Human cathelicidin LL-37 is an antimicrobial peptide that has a broad spectrum of antimicrobial activities but also acts on host cells to exert immunomodulatory functions. It has been suggested that the increase of LL-37 in atherosclerotic aortas and the dysregulated autophagy of endothelial cells are involved in the pathogenesis of atherosclerosis. In this study, to elucidate the role of LL-37 in atherosclerosis, we investigated the effect of LL-37 on autophagy in endothelial cells using HUVECs. First, LL-37 upregulated LC3-II (an autophagosomal membrane marker) and enhanced the formation of LC3-positive puncta in the cells, suggesting that LL-37 induces autophagy in endothelial cells. Second, LL-37 was associated with p62, which recognizes ubiquitinated proteins and transfers them to autophagosomes, suggesting that LL-37 is ubiquitinated and recognized by p62. Third, the degradation of LL-37 was delayed, and LL-37 induced cell death in atg7 knockdown cells, which was accompanied by the formation of protein aggregates in the cells. Taken together, these observations suggest that LL-37 induces autophagy in endothelial cells but enhances cell death in autophagy-dysfunctional conditions, in which the intracellular degradation of LL-37 is disturbed. Thus, LL-37 may exert an adverse action on autophagy-dysfunctional endothelial cells to induce cell death in the pathogenesis of atherosclerosis.

Cathelicidins are a family of antimicrobial peptides that are expressed in several mammalian species. They are characterized by highly conserved cathelin-like prosequences and variable C-terminal sequences that correspond to the mature antibacterial peptides (1). LL-37 is the sole mature peptide of human cathelicidin; it comprises 37 aa and contains the first two leucine residues and an amphipathic α-helical conformation, which is cleaved from the proform of a human cationic antibacterial protein of 18 kDa (2). In addition to its membrane-disrupting antimicrobial properties, LL-37 binds to LPS (3), an outer membrane component of Gram-negative bacteria, and exerts an LPS-neutralizing action, as well as membrane-disrupting antimicrobial properties (1). Moreover, it has been shown that LL-37 exerts multidirectional actions on host cells (4). LL-37 is directly chemotactic for neutrophils, monocytes, and T cells, whereas LL-37 promotes the migration of dendritic cells through the ability to degranulate mast cells. LL-37 activates epithelial cells to induce cytokine and chemokine production. Furthermore, LL-37 acts on endothelial cells to promote cell proliferation (angiogenesis) and induce inflammatory responses (such as the upregulation of ICAM-1) (5).

Atherosclerosis is a chronic inflammatory and immune disease of vascular walls involving multiple cell types (monocytes, macrophages, T cells, endothelial cells, and vascular smooth muscle cells) (6). Endothelial dysfunction, such as the decrease of NO production, is a critical early event of the pathogenesis of atherosclerosis and contributes to the initiation and progression of plaque formation. In the advanced stage, damaged or apoptotic endothelial cells become procoagulant in the plaque lesions, thus promoting the formation of thrombi. Notably, recent studies demonstrated the involvement of cathelicidins (human LL-37 and its mouse ortholog CRAMP [cathelin-related antimicrobial peptide]) in the pathogenesis of atherosclerosis; it has been shown that LL-37 is deposited in the atherosclerotic lesions of human aortas (5), and that the deletion of CRAMP results in the reduction of plaque lesions and macrophage accumulation in apolipoprotein E (ApoE)−/− atherosclerotic mice (7).

Autophagy is a lysosome-dependent cellular degradation process that is triggered by cellular stresses. In endothelial cells, the lack of autophagy results in the intracellular accumulation of the low-density lipoprotein and oxidized low-density lipoprotein (8), the impaired secretion of the von Willebrand factor (9), and decreased production of NO (10). The lack of autophagy also induces the inflammatory activation in endothelial cells (such as the upregulation of ICAM-1) (11). Importantly, the autophagy of endothelial cells is modulated in the pathogenesis of atherosclerosis; in its early stage, autophagy is activated in endothelial cells; however, autophagy becomes dysfunctional in the advanced stage of the disease, leading to apoptotic cell death and plaque instability (12).

The observations described earlier led to the hypothesis that the upregulation of LL-37 in atherosclerotic aortas and the dysregulated autophagy of endothelial cells are involved in the pathogenesis of atherosclerosis. However, it remains to be elucidated how LL-37 modulates the autophagy of endothelial cells in the pathogenesis of this disease. Therefore, in this study, we evaluated the effect of LL-37 on the autophagy of endothelial cells using HUVECs and human coronary artery endothelial cells (HCAECs). Our results indicated that LL-37 enhances autophagy in endothelial cells but induces cell death in autophagy-dysfunctional endothelial cells.

LL-37 (L1LGDFFRKSKEKIGKEFKRIVQRIKDFLRMLVPRTES37) was synthesized on a peptide synthesizer (model PSSM-8; Shimadzu, Kyoto, Japan) using the solid-phase method and F-moc chemistry and was then purified as described previously (13). The lysosomal protease inhibitors E-64d and pepstatin A were purchased from Sigma-Aldrich (St. Louis, MO). Human recombinant IL-1β was purchased from PeproTech (Cranbury, NJ). The anti–LL-37 antiserum was raised in rabbits using LL-37 covalently coupled to keyhole limpet hemocyanin, as described previously (14). Abs to LC3 (rabbit PM036 and mouse M152-3), Atg7 (rabbit PM039), p62 (rabbit PM045), and normal rabbit IgG (PM035) were from MBL (Nagoya, Japan); anti-ubiquitin Abs (rabbit E4I2J and mouse FK2) were from Cell Signaling Technology (Danvers, MA); the anti-CRAMP Ab (PA-CRPL-100) was from Innovagen (Lund, Sweden); and the anti-LL-37 Ab (D-5) was from Santa Cruz (Dallas, TX).

HUVECs and HCAECs were purchased from Lonza (Basel, Switzerland) and maintained in endothelial cell growth medium (EGM)-2 (containing 2% FBS and the endothelial cell growth supplement EGM-2 Bullet kit) and EGM-2MV (containing 5% FBS and the EGM-2MV Bullet kit), respectively, in the presence of antibiotics at 37°C in 5% CO2. The cells of passages 5–12 were used in all experiments. Tissue culture–treated dishes and plates were obtained from Iwaki (Tokyo, Japan).

HUVECs were seeded in six-well plates and cultured overnight. Cells were transfected with 5 nM of an atg7 small interfering RNA (siRNA; s20650) or a nontargeting control siRNA (Silencer Select; Ambion, Thermo Fisher Scientific, Waltham, MA) using the Lipofectamine RNAiMAX transfection reagent (Thermo Fisher Scientific) for 6 h. Thereafter, cells were washed with EGM-2 and further incubated for up to 72 h. Subsequently, cell lysates were prepared and the expression of Atg7, p62, and GAPDH was detected by Western blotting, as described later. The expression of Atg7 was substantially suppressed in the atg7-siRNA–transfected cells compared with nontargeting control siRNA-transfected cells at 48 and 72 h after transfection (Fig. 4B). Thus, in this study, 48 h after transfection, the siRNA-transfected HUVECs were incubated with LL-37 for the indicated periods and subjected to the following experiments.

HUVECs or HCAECs were incubated with LL-37 (2, 5, and 10 µg/ml) for 24 h in the absence or presence of lysosomal protease inhibitors (E-64d and pepstatin A, 10 µg/ml each) to detect LC3. Alternatively, to evaluate the time-dependent degradation of LL-37, we incubated HUVECs (atg7 knockdown [KD] and control cells) with LL-37 (5 µg/ml) for 15 min, extensively washed them with HBSS (to remove unbound LL-37), and incubated them in EGM-2 for 1–12 h. Cells were lysed in radioimmunoprecipitation buffer (50 mM Tris–HCl [pH 8.0], 150 mM NaCl, 0.5% sodium deoxycholate, 1% Nonidet P-40, and 0.1% SDS; Fujifilm Wako Pure Chemical, Osaka, Japan) containing the Complete protease inhibitor mixture (Sigma-Aldrich). Cell lysates (10 µg of protein/lane) were mixed with Laemmli sample buffer containing 2-ME, subjected to SDS-PAGE, and transferred to polyvinylidene fluoride membranes (Immobilon-P; Millipore, Billerica, MA) using a semidry transfer system (Trans-Blot; Bio-Rad, Hercules, CA). Membranes were blocked with BlockAce (DS Pharma Biomedical, Osaka, Japan) or skim milk (Nacalai Tesque, Kyoto, Japan), then probed with rabbit anti-LC3 (PM036), anti-Atg7, anti-p62, anti-ubiquitin (E4I2J), or anti–LL-37 (D-5) Abs, followed by incubation with HRP-conjugated goat anti-rabbit IgG (Chemicon International, Temecula, CA) or goat anti-mouse IgG/IgM (Jackson ImmunoResearch Laboratories, West Grove, PA). Signals were developed using the SuperSignal West Pico/Dura Chemiluminescent Substrate (Pierce Biotechnology, Rockford, IL) and detected using a FUSION FX luminescent image analyzer (Vilber Lourmat, Collégien, France). GAPDH was detected with an HRP-conjugated anti-GAPDH Ab (5A12; Fujifilm Wako Pure Chemical) as a loading control.

HUVECs or HCAECs were seeded in Nunc Lab Tek II CC2 chamber slides (Thermo Fisher Scientific) and cultured overnight. Cells were incubated with LL-37 (5 µg/ml) for 24 h in the presence of E-64d and pepstatin A. Alternatively, atg7 KD and nontargeting control cells were incubated with LL-37 (5 µg/ml) for 24 h. Cells were then fixed with 2% paraformaldehyde (PFA), permeabilized with 0.2% saponin, blocked with 5% skim milk, and incubated with the mouse anti-LC3 Ab (M152-3) or rabbit LL-37 antiserum. After washing with HBSS, cells were further incubated with Alexa Fluor 594–labeled goat anti-mouse IgG or Alexa Fluor 488–labeled goat anti-rabbit IgG, followed by mounting with Vectashield Hardset with DAPI (Vector Laboratories, San Diego, CA). Fluorescent images were captured with a BZ-X710 microscope (Keyence, Itasca, IL) or an FV1000-D confocal microscope (Olympus, Tokyo, Japan). The rate of LC3-positive puncta was calculated as a percentage by dividing by the number of total cells (DAPI staining). The colocalization analysis of LL-37 and LC3 was performed on a pixel-by-pixel basis using the FLUOVIEW software (Olympus).

HUVECs (atg7 KD and nontargeting control cells) or HCAECs (in the presence/absence of E-64d and pepstatin A) were incubated with LL-37 (2, 5, and 10 µg/ml) for 24 h. Cells were detached with 0.05% trypsin-EDTA, collected in test tubes, and suspended in HBSS. Viable trypan blue–excluded cells were counted with a hemocytometer. Alternatively, cells were incubated with Annexin V–FITC and propidium iodide (PI) solutions (Annexin V-FITC Apoptosis Detection Kit; BioVision, Mountain View, CA) for 5 min, then analyzed by flow cytometry (FACSCalibur, Becton Dickinson, Franklin Lakes, NJ). Annexin V+/PI+ cells were classified as dead cells with apoptosis, whereas Annexin V/PI+ cells were classified as dead cells with necrosis.

HUVECs were incubated with LL-37 (5 µg/ml) in the absence or presence of E-64d and pepstatin A for 4 h, and cell lysates were prepared using immunoprecipitation (IP) lysis buffer (25 mM Tris–HCl [pH 7.4], 150 mM NaCl, 1% Nonidet P-40, 1 mM EDTA, and 5% glycerol; Pierce, Thermo Fisher Scientific) containing the Halt protease and phosphatase inhibitor mixture (Thermo Fisher Scientific). IP was performed using the Dynabeads protein A IP kit (Thermo Fisher Scientific), according to the manufacturer’s instructions. Briefly, protein A–conjugated magnetic beads were incubated with anti-p62 or anti-ubiquitin (FK2) Abs or normal IgG, washed, and then incubated with the cell lysates overnight. Proteins were eluted with Laemmli sample buffer containing 2-ME and subjected to Western blotting using anti-p62, anti–LL-37 (D-5), or anti-ubiquitin (E4I2J) Abs, as described earlier.

HUVECs (atg7 KD and nontargeting control cells) were incubated with LL-37 (5 µg/ml) for 4 h, detached with 0.05% trypsin-EDTA, and collected in test tubes. Cells were fixed with 2% PFA, permeabilized with 0.5% Triton X-100, washed with HBSS, incubated with the PROTEOSTAT aggresome detection dye (ENZO Life Sciences, Farmingdale, NY) (15), and analyzed by flow cytometry (FACSCalibur). To analyze the colocalization of LL-37 with PROTEOSTAT dye–positive aggregates, we incubated atg7 KD cells in chamber slides with LL-37 (5 µg/ml), fixed them with 2% PFA, permeabilized with 0.5% Triton X-100, washed with HBSS, and incubated with the anti–LL-37 rabbit antiserum; after washing with HBSS, cells were further incubated with Alexa Fluor 488–labeled goat anti-rabbit IgG and the PROTEOSTAT dye, followed by mounting in Vectashield Hardset with DAPI. Fluorescent images were captured using a BZ-X710 microscope. The PROTEOSTAT-positive and LL-37–positive cells were counted among dead cells with condensed nuclei or live cells with larger round nuclei (DAPI staining), and the percentages of LL-37+ and LL-37 cells among the PROTEOSTAT+ dead or live cells were calculated.

All animal experimental procedures were approved by the Ethics Committee for the Use of Laboratory Animals of Juntendo University (2021291, June 2021). Ten-week-old male C57BL/6 mice and spontaneously hyperlipidemic male C57BL/6.KOR-ApoEshl mice were obtained from Japan SLC (Shizuoka, Japan). Mice were transcardially perfused with cold heparin/PBS followed by 4% PFA under anesthesia. The aortic roots were isolated, postfixed with 4% PFA, cryoprotected with 30% sucrose/PBS, and then embedded in optimal cutting temperature compound (Sakura Finetechnical, Tokyo, Japan). Frozen sections (thickness, 8 µm) were prepared using a CM3050S cryostat (Leica Biosystems, Wetzlar, Germany) and incubated with 0.5% Triton X-100/PBS at 80°C for 20 min (for Ag retrieval), followed by incubation with an anti-CRAMP Ab (1:1000) and Alexa Fluor 594–labeled goat anti-rabbit IgG. Sections were further incubated with the TUNEL reaction buffer containing fluorescein-dUTP (in situ cell death detection kit, Fluorescein; Roche, Darmstadt, Germany) at 37°C for 1 h, followed by mounting in mounting media. Fluorescent images were captured using a BZ-X710 microscope.

Data are presented as the mean ± SD. Statistical significance was determined by two-way ANOVA, followed by Dunnett’s multiple comparisons test (GraphPad Prism 8, GraphPad Software, San Diego, CA). Differences were considered statistically significant at p < 0.05.

A previous study indicated that LL-37 upregulates autophagy-related gene expression in macrophages and induces autophagosome formation, to promote the killing of intracellular bacteria (16). To examine whether LL-37 induces autophagy in endothelial cells, we incubated HUVECs with LL-37 (2, 5, and 10 µg/ml) for 24 h in the absence or presence of lysosomal protease inhibitors (E-64d and pepstatin A); subsequently, LC3-I (cytosolic LC3) and LC3-II (autophagosomal membrane–bound LC3) were detected by Western blotting. LC3-I (upper band) was faintly detected in LL-37–treated cells in the absence of inhibitors, and LL-37 weakly but substantially increased the level of LC3-II (lower band) shifted from LC3-I in the absence of protease inhibitors (Fig. 1A, left panel). However, LL-37 apparently increased the level of LC3-II (expressed not only as the LC3-II/GAPDH ratio but also as the LC3-II/LC3-I ratio) in a dose-dependent manner in the presence of protease inhibitors (Fig. 1A, right panel, 1B, 1C). Furthermore, an immunocytochemical analysis was performed using an anti-LC3 Ab. LL-37 (5 µg/ml) obviously enhanced the formation of LC3-positive puncta and increased the number of cells containing these puncta in the presence of protease inhibitors (Fig. 1D, 1E). In addition, using HCAECs, we confirmed that LL-37 upregulates LC3-II and increases the number of cells containing LC3-positive puncta (Fig. 2A–C). These observations suggest that LL-37 induces autophagy in endothelial cells.

FIGURE 1.

LL-37 induces autophagy in HUVECs. HUVECs were incubated without (0 µg/ml) or with LL-37 (2, 5, and 10 µg/ml) in the absence or presence of E-64d and pepstatin A (10 µg/ml each) for 24 h. LC3 and GAPDH (a loading control) were detected by Western blotting, and the signals were quantified using an image analyzer. Membrane-bound LC3-II was detected as a lower band compared with cytosolic LC3-I (A). LC3-II/GAPDH (B) and LC3-II/LC3-I (C) were expressed as a ratio to the cells incubated without LL-37 (0 µg/ml) in the presence of E-64d and pepstatin A. Images are representative of four independent experiments. Data are the mean ± SD of four independent experiments. Values were compared between the cells without (0 µg/ml) and with LL-37 (2, 5, and 10 µg/ml) (A–C). (D) HUVECs were seeded in chamber slides and incubated without or with LL-37 (5 µg/ml) in the presence of E-64d and pepstatin A for 24 h. Cells were then fixed/permeabilized and incubated with the anti-LC3 Ab, followed by Alexa Fluor 594–labeled anti-rabbit IgG incubation (red). Images are representative of four independent experiments. The area containing the LC3-positive puncta is enclosed by a white dotted line and shown as an expanded image. (E) Total cells (evaluated by DAPI staining) and cells with LC3 puncta were counted in fluorescence images, and the percentage of cells containing LC3 puncta was calculated among the cells incubated without (0 µg/ml) and with LL-37 (5 µg/ml). Data are the mean ± SD of four independent experiments. Values were compared between the cells without and with LL-37 (E). *p < 0.05, **p < 0.01. Scale bars, 20 µm.

FIGURE 1.

LL-37 induces autophagy in HUVECs. HUVECs were incubated without (0 µg/ml) or with LL-37 (2, 5, and 10 µg/ml) in the absence or presence of E-64d and pepstatin A (10 µg/ml each) for 24 h. LC3 and GAPDH (a loading control) were detected by Western blotting, and the signals were quantified using an image analyzer. Membrane-bound LC3-II was detected as a lower band compared with cytosolic LC3-I (A). LC3-II/GAPDH (B) and LC3-II/LC3-I (C) were expressed as a ratio to the cells incubated without LL-37 (0 µg/ml) in the presence of E-64d and pepstatin A. Images are representative of four independent experiments. Data are the mean ± SD of four independent experiments. Values were compared between the cells without (0 µg/ml) and with LL-37 (2, 5, and 10 µg/ml) (A–C). (D) HUVECs were seeded in chamber slides and incubated without or with LL-37 (5 µg/ml) in the presence of E-64d and pepstatin A for 24 h. Cells were then fixed/permeabilized and incubated with the anti-LC3 Ab, followed by Alexa Fluor 594–labeled anti-rabbit IgG incubation (red). Images are representative of four independent experiments. The area containing the LC3-positive puncta is enclosed by a white dotted line and shown as an expanded image. (E) Total cells (evaluated by DAPI staining) and cells with LC3 puncta were counted in fluorescence images, and the percentage of cells containing LC3 puncta was calculated among the cells incubated without (0 µg/ml) and with LL-37 (5 µg/ml). Data are the mean ± SD of four independent experiments. Values were compared between the cells without and with LL-37 (E). *p < 0.05, **p < 0.01. Scale bars, 20 µm.

Close modal
FIGURE 2.

Effect of LL-37 on autophagy and cell death of HCAECs. HCAECs were incubated without or with LL-37 (2, 5, or 10 µg/ml) in the presence of E-64d and pepstatin A for 24 h. LC3 and GAPDH were detected by Western blotting, and the signals were quantified using an image analyzer. LC3-II/GAPDH (A) and LC3-II/LC3-I (B) were expressed as a ratio to the cells incubated without LL-37. Data are the mean ± SD of three independent experiments. Values were compared between the cells without (0) and with LL-37 (2, 5, 10) (A and B). HCAECs were seeded in chamber slides and incubated with LL-37 (5 µg/ml) in the presence of E-64d and pepstatin A for 24 h. Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum and anti-LC3 Ab, followed by Alexa Fluor 488–labeled anti-rabbit IgG and Alexa Fluor 594–labeled anti-mouse IgG incubation. Total cells and cells with LC3-puncta were counted in fluorescence images, and the percentage of cells with LC3-puncta was calculated (C). Data are the mean ± SD of four independent experiments. Values were compared between the cells without (0) and with LL-37 (5). Images are representative of four independent experiments (C and D). HCAECs were incubated without or with LL-37 (2, 5, or 10 µg/ml) in the absence or presence of E-64d and pepstatin A for 24 h. Cells were collected in test tubes, and the number of viable cells was counted using trypan blue (E). Alternatively, cell death was analyzed by flow cytometry, and the percentages of PI+ dead cells were calculated (F). Data are the mean ± SD of six independent experiments. Values were compared between the incubation without (0) and with LL-37 (5, 10), or between the cells in the absence and presence of E-64d and pepstatin A (E and F). *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 2.

Effect of LL-37 on autophagy and cell death of HCAECs. HCAECs were incubated without or with LL-37 (2, 5, or 10 µg/ml) in the presence of E-64d and pepstatin A for 24 h. LC3 and GAPDH were detected by Western blotting, and the signals were quantified using an image analyzer. LC3-II/GAPDH (A) and LC3-II/LC3-I (B) were expressed as a ratio to the cells incubated without LL-37. Data are the mean ± SD of three independent experiments. Values were compared between the cells without (0) and with LL-37 (2, 5, 10) (A and B). HCAECs were seeded in chamber slides and incubated with LL-37 (5 µg/ml) in the presence of E-64d and pepstatin A for 24 h. Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum and anti-LC3 Ab, followed by Alexa Fluor 488–labeled anti-rabbit IgG and Alexa Fluor 594–labeled anti-mouse IgG incubation. Total cells and cells with LC3-puncta were counted in fluorescence images, and the percentage of cells with LC3-puncta was calculated (C). Data are the mean ± SD of four independent experiments. Values were compared between the cells without (0) and with LL-37 (5). Images are representative of four independent experiments (C and D). HCAECs were incubated without or with LL-37 (2, 5, or 10 µg/ml) in the absence or presence of E-64d and pepstatin A for 24 h. Cells were collected in test tubes, and the number of viable cells was counted using trypan blue (E). Alternatively, cell death was analyzed by flow cytometry, and the percentages of PI+ dead cells were calculated (F). Data are the mean ± SD of six independent experiments. Values were compared between the incubation without (0) and with LL-37 (5, 10), or between the cells in the absence and presence of E-64d and pepstatin A (E and F). *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

Next, we investigated the association between LL-37 and the autophagosomal membrane and autophagosome-related molecules in endothelial cells. Double immunostaining of LL-37–treated HUVECs using an anti–LL-37 antiserum and an anti-LC3 Ab revealed the colocalization of LL-37 and LC3 puncta around the nuclei in the presence of E-64d and pepstatin A (Fig. 3A). We also investigated the localization of LL-37 and LC3 using confocal microscopy, which indicated that LL-37 partially but substantially colocalized with LC3 around nuclei (Fig. 3B). In fact, a pixel-based colocalization analysis indicated that, in the perinuclear area of the cells, pixels that were positive for LL-37 and LC3 were more frequent than those positive for LC3 or LL-37 alone (Fig. 3B, 3C), although these differences were not statistically significant. In addition, we confirmed the colocalization of LL-37 and LC3 puncta around nuclei in the presence of E-64d and pepstatin A in HCAECs (Fig. 2D). Because LC3 is localized at the autophagosomal membrane, the interaction between LL-37 and p62, which is an autophagosome membrane–associated molecule, was further examined.

p62 recognizes ubiquitinated substrates (such as misfolded or aggregated proteins) via its ubiquitin-associated domain and shuttles the ubiquitinated substrates to autophagosomes for degradation (17). Thus, we first evaluated LL-37, p62, and ubiquitinated proteins without IP in the lysates of cells treated with or without LL-37 in the absence or presence of E-64d and pepstatin A (Fig. 3D). LL-37 was detected exclusively in the LL-37–treated cell lysates, whereas p62 and ubiquitinated proteins were detected in both LL-37–treated and untreated cells. Interestingly, smear bands of LL-37 were detected in long exposure blots of the LL-37–treated cell lysates without inhibitors and were further increased by the presence of inhibitors. Second, the lysates of LL-37–treated cells were immunoprecipitated with the anti-p62 Ab, followed by the detection of the LL-37, p62, and ubiquitinated proteins (Fig. 3E). LL-37 and ubiquitinated proteins were recovered with p62. Importantly, LL-37 and p62 were apparently increased by the presence of inhibitors. Third, the lysates of LL-37–treated cells were immunoprecipitated with the anti-ubiquitin Ab, followed by the detection of LL-37, p62, and ubiquitinated proteins (Fig. 3F). LL-37, p62, and ubiquitinated proteins were increased by the presence of inhibitors; notably, smear bands of LL-37 were detected in the cell lysates without inhibitors and were further increased by the presence of inhibitors.

FIGURE 3.

LL-37 colocalizes with LC3 and associates with p62 in HUVECs. HUVECs were seeded in chamber slides and incubated with LL-37 (5 µg/ml) in the presence of E-64d and pepstatin A for 24 h. Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum (green) and an anti-LC3 Ab (red), followed by Alexa Fluor 488–labeled anti-rabbit IgG and Alexa Fluor 594–labeled anti-mouse IgG incubation. Nuclei were stained with DAPI (blue). Images are representative of three independent experiments (A). The localization of LL-37 and LC3 was analyzed using confocal microscopy, and pixels that were positive for LC3 (LC3+), LL-37 (LL-37+), and LC3+LL-37 (LC3+/LL-37+) were calculated as percentages of the total pixels in the perinuclear area of the cells. Image is representative of nine independent experiments (B). Data are the mean ± SD of nine independent experiments (C). HUVECs were incubated without or with LL-37 (5 µg/ml) for 4 h in the absence (−) or presence (+) of E-64d and pepstatin A (Inhibitors), and cell lysates were prepared. LL-37, ubiquitin, p62, and GAPDH were detected by Western blotting. The signal of LL-37 was developed using shorter (short exposure) and longer (long exposure) exposure times. Images are representative of three independent experiments (D). The LL-37–treated cell lysates were immunoprecipitated with anti-p62 (α-p62), anti-ubiquitin (α-Ub), or normal rabbit IgG. The precipitates were subjected to SDS-PAGE, and ubiquitinated proteins, LL-37, and p62 were detected by Western blotting. The heavy chain of IgG (IgG, 50 kDa) was detected in the images of immunoblotting (LL-37, ubiquitin, and p62) after IP with α-p62, α-Ub, and normal IgG. Images are representative of three independent experiments (E and F). Scale bars, 20 µm.

FIGURE 3.

LL-37 colocalizes with LC3 and associates with p62 in HUVECs. HUVECs were seeded in chamber slides and incubated with LL-37 (5 µg/ml) in the presence of E-64d and pepstatin A for 24 h. Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum (green) and an anti-LC3 Ab (red), followed by Alexa Fluor 488–labeled anti-rabbit IgG and Alexa Fluor 594–labeled anti-mouse IgG incubation. Nuclei were stained with DAPI (blue). Images are representative of three independent experiments (A). The localization of LL-37 and LC3 was analyzed using confocal microscopy, and pixels that were positive for LC3 (LC3+), LL-37 (LL-37+), and LC3+LL-37 (LC3+/LL-37+) were calculated as percentages of the total pixels in the perinuclear area of the cells. Image is representative of nine independent experiments (B). Data are the mean ± SD of nine independent experiments (C). HUVECs were incubated without or with LL-37 (5 µg/ml) for 4 h in the absence (−) or presence (+) of E-64d and pepstatin A (Inhibitors), and cell lysates were prepared. LL-37, ubiquitin, p62, and GAPDH were detected by Western blotting. The signal of LL-37 was developed using shorter (short exposure) and longer (long exposure) exposure times. Images are representative of three independent experiments (D). The LL-37–treated cell lysates were immunoprecipitated with anti-p62 (α-p62), anti-ubiquitin (α-Ub), or normal rabbit IgG. The precipitates were subjected to SDS-PAGE, and ubiquitinated proteins, LL-37, and p62 were detected by Western blotting. The heavy chain of IgG (IgG, 50 kDa) was detected in the images of immunoblotting (LL-37, ubiquitin, and p62) after IP with α-p62, α-Ub, and normal IgG. Images are representative of three independent experiments (E and F). Scale bars, 20 µm.

Close modal

Based on these observations, it is reasonable to speculate that: (1) LL-37 is ubiquitinated, recognized by p62 to form an LL-37/p62 complex, and colocalizes with LC3 at the autophagosomal membrane; and (2) the ubiquitination is enhanced in the presence of lysosomal protease inhibitors.

The degradation of LL-37 by HUVECs was examined by incubating the cells with LL-37 (5 µg/ml) for 24 h in the absence or presence of E-64d and pepstatin A, followed by immunocytochemistry using the anti–LL-37 antiserum. Interestingly, a small amount of LL-37 was detected in the cells without E-64d and pepstatin A; in contrast, LL-37 accumulated in the cells as puncta in the perinuclear area in the presence of inhibitors at 24 h after LL-37 treatment (Fig. 4A). Subsequently, the degradation of LL-37 was examined using atg7 KD cells. Atg7 is essential for autophagosome formation, and autophagy is dysfunctional in atg7 KD endothelial cells (8, 9). As shown in (Fig. 4B, the expression of Atg7 was successfully suppressed in atg7-siRNA–transfected cells compared with nontargeting control siRNA-transfected cells; in contrast, the expression of p62 was increased in atg7 KD cells, in which autophagy is dysregulated and the degradation of p62 is suppressed (18). Interestingly, LL-37 accumulated as puncta in the perinuclear area in atg7 KD cells compared with nontargeting control cells (Fig. 4C).

FIGURE 4.

The degradation of LL-37 in HUVECs is delayed by lysosomal protease inhibitors and by KD of atg7. HUVECs were seeded in chamber slides and incubated without or with LL-37 (5 µg/ml) for 24 h in the absence (−) or presence (+) of E-64d and pepstatin A (A). Alternatively, HUVECs were incubated without or with LL-37 (5 µg/ml) for 24 h after the transfection of a nontargeting control siRNA (siCont) and an atg7 siRNA (siAtg7) (C). Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum, followed by Alexa Fluor 488–labeled anti-rabbit IgG (green). Images are representative of three independent experiments (A and C). To evaluate the KD of Atg7 in the siRNA-transfected cells, we incubated HUVECs for 72 h after siRNA transfection, and the expression of Atg7, p62, and GAPDH was detected by Western blotting at 48 and 72 h after transfection using an image analyzer. Images are representative of five independent experiments (B). (D and E) The siCont and atg7 KD cells were incubated without (0 µg/ml) or with LL-37 (5 µg/ml) for 15 min, extensively washed with HBSS, and incubated for 1–12 h without LL-37. LL-37 and GAPDH were detected by Western blotting, and the signals were quantified using an image analyzer. The amount of LL-37 was corrected using that of GAPDH and expressed as a ratio of the cells (0 h) incubated with LL-37 for 15 min, but not incubated after washing. Images are representative of four independent experiments (D). Data are the mean ± SD of four independent experiments. Values were compared between the cells without (0 h) and with incubation (1, 3, 6, and 12 h), or between siCont and siAtg7 cells (E). #p < 0.05, ##p < 0.01, ###p < 0.001 between the cells without (0 h) and with incubation (1, 3, 6, and 12 h); *p < 0.05 between siCont and siAtg7 cells. Scale bars, 20 µm.

FIGURE 4.

The degradation of LL-37 in HUVECs is delayed by lysosomal protease inhibitors and by KD of atg7. HUVECs were seeded in chamber slides and incubated without or with LL-37 (5 µg/ml) for 24 h in the absence (−) or presence (+) of E-64d and pepstatin A (A). Alternatively, HUVECs were incubated without or with LL-37 (5 µg/ml) for 24 h after the transfection of a nontargeting control siRNA (siCont) and an atg7 siRNA (siAtg7) (C). Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum, followed by Alexa Fluor 488–labeled anti-rabbit IgG (green). Images are representative of three independent experiments (A and C). To evaluate the KD of Atg7 in the siRNA-transfected cells, we incubated HUVECs for 72 h after siRNA transfection, and the expression of Atg7, p62, and GAPDH was detected by Western blotting at 48 and 72 h after transfection using an image analyzer. Images are representative of five independent experiments (B). (D and E) The siCont and atg7 KD cells were incubated without (0 µg/ml) or with LL-37 (5 µg/ml) for 15 min, extensively washed with HBSS, and incubated for 1–12 h without LL-37. LL-37 and GAPDH were detected by Western blotting, and the signals were quantified using an image analyzer. The amount of LL-37 was corrected using that of GAPDH and expressed as a ratio of the cells (0 h) incubated with LL-37 for 15 min, but not incubated after washing. Images are representative of four independent experiments (D). Data are the mean ± SD of four independent experiments. Values were compared between the cells without (0 h) and with incubation (1, 3, 6, and 12 h), or between siCont and siAtg7 cells (E). #p < 0.05, ##p < 0.01, ###p < 0.001 between the cells without (0 h) and with incubation (1, 3, 6, and 12 h); *p < 0.05 between siCont and siAtg7 cells. Scale bars, 20 µm.

Close modal

To further examine the uptake and degradation of LL-37 by endothelial cells, we incubated HUVECs (atg7 KD and nontargeting control cells) with LL-37 (5 µg/ml) for 15 min, extensively washed them (to remove unbound LL-37), and incubated them for 1–12 h, followed by the detection of LL-37 using Western blotting. The level of LL-37 was time dependently decreased in the nontargeting control cells (Fig. 4D, 4E). In contrast, the degradation of LL-37 was delayed, and the LL-37 level remained constant in the atg7 KD cells compared with the control cells; in fact, the half-life of LL-37 was 3–6 h in the control cells, whereas it was >12 h in the atg7 KD cells (Fig. 4E). In addition, we confirmed that virtually the same amounts of LL-37 were detected early (within 15 min) after LL-37 treatment both in atg7 KD and control cells; thus, the uptake of LL-37 by endothelial cells was not different between atg7 KD and control cells (Fig. 4D). Taken together, these observations suggest that LL-37 is rapidly internalized by endothelial cells and is subsequently degraded by lysosomal proteases; however, in autophagy-dysfunctional endothelial cells, LL-37 is internalized by endothelial cells, but its degradation is disturbed, resulting in the accumulation of LL-37 in these cells.

It has been reported that the intracellular accumulation of proteins leads to cell death in autophagy-dysfunctional conditions (19, 20). To determine whether LL-37 induces cell death in autophagy-dysfunctional endothelial cells, we evaluated the effect of LL-37 on the viability of atg7 KD and nontargeting control HUVECs via trypan blue staining. Viability was slightly decreased in the atg7 KD cells compared with nontargeting control cells in the absence of LL-37, although this difference was not statistically significant (Fig. 5A). LL-37 (2, 5, and 10 µg/ml) did not affect the viability of control cells. In contrast, LL-37 significantly reduced the number of viable cells in the atg7 KD cells at 5 and 10 µg/ml. Furthermore, Annexin V/PI staining revealed that LL-37 (2, 5, and 10 µg/ml) did not essentially increase the number of apoptotic (Annexin V+/PI+), necrotic (Annexin V/PI+), and dead (Annexin V+/PI+ and Annexin V/PI+) cells in the control group. In contrast, in the atg7 KD cells, LL-37 significantly increased the number of apoptotic and necrotic cells (at 10 µg/ml; (Fig. 5B, 5C), as well as the dead cell number (at 5 and 10 µg/ml; (Fig. 5D). In addition, we confirmed that LL-37 decreased the cell viability and increased the cell death of HCAECs in the presence of protease inhibitors, as assessed by trypan blue staining and PI staining, respectively (Fig. 2E, 2F). These observations suggest that LL-37 enhances cell death (apoptosis and necrosis) in autophagy-dysfunctional endothelial cells.

FIGURE 5.

LL-37 decreases the viability and enhances the death of atg7 KD cells. Nontargeting control siRNA (siCont) and atg7 KD (siAtg7) cells were incubated without (0 µg/ml) or with LL-37 (2, 5, and 10 µg/ml) for 24 h. Cells were collected in test tubes, and the number of viable cells was counted using trypan blue. Data are the mean ± SD of six independent experiments. Values were compared between cells without (0 µg/ml) and with LL-37 (5, 10 µg/ml) (A). Alternatively, cell death was analyzed using Annexin V/PI staining by flow cytometry, and the percentages of apoptotic cells (Annexin V+/PI+) (B), necrotic cells (Annexin V/PI+) (C), or dead cells (Annexin V/PI+ and Annexin V+/PI+) (D) were calculated. Data are the mean ± SD of five independent experiments. Values were compared between cells without (0 µg/ml) and with LL-37 (2, 5, 10 µg/ml), or between siCont and siAtg7 cells (B–D). *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 5.

LL-37 decreases the viability and enhances the death of atg7 KD cells. Nontargeting control siRNA (siCont) and atg7 KD (siAtg7) cells were incubated without (0 µg/ml) or with LL-37 (2, 5, and 10 µg/ml) for 24 h. Cells were collected in test tubes, and the number of viable cells was counted using trypan blue. Data are the mean ± SD of six independent experiments. Values were compared between cells without (0 µg/ml) and with LL-37 (5, 10 µg/ml) (A). Alternatively, cell death was analyzed using Annexin V/PI staining by flow cytometry, and the percentages of apoptotic cells (Annexin V+/PI+) (B), necrotic cells (Annexin V/PI+) (C), or dead cells (Annexin V/PI+ and Annexin V+/PI+) (D) were calculated. Data are the mean ± SD of five independent experiments. Values were compared between cells without (0 µg/ml) and with LL-37 (2, 5, 10 µg/ml), or between siCont and siAtg7 cells (B–D). *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

It has been reported that the formation of intracellular protein aggregates leads to cell death in autophagy-dysfunctional conditions (19, 20). Because LL-37 enhanced cell death in autophagy-dysfunctional endothelial cells (Fig. 5), we hypothesized that LL-37 increases protein aggregate formation in autophagy-dysfunctional HUVECs. To examine whether LL-37 increases aggregate formation in HUVECs (atg7 KD and nontargeting control cells), these cells were incubated with LL-37 (5 µg/ml) for 24 h, and the formation of intracellular aggregates was examined by immunocytochemical staining and flow cytometry using the PROTEOSTAT aggregate detection dye, which specifically intercalates into aggregated proteins (15). An immunocytochemical analysis indicated that aggregates were weakly detected in LL-37–treated nontargeting control cells; in contrast, aggregates were apparently increased in LL-37–treated atg7 KD cells (Fig. 6A). Moreover, a flow cytometrical analysis indicated that the level of aggregates was slightly but significantly increased by LL-37 in the nontargeting control cells. Notably, the aggregate level was further increased by LL-37 in the atg7 KD cells (Fig. 6B, 6C). These observations indicate that LL-37 enhances aggregate formation in autophagy-dysfunctional endothelial cells.

FIGURE 6.

LL-37 increases aggregate formation in HUVECs. Nontargeting control siRNA (siCont) and atg7 KD (siAtg7) cells in chamber slides were incubated without or with LL-37 (5 µg/ml) for 24 h. Cells were fixed/permeabilized and incubated with the PROTEOSTAT dye (red), and the aggregates in cells were detected using fluorescence microscopy. Images are representative of three independent experiments (A). The siCont and siAtg7 cells were treated as described earlier, collected in test tubes, fixed/permeabilized, and incubated with the PROTEOSTAT dye. The aggregates in cells were analyzed by flow cytometry and are shown as fluorescence intensity histograms. The dotted lines indicate the mean fluorescence intensity (MFI) of siCont cells incubated without LL-37 (upper left) (B). The relative amounts of aggregates in the cells were expressed as a ratio of MFI to the nontargeting control cells (siCont) without LL-37 (−) (C). Data are the mean ± SD of four independent experiments. Values were compared between the cells without (−) and with (+) LL-37 (LL-37), or between siCont and siAtg7 cells (B and C). *p < 0.05, ***p < 0.001. Scale bar, 50 µm.

FIGURE 6.

LL-37 increases aggregate formation in HUVECs. Nontargeting control siRNA (siCont) and atg7 KD (siAtg7) cells in chamber slides were incubated without or with LL-37 (5 µg/ml) for 24 h. Cells were fixed/permeabilized and incubated with the PROTEOSTAT dye (red), and the aggregates in cells were detected using fluorescence microscopy. Images are representative of three independent experiments (A). The siCont and siAtg7 cells were treated as described earlier, collected in test tubes, fixed/permeabilized, and incubated with the PROTEOSTAT dye. The aggregates in cells were analyzed by flow cytometry and are shown as fluorescence intensity histograms. The dotted lines indicate the mean fluorescence intensity (MFI) of siCont cells incubated without LL-37 (upper left) (B). The relative amounts of aggregates in the cells were expressed as a ratio of MFI to the nontargeting control cells (siCont) without LL-37 (−) (C). Data are the mean ± SD of four independent experiments. Values were compared between the cells without (−) and with (+) LL-37 (LL-37), or between siCont and siAtg7 cells (B and C). *p < 0.05, ***p < 0.001. Scale bar, 50 µm.

Close modal

The involvement of aggregate formation in the LL-37–induced death of atg7 KD cells was also evaluated. PROTEOSTAT-positive aggregates were localized at the perinuclear area of LL-37–treated atg7 KD cells (Fig. 7A). LL-37 also accumulated as puncta at the perinuclear area in cells and colocalized with PROTEOSTAT-positive aggregates (Fig. 7A). Furthermore, the percentages of aggregate (PROTEOSTAT)-positive cells without and with LL-37 accumulation among live and dead cells were evaluated. The results of this experiment indicated that the percentage of aggregate-positive cells without LL-37 accumulation was higher among dead cells (14%) compared with that detected among live cells (6%), even though this difference was not significant (Fig. 7B). Importantly, the percentage of aggregate-positive cells with LL-37 accumulation was significantly higher among dead cells (59%) compared with live cells (3%) (Fig. 7B). These observations suggest that the LL-37–induced aggregate formation is associated with the death of autophagy-dysfunctional endothelial cells.

FIGURE 7.

Accumulation of LL-37 induces cell death in atg7 KD cells. The atg7 KD cells in chamber slides were incubated with LL-37 (5 µg/ml) for 24 h. Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum, followed by Alexa Fluor 488–labeled anti-rabbit IgG (green) and the PROTEOSTAT dye (red), and finally mounted with mounting media containing DAPI. The arrows in the image of DAPI staining indicate dead cells containing condensed nuclei. Images are representative of three independent experiments (A). Cells that were positive for the PROTEOSTAT dye and LL-37 were counted in the above images, and the percentages of aggregate (PROTEOSTAT)-positive cells without and with LL-37 accumulation were calculated among live and dead cells. Data are the mean ± SD of four independent experiments. Values were compared between aggregate-positive cells without and with LL-37 accumulation among dead cells and between live and dead cells for aggregate-positive cells with LL-37 accumulation (B). ***p < 0.001.

FIGURE 7.

Accumulation of LL-37 induces cell death in atg7 KD cells. The atg7 KD cells in chamber slides were incubated with LL-37 (5 µg/ml) for 24 h. Cells were then fixed/permeabilized and incubated with the anti–LL-37 antiserum, followed by Alexa Fluor 488–labeled anti-rabbit IgG (green) and the PROTEOSTAT dye (red), and finally mounted with mounting media containing DAPI. The arrows in the image of DAPI staining indicate dead cells containing condensed nuclei. Images are representative of three independent experiments (A). Cells that were positive for the PROTEOSTAT dye and LL-37 were counted in the above images, and the percentages of aggregate (PROTEOSTAT)-positive cells without and with LL-37 accumulation were calculated among live and dead cells. Data are the mean ± SD of four independent experiments. Values were compared between aggregate-positive cells without and with LL-37 accumulation among dead cells and between live and dead cells for aggregate-positive cells with LL-37 accumulation (B). ***p < 0.001.

Close modal

The involvement of LL-37, which is an antimicrobial peptide of human cathelicidin, in the pathogenesis of atherosclerosis has been speculated, because LL-37 accumulates in human atherosclerotic aortas (5), and the deletion of CRAMP resulted in the reduction of plaque formation and macrophage accumulation in ApoE−/− atherosclerotic mice (7). Actually, using ApoE-deficient atherosclerotic mice, we confirmed that CRAMP accumulated in TUNEL+ dead endothelial cells at the surface of aortic valves (Supplemental Fig. 2). In turn, the autophagy of endothelial cells is dysregulated in atherosclerosis (12). In this study, to elucidate the role of LL-37 in atherosclerosis, we investigated the effect of LL-37 on autophagy in endothelial cells. First, the level of LC3-II, which is an autophagosomal membrane marker, was evaluated in LL-37–treated HUVECs. LL-37 only weakly upregulated LC3-II in the absence of lysosomal protease inhibitors (E-64d and pepstatin A); in contrast, LL-37 apparently upregulated LC3-II in the presence of inhibitors (Fig. 1A–C). Because LC3-II is degraded by lysosomal proteases via autophagy (21), LC3-II is likely increased in LL-37–treated HUVECs in the presence of lysosomal protease inhibitors. Furthermore, LL-37 enhanced the formation of LC3-containing puncta (representing autophagosomes) in the presence of inhibitors (Fig. 1D, 1E). These observations indicate that LL-37 induces autophagy in endothelial cells.

Previous studies demonstrated that LL-37 is internalized and colocalizes with LC3 puncta in macrophages and plasmacytoid dendritic cells during autophagy (16, 22, 23). Thus, the colocalization of LL-37 with LC3 was examined in HUVECs by immunocytochemistry. The results of this experiment indicated that LL-37 colocalized with LC3 puncta in the perinuclear area in the presence of lysosomal protease inhibitors (Fig. 3A–C), suggesting the association between LL-37 and the autophagosomal membrane. In the autophagic process, ubiquitinated substrate proteins are recognized by p62 (which is an LC3-II–bound autophagosomal membrane-associated molecule) and transferred to autophagosomes. Thus, the association between LL-37 and p62 and the ubiquitination of LL-37 were examined by IP. Interestingly, LL-37 was recovered using not only an anti-p62 Ab but also an anti-ubiquitin Ab (Fig. 3E, 3F). These observations suggest that LL-37 is ubiquitinated, recognized by p62 to form a complex, and possibly transferred to autophagosomes as a substrate.

Furthermore, the degradation of LL-37 was examined. The degradation of LL-37 was delayed in atg7 KD cells (Fig. 4D, 4E) and in the presence of lysosomal protease inhibitors (data not shown) during the incubation with LL-37. These observations suggest that LL-37 is internalized by endothelial cells and degraded by lysosomal proteases via autophagy; in contrast, the degradation of LL-37 is delayed, and LL-37 accumulates in the autophagy-dysfunctional cells.

Several reports have indicated that LL-37 is internalized by various types of cell (including macrophages, mast cells, epithelial cells, and endothelial cells) by direct translocation (across the membrane) or endocytosis, depending on the cell type. Importantly, we previously revealed that LL-37 is internalized by liver sinusoidal endothelial cells via an interaction with cell-surface heparan sulfate proteoglycans by endocytosis, then transferred to lysosomes (24). Thus, in this study, LL-37 was possibly incorporated into endothelial cells by endocytosis and transferred to lysosomes for degradation.

It has been reported that the intracellular accumulation of proteins leads to cell death under autophagy-dysfunctional conditions (19, 20). Therefore, the effect of LL-37 on the death of atg7 KD and nontargeting control cells was examined. Our results indicated that LL-37 decreased the number of viable cells and increased that of dead cells (apoptotic and necrotic cells) in atg7 KD cells (Fig. 5), suggesting that LL-37 enhances cell death in autophagy-dysfunctional endothelial cells, in which LL-37 is intracellularly accumulated. To elucidate further the mechanism underlying the LL-37–induced death of atg7 KD cells, we evaluated intracellular protein accumulation (aggregate formation) using the PROTEOSTAT dye. The results demonstrated that LL-37 apparently increased aggregate formation in atg7 KD cells (Fig. 6). Moreover, LL-37 colocalized with PROTEOSTAT+ aggregates in atg7 KD cells (Fig. 7A), and the number of aggregate-positive cells with LL-37 accumulation was significantly increased among dead cells compared with live cells (Fig. 7B). These observations suggest that LL-37 increases protein aggregates intracellularly, thereby inducing cell death in autophagy-dysfunctional endothelial cells.

It has been reported that ubiquitinated protein aggregates are accumulated in autophagy-dysfunctional cells (17). The results of this study indicated that LL-37 is possibly ubiquitinated (Fig. 3F) and that it colocalized with the PROTEOSTAT dye in atg7 KD cells (Fig. 7A). The PROTEOSTAT dye specifically intercalates into the β-sheet structures that are typically found in misfolded and aggregated proteins (15). Notably, LL-37 forms β-sheets and β-fibrils in the presence of lipids or the lipid membrane (25). Taken together, these observations suggest that the ubiquitinated LL-37 accumulates intracellularly and forms protein aggregates with the PROTEOSTAT dye in autophagy-dysfunctional HUVECs, which is accompanied by cell death.

The human cathelicidin peptide LL-37 acts on endothelial cells to promote cell proliferation (angiogenesis) (26) and maintains endothelial barrier permeability (27). However, in this study, we revealed that LL-37 induced autophagy in endothelial cells but enhanced cell death in autophagy-dysfunctional conditions, in which the intracellular degradation of LL-37 is disturbed, and protein aggregates accumulate. Interestingly, we confirmed that IL-1β, which is an atherosclerosis-associated inflammatory cytokine (28), upregulated LC3-II, and LL-37 further increased the IL-1β–induced expression of LC3-II (Supplemental Fig. 1A–C). Moreover, cell death was increased by IL-1β and further increased by LL-37, which colocalized with LC3 around nuclei (Supplemental Fig. 1D, 1E). Importantly, it has been reported that the autophagy of endothelial cells is dysregulated in atherosclerosis (12), and that LL-37 is accumulated in atherosclerotic lesions (5). Taken together, these findings suggest that not only IL-1β but also LL-37 plays a role in atherosclerosis, and LL-37 exerts an adverse action on autophagy-dysfunctional endothelial cells to induce cell death in the pathogenesis of atherosclerosis, a chronic inflammatory disease of vascular walls.

We thank members of the Laboratory of Proteomics and Biomolecular Science (Drs. Yuka Hiraoka, Mika Kikkawa, and Yoshiki Miura), the Laboratory of Morphology and Image Analysis (Dr. Shinji Nakamura), the Laboratory of Molecular and Biochemical Research, Biomedical Research Core Facilities (Dr. Tomomi Ikeda and Prof. Hiroshi Koide), and the Division of Cell Biology (Drs. Akemi Koyanagi and Tamami Sakanishi), Research Support Center, Juntendo University Graduate School of Medicine for synthesizing LL-37 peptide or technical assistance.

This work was supported in part by a Grant-in-Aid (Grant 19K07545) for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology, Japan, Japan Society for the Promotion of Science and a Grant-in-Aid (Grant S1411007) from the Ministry of Education, Culture, Sports, Science, and Technology, Japan for the Foundation of Strategic Research Projects in Private Universities.

The online version of this article contains supplemental material.

Abbreviations used in this article:

ApoE

apolipoprotein E

CRAMP

cathelin-related antimicrobial peptide

EGM, endothelial cell growth medium; HCAEC

human coronary artery endothelial cell

IP

immunoprecipitation

KD

knockdown

PFA

paraformaldehyde

PI

propidium iodide

1.
Zanetti
M.
2005
.
The role of cathelicidins in the innate host defenses of mammals.
Curr. Issues Mol. Biol.
7
:
179
196
.
2.
Larrick
J. W.
,
M.
Hirata
,
R. F.
Balint
,
J.
Lee
,
J.
Zhong
,
S. C.
Wright
.
1995
.
Human CAP18: a novel antimicrobial lipopolysaccharide-binding protein.
Infect. Immun.
63
:
1291
1297
.
3.
Hirata
M.
,
M.
Yoshida
,
K.
Inada
,
T.
Kirikae
.
1990
.
Investigation of endotoxin binding cationic proteins from granulocytes; agglutination of erythrocytes sensitized with Re-LPS.
Adv. Exp. Med. Biol.
256
:
287
299
.
4.
Niyonsaba
F.
,
I.
Nagaoka
,
H.
Ogawa
.
2006
.
Human defensins and cathelicidins in the skin: beyond direct antimicrobial properties.
Crit. Rev. Immunol.
26
:
545
576
.
5.
Edfeldt
K.
,
B.
Agerberth
,
M. E.
Rottenberg
,
G. H.
Gudmundsson
,
X. B.
Wang
,
K.
Mandal
,
Q.
Xu
,
Z. Q.
Yan
.
2006
.
Involvement of the antimicrobial peptide LL-37 in human atherosclerosis.
Arterioscler. Thromb. Vasc. Biol.
26
:
1551
1557
.
6.
Galkina
E.
,
K.
Ley
.
2009
.
Immune and inflammatory mechanisms of atherosclerosis (*).
Annu. Rev. Immunol.
27
:
165
197
.
7.
Döring
Y.
,
M.
Drechsler
,
S.
Wantha
,
K.
Kemmerich
,
D.
Lievens
,
S.
Vijayan
,
R. L.
Gallo
,
C.
Weber
,
O.
Soehnlein
.
2012
.
Lack of neutrophil-derived CRAMP reduces atherosclerosis in mice.
Circ. Res.
110
:
1052
1056
.
8.
Torisu
K.
,
K. K.
Singh
,
T.
Torisu
,
F.
Lovren
,
J.
Liu
,
Y.
Pan
,
A.
Quan
,
A.
Ramadan
,
M.
Al-Omran
,
N.
Pankova
, et al
2016
.
Intact endothelial autophagy is required to maintain vascular lipid homeostasis.
Aging Cell
15
:
187
191
.
9.
Torisu
T.
,
K.
Torisu
,
I. H.
Lee
,
J.
Liu
,
D.
Malide
,
C. A.
Combs
,
X. S.
Wu
,
I. I.
Rovira
,
M. M.
Fergusson
,
R.
Weigert
, et al
2013
.
Autophagy regulates endothelial cell processing, maturation and secretion of von Willebrand factor.
Nat. Med.
19
:
1281
1287
.
10.
LaRocca
T. J.
,
G. D.
Henson
,
A.
Thorburn
,
A. L.
Sindler
,
G. L.
Pierce
,
D. R.
Seals
.
2012
.
Translational evidence that impaired autophagy contributes to arterial ageing.
J. Physiol.
590
:
3305
3316
.
11.
Vion
A. C.
,
M.
Kheloufi
,
A.
Hammoutene
,
J.
Poisson
,
J.
Lasselin
,
C.
Devue
,
I.
Pic
,
N.
Dupont
,
J.
Busse
,
K.
Stark
, et al
2017
.
Autophagy is required for endothelial cell alignment and atheroprotection under physiological blood flow.
Proc. Natl. Acad. Sci. USA
114
:
E8675
E8684
.
12.
De Meyer
G. R.
,
M. O.
Grootaert
,
C. F.
Michiels
,
A.
Kurdi
,
D. M.
Schrijvers
,
W.
Martinet
.
2015
.
Autophagy in vascular disease.
Circ. Res.
116
:
468
479
.
13.
Nagaoka
I.
,
S.
Hirota
,
F.
Niyonsaba
,
M.
Hirata
,
Y.
Adachi
,
H.
Tamura
,
D.
Heumann
.
2001
.
Cathelicidin family of antibacterial peptides CAP18 and CAP11 inhibit the expression of TNF-alpha by blocking the binding of LPS to CD14(+) cells.
J. Immunol.
167
:
3329
3338
.
14.
Nagaoka
I.
,
M.
Hirata
,
K.
Sugimoto
,
Y.
Tsutsumi-Ishii
,
A.
Someya
,
K.
Saionji
,
J.
Igari
.
1998
.
Evaluation of the expression of human CAP18 gene during neutrophil maturation in the bone marrow.
J. Leukoc. Biol.
64
:
845
852
.
15.
Shen
D.
,
J.
Coleman
,
E.
Chan
,
T. P.
Nicholson
,
L.
Dai
,
P. W.
Sheppard
,
W. F.
Patton
.
2011
.
Novel cell- and tissue-based assays for detecting misfolded and aggregated protein accumulation within aggresomes and inclusion bodies.
Cell Biochem. Biophys.
60
:
173
185
.
16.
Yuk
J. M.
,
D. M.
Shin
,
H. M.
Lee
,
C. S.
Yang
,
H. S.
Jin
,
K. K.
Kim
,
Z. W.
Lee
,
S. H.
Lee
,
J. M.
Kim
,
E. K.
Jo
.
2009
.
Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin.
Cell Host Microbe
6
:
231
243
.
17.
Stolz
A.
,
A.
Ernst
,
I.
Dikic
.
2014
.
Cargo recognition and trafficking in selective autophagy.
Nat. Cell Biol.
16
:
495
501
.
18.
Komatsu
M.
,
S.
Waguri
,
M.
Koike
,
Y. S.
Sou
,
T.
Ueno
,
T.
Hara
,
N.
Mizushima
,
J.
Iwata
,
J.
Ezaki
,
S.
Murata
, et al
2007
.
Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice.
Cell
131
:
1149
1163
.
19.
Shigihara
N.
,
A.
Fukunaka
,
A.
Hara
,
K.
Komiya
,
A.
Honda
,
T.
Uchida
,
H.
Abe
,
Y.
Toyofuku
,
M.
Tamaki
,
T.
Ogihara
, et al
2014
.
Human IAPP-induced pancreatic β cell toxicity and its regulation by autophagy.
J. Clin. Invest.
124
:
3634
3644
.
20.
Hung
S. Y.
,
W. P.
Huang
,
H. C.
Liou
,
W. M.
Fu
.
2009
.
Autophagy protects neuron from Abeta-induced cytotoxicity.
Autophagy
5
:
502
510
.
21.
Barth
S.
,
D.
Glick
,
K. F.
Macleod
.
2010
.
Autophagy: assays and artifacts.
J. Pathol.
221
:
117
124
.
22.
Zhang
Z.
,
P.
Meng
,
Y.
Han
,
C.
Shen
,
B.
Li
,
M. A.
Hakim
,
X.
Zhang
,
Q.
Lu
,
M.
Rong
,
R.
Lai
.
2015
.
Mitochondrial DNA-LL-37 complex promotes atherosclerosis by escaping from autophagic recognition.
Immunity
43
:
1137
1147
.
23.
Rekha
R. S.
,
S. S.
Rao Muvva
,
M.
Wan
,
R.
Raqib
,
P.
Bergman
,
S.
Brighenti
,
G. H.
Gudmundsson
,
B.
Agerberth
.
2015
.
Phenylbutyrate induces LL-37-dependent autophagy and intracellular killing of Mycobacterium tuberculosis in human macrophages.
Autophagy
11
:
1688
1699
.
24.
Suzuki
K.
,
T.
Murakami
,
Z.
Hu
,
H.
Tamura
,
K.
Kuwahara-Arai
,
T.
Iba
,
I.
Nagaoka
.
2016
.
Human host defense cathelicidin peptide LL-37 enhances the lipopolysaccharide uptake by liver sinusoidal endothelial cells without cell activation.
J. Immunol.
196
:
1338
1347
.
25.
Sancho-Vaello
E.
,
D.
Gil-Carton
,
P.
François
,
E. J.
Bonetti
,
M.
Kreir
,
K. R.
Pothula
,
U.
Kleinekathöfer
,
K.
Zeth
.
2020
.
The structure of the antimicrobial human cathelicidin LL-37 shows oligomerization and channel formation in the presence of membrane mimics.
Sci. Rep.
10
:
17356
.
26.
Koczulla
R.
,
G.
von Degenfeld
,
C.
Kupatt
,
F.
Krötz
,
S.
Zahler
,
T.
Gloe
,
K.
Issbrücker
,
P.
Unterberger
,
M.
Zaiou
,
C.
Lebherz
, et al
2003
.
An angiogenic role for the human peptide antibiotic LL-37/hCAP-18.
J. Clin. Invest.
111
:
1665
1672
.
27.
Byfield
F. J.
,
Q.
Wen
,
K.
Leszczynska
,
A.
Kulakowska
,
Z.
Namiot
,
P. A.
Janmey
,
R.
Bucki
.
2011
.
Cathelicidin LL-37 peptide regulates endothelial cell stiffness and endothelial barrier permeability.
Am. J. Physiol. Cell Physiol.
300
:
C105
C112
.
28.
Ridker
P. M.
,
B. M.
Everett
,
T.
Thuren
,
J. G.
MacFadyen
,
W. H.
Chang
,
C.
Ballantyne
,
F.
Fonseca
,
J.
Nicolau
,
W.
Koenig
,
S. D.
Anker
, et al
CANTOS Trial Group
.
2017
.
Antiinflammatory therapy with canakinumab for atherosclerotic disease.
N. Engl. J. Med.
377
:
1119
1131
.

The authors have no financial conflicts of interest.

This article is distributed under The American Association of Immunologists, Inc., Reuse Terms and Conditions for Author Choice articles.

Supplementary data