstad-O’Brien P, Cooper JA. Absence of Fyn and Src causes a reeler-like phenotype. J Neurosci. 2005;25:8578–8586. doi: 10.1523/JNEUROSCI.1656-05.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Carotti S, Perrone G, Amato M, Vespasiani Gentilucci U, Righi D, Francesconi M, Pellegrini C, Zalfa F, Zingariello M, Picardi A, Onetti Muda A, Morini S. Reelin expression in human liver of patients with chronic hepatitis C infection. Eur J Histochem. 2017;61:2745. doi: 10.4081/ejh.2017.2745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Samama B, Boehm N. Reelin immunoreactivity in lymphatics and liver during development and adult life. Anat Rec A Discov Mol Cell Evol Biol. 2005;285:595–599. doi: 10.1002/ar.a.20202 [DOI] [PubMed] [Google Scholar]
- 32.Botella-Lopez A, de Madaria E, Jover R, Bataller R, Sancho-Bru P, Candela A, Compañ A, Pérez-Mateo M, Martinez S, Sáez-Valero J. Reelin is overexpressed in the liver and plasma of bile duct ligated rats and its levels and glycosylation are altered in plasma of humans with cirrhosis. Int J Biochem Cell Biol. 2008;40:766–775. [DOI] [PubMed] [Google Scholar]
- 33.Smalheiser NR, Costa E, Guidotti A, Impagnatiello F, Auta J, Lacor P, Kriho V, Pappas GD. Expression of reelin in adult mammalian blood, liver, pituitary pars intermedia, and adrenal chromaffin cells. Proc Natl Acad Sci USA. 2000;97:1281–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lane-Donovan C, Herz J. The ApoE receptors Vldlr and Apoer2 in central nervous system function and disease. J Lipid Res. 2017;58:1036–1043. doi: 10.1194/jlr.R075507 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bock HH, May P. Canonical and Non-canonical Reelin Signaling. Front Cell Neurosci. 2016;10:166. doi: 10.3389/fncel.2016.00166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lee GH, D’Arcangelo G. New Insights into Reelin-Mediated Signaling Pathways. Front Cell Neurosci. 2016;10:122. doi: 10.3389/fncel.2016.00122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Howell BW, Hawkes R, Soriano P, Cooper JA. Neuronal position in the developing brain is regulated by mouse disabled-1. Nature. 1997;389:733–737. doi: 10.1038/39607 [DOI] [PubMed] [Google Scholar]
- 38.Beffert U, Morfini G, Bock HH, Reyna H, Brady ST, Herz J. Reelin-mediated signaling locally regulates protein kinase B/Akt and glycogen synthase kinase 3beta. J Biol Chem. 2002;277:49958–49964. doi: 10.1074/jbc.M209205200 [DOI] [PubMed] [Google Scholar]
- 39.Durakoglugil MS, Chen Y, White CL, Kavalali ET, Herz J. Reelin signaling antagonizes beta-amyloid at the synapse. Proc Natl Acad Sci USA. 2009;106:15938–15943. doi: 10.1073/pnas.0908176106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sacharidou A, Chambliss KL, Ulrich V, Salmon JE, Shen Y-M, Herz J, Hui DY, Terada LS, Shaul PW, Mineo C. Antiphospholipid antibodies induce thrombosis by PP2A activation via apoER2-Dab2-SHC1 complex formation in endothelium. Blood. 2018;131:2097–2110. doi: 10.1182/blood-2017-11-814681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lacolley P, Regnault V, Segers P, Laurent S. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease. Physiol Rev. 2017;97:1555–1617. doi: 10.1152/physrev.00003.2017 [DOI] [PubMed] [Google Scholar]
- 42.Basatemur GL, Jørgensen HF, Clarke MCH, Bennett MR, Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat Rev Cardiol. 2019;16:727–744. doi: 10.1038/s41569-019-0227-9 [DOI] [PubMed] [Google Scholar]
- 43.Mineo C Lipoprotein Receptor Signaling in Atherosclerosis. Cardiovasc Res. Published online December 13, 2019. doi: 10.1093/cvr/cvz338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lin L, Zhang X, Cao L, An Q, Hao J, Zhang Y, Jin R, Chang Y, Huang X, Lu J, Ge Q. Reelin promotes adhesion of multiple myeloma cells to bone marrow stromal cells via integrin β1 signaling. J Cancer. 2017;8:2212–2222. doi: 10.7150/jca.18808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lin L, Yan F, Zhao D, Lv M, Liang X, Dai H, Qin X, Zhang Y, Hao J, Sun X, Yin Y, Huang X, Zhang J, Lu J, Ge Q. Reelin promotes the adhesion and drug resistance of multiple myeloma cells via integrin β1 signaling and STAT3. Oncotarget. 2016;7:9844–9858. doi: 10.18632/oncotarget.7151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Davenpeck KL, Marcinkiewicz C, Wang D, Niculescu R, Shi Y, Martin JL, Zalewski A. Regional differences in integrin expression: role of alpha(5)beta(1) in regulating smooth muscle cell functions. Circ Res. 2001;88:352–358. doi: 10.1161/01.res.88.3.352 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
