1. Introduction
Atherosclerosis (AS) is a chronic disease characterized by plaque accumulation within the arterial walls, resulting in severe cardiovascular events such as heart attack and stroke [
1]. Foam cell formation is a distinguished feature of AS and contributes to the formation of fatty streaks, which are the initial visible indication of AS [
2,
3,
4]. The process begins with abnormal lipid metabolism and the accumulation of low-density lipoprotein (LDL) in the arterial walls [
5]. Next, LDL interacts with oxidizing agents such as free radicals or reactive oxygen species, which initiate oxidation reactions and lead to the formation of oxidized low-density lipoprotein (ox-LDL) [
6]. The deposition of ox-LDL triggers the release of proinflammatory factors and promotes monocyte differentiation into macrophages, which further develop into foam cells through the regulation of ox-LDL uptake and cholesterol efflux [
7,
8]. Peroxisome proliferator-activated receptor γ (PPARγ) is a critical transcription factor to regulate ox-LDL uptake and cholesterol efflux in foam cells [
9]. The activation of PPARγ upregulates ATP binding box transporter A1/G1 (ABCA1/ABCG1) expression, thereby promoting cellular cholesterol efflux [
10]. In addition, PPARγ binds with RXRα to form heterodimers, which interact with the peroxisome proliferator-responsive element (PPRE) motif in the cluster of differentiation 36 (CD36) promoter, leading to ox-LDL uptake [
11,
12]. Targeting PPARγ to regulate cholesterol uptake and efflux is an important strategy for treating AS [
13].
Statins are typically the first choice for the treatment of AS due to their ability to lower low-density lipoprotein cholesterol (LDL-C) levels, reduce inflammatory responses, and stabilize atherosclerotic plaques [
14,
15]. Although statins display considerable efficacy and wide applicability, their side effects, such as muscle soreness and hepatotoxicity, cause numerous patients to cease therapy even if their lipid levels fail to meet the recommended guidelines for ceasing treatment [
16]. Rosiglitazone (RSG), a type of thiazolidinedione (TZD) that activates PPARγ and upregulates ABCA1 and ABCG1 expression, thereby increasing cholesterol efflux from macrophages [
17,
18]. However, it should be used cautiously because of the risk of mild anemia and the potential for increased heart failure [
19]. In recent years, several novel drugs that target lipid metabolism have been approved for the treatment of AS. For example, alirocumab (AOC) and evolocumab (EVO), two monoclonal antibodies that inhibit PCSK9, increase the expression of LDL receptors on the surface of hepatocytes, thereby reducing LDL-C levels [
20,
21]. Although monoclonal antibody drugs are highly effective and precise, they are expensive and may cause immune reactions [
22]. Therefore, developing safer, more effective and cost-efficient alternative therapies is crucial.
Antimicrobial peptides (AMPs), a subclass of bioactive peptides, are evolutionarily conserved defense molecules produced by all living organisms, typically comprising 12–50 amino acids [
23]. AMPs can be classified based on their biological sources, structural features, and target microorganisms [
23]. Based on biological sources, they are categorized as animal-derived, plant-derived, microbial-derived, or synthetic/engineered variants. Structurally, these peptides adopt distinct conformations including α-helical, β-sheet, extended/linear, and cyclic architectures. Functionally, AMPs are differentiated into antibacterial, antifungal, antiviral, and broad-spectrum subgroups. Unlike other bioactive peptides, AMPs are traditionally recognized for their direct antibacterial activities via membrane disruption [
24].However, recent research has uncovered unconventional roles of AMPs in: modulating immune responses (e.g., human cathelicidin LL-37) [
25,
26], promoting wound repair (e.g., β-defensin) [
27], and regulating metabolic pathways (e.g., adipocetin) [
28].
Recent studies have explored the potential of AMPs in combating AS because of their ability to modulate lipid metabolism and reduce inflammation [
29]. LL-37 is a cathelicidin-like AMP found in humans that is expressed predominantly in macrophages, and the levels of LL-37 are higher in atherosclerotic lesions than in normal blood vessels [
30]. A recent study reported that LL-37 inhibits cholesterol accumulation by decreasing the expression of CD36 through the ERK signaling pathway [
31]. Additionally, PR-39 is a natural AMP that has cardioprotective effects by inhibiting inflammation through inactivating the proteasome-mediated degradation of IκBα [
31].
Our previous research revealed that the frog skin AMP temporin-1CEa and its analogs exhibited anti-atherosclerotic effects by reducing the formation of macrophage-derived foam cells [
32]. The frog skin AMP chensinin-1b was designed in our lab by replacing the hydrophobic and hydrophilic residues on the opposite side of the parent peptide chensinin-1 [
33]. Moreover, AMP 3-13 was synthesized by truncating positions 3 to 13 of chensinin-1b, and AMP W3R6 was obtained by replacing the His residues at positions 4 and 10 with Arg residues of AMP 3-13 [
34,
35]. Our previous study has revealed the antibacterial activity and the structural characteristics of the AMPs [
34]. All of the AMPs exhibited a relatively high positive charge density. Notably, the NMR structure of chensinin-1b revealed a partially α-helical region (residues 8–14) in a membrane-mimetic environment. In comparison, AMP 3-13 demonstrated a higher α-helical content than chensinin-1b [
34]. In this study,
ApoE-/- mice were fed a high-fat diet (HFD) to establish an AS mouse model, and THP-1 cells were induced with ox-LDL to establish foam cell model. The effects of AMP 3-13 and its analogs, particularly AMP 3-13, on
ApoE-/- AS mice and foam cell formation were investigated. In addition, the underlying mechanism of how ox-LDL uptake and cholesterol efflux are regulated in foam cells was elucidated. This study provides novel insights into the potential of 3-13 and its analogs as therapeutic agents for the prevention and treatment of AS.
2. Materials and Methods
2.1. Reagents
ox-LDL was purchased from Yiyuan Biotechnology Co., Ltd. (Guangzhou, China). The frog skin peptides chensinin-1b, W3R6 and 3-13 were synthesized by GL Biochemistry, Inc. (Shanghai, China), and the purity of the peptides was greater than 95%. The amino acid sequences and physicochemical information of chensinin-1b, W3R6 and 3-13 are summarized in
Table S1. RSG and T0070907 was purchased from MedChemExpress Co., Ltd. (Shanghai, China), and dissolved in DMSO (solubilities 175 and 62.5 mg/mL).
2.2. ApoE-/- Mouse Model of AS
All experimental protocols were approved by the Medical Ethics Committee of Liaoning Normal University. ApoE-/- mice were purchased from Vital River Laboratory Animal Technology (Beijing, China). ApoE-/- mice were 6 weeks old and maintained on a SPF C57BL/6 background. ApoE-/- mice were identified at 4 weeks after birth. The mouse tail genotype was identified by using a mouse genotyping kit, and ApoE-/- homozygous mice were used in subsequent experiments. ApoE-/- female mice aged 6 weeks were fed a HFD to establish an AS mouse model. After the HFD was continued for 6 weeks, the mice were randomly divided into the ctrl, HFD and HFD+AMP 3-13 groups, with 10 mice in each group. The AMP 3-13 were dissolved in 0.9% saline and administered intraperitoneally daily for 14 days. The body weight, activity and mental status of the mice in each group were observed and recorded every day.
2.3. Aorta Oil Red O (ORO) Staining
After anesthesia, plasma was collected from the mice in each group. The abdominal cavity and thoracic cavity of each mouse were cut open, the excess tissue around the aortic vessels was removed to expose the intact aortic vessels, and the perivascular adipose tissue was carefully removed. The whole aortic vessel was isolated by clipping from the proximal end, along the spine of the mouse to the iliac artery, with small spring-loaded scissors. The scissors were inserted into the proximal end of the aorta, and the aortic arch, non-aorta, left common carotid artery, the left subclavian artery, the thoracic aorta, the abdominal aorta and the left and right iliac arteries were harvested along the inner curvature of the aortic arch. Finally, the whole blood vessels were isolated. Staining was performed with an ORO staining kit (G1260, Solarbio, Shanghai, China).
2.4. Histological Detection
The aortic vessel was fixed in 4% paraformaldehyde in phosphate-buffered saline and embedded in paraffin. The paraffin-embedded tissue was cut into 4 μm-thick sections. The sections were stained with hematoxylin and eosin (HE, Sigma-Aldrich, St. Louis, MO, USA).
2.5. Plasma Lipid Factor Assay
After 14 days, the mice in each group were anesthetized, and the eyeball blood was collected in an anticoagulant tube containing EDTA. The whole blood was mixed with the anticoagulant thoroughly and centrifuged at 3000 rpm for 10 min, and the resulting mixture was stored at −80 °C. Total cholesterol (T-CHO), LDL-C, triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C) kits from Nanjing Jian Cheng Bioengineering Institute Co., Ltd. (Item No.: A111-1-1, A113-1-1, A110-1-1 and A112-1-1) were used to detect blood lipid parameters. The detection ranges of the kits are 0–19.39, 0–10.40, 0.30–11.40 and 0–5.16 mmol/L, respectively. The sensitivities are 2.00 mmol/L (absorbance 0.10–0.40), 2.60 mmol/L (absorbance 0.18–0.28), 2.70 mmol/L (absorbance 0.20–0.40) and 1.00 mmol/L (absorbance > 0.04), respectively. The results were obtained using Multiskan FC microplate reader (Thermo Fisher Scientific, Waltham, MA, USA).
2.6. RNA-Seq Assay
HFD-induced ApoE-/- AS mice were treated with AMP 3-13 as previously described. The mice were randomly divided into the following groups: the ctrl, HFD and HFD+3-13 groups with three replicates in each group. The mice in each group were anesthetized with sodium pentobarbital (50 mg/kg I.P), the abdominal cavity was washed with about 1 to 2 mL cold PBS containing 10 mM EDTA, the peritoneal exudate was collected and centrifuged at 400 g for 10 min. The cell pellets were washed twice with PBS and then resuspended in 0.2 mL DMEM to count the cells. The isolated macrophages were plated separately in 6-well tissue culture plates and cultured with RPMI/10% FBS overnight. The next morning, nonadherent cells were removed by aspiration, adherent cells (macrophages) were washed with PBS three times and harvested for the experiments. 600 μL Trizol lysate was added to collect the cells, and then the cells were frozen in liquid nitrogen for 20 min and transferred to the −80 °C refrigerator for storage. Samples were sent to the company for RNA-seq analysis.
Total RNA purity and concentration were assessed using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA). RNA integrity was verified with an Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA), where all samples met the following quality thresholds: RNA Integrity Number (RIN) ≥ 8.0, OD
260/280 > 1.9 and Total RNA > 500 ng. Sequencing libraries were constructed from 1 μg qualified RNA using the VAHTS Universal V6 RNA-seq Library Prep Kit (Vazyme Biotech, Nanjing, China) according to manufacturer protocols. The transcriptome sequencing and analysis were conducted by OE Biotech Co., Ltd. (Shanghai, China). Library sequencing was performed on an Illumina NovaSeq 6000 platform (Illumina Inc., San Diego, CA, USA) to generate 150 bp paired-end reads. Adaptor trimming and low-quality read removal using fastp (v0.23.2) [
36]. Clean reads were mapped to the
Mus musculus reference genome (Genome assembly GRCm39) using HISAT2 (v2.2.1) [
37] with default parameters. Gene-level counts were generated by HTSeq-count (v0.13.5) [
38]. Differentially expressed genes (DEGs) were identified using DESeq2 (v1.30.1) [
39] with the following criteria: |log
2 (fold change)| > 1 and Benjamini–Hochberg adjusted
p-value < 0.05. Hierarchical clustering of DEGs was performed in R (v4.2.0) using Euclidean distance with complete linkage. Volcano plot and heatmaps generated with bioinformatics platform, and WikiPathways enrichment bubble plots created using ggplot2 (v3.3.5). The raw RNA-seq data has been submitted to the NCBI SRA datasets with accession number PRJNA1290911.
2.7. Cell Culture
The human leukemia monocytic cell line THP-1 was purchased from Jiangsu Kaiji Biology Co., Ltd. (Nanjing, China). The cells were cultured in RPMI 1640 medium supplemented with 10% FBS and 1% penicillin and streptomycin. Next, THP-1 cells were induced to differentiate into macrophages by incubation with 100 ng/mL phorbol 12-myristate 13-acetate (PMA, MedChemExpress Co., Ltd., Shanghai, China) for 48 h, during which the cells adhered to the culture surface. Finally, to synchronize the macrophages, the medium was replaced with serum-free RPMI 1640 medium and the cells were cultured for an additional 24 h. The in vitro assays were divided into the control group (ctrl, THP-1 cells), ox-LDL group (ox-LDL, 100 μg/mL), PPARγ agonist group (ox-LDL+RSG, 20 μM), PPARγ inhibitor group (ox-LDL+T0070907, 10 μM) and AMPs groups (ox-LDL+3-13 or its analogs, 6.25, 12.5 and 25 μM).
2.8. Quantitative Real-Time PCR
The samples of aortic arch tissue in each group were the same as before. Macrophages were seeded in 6-well plates and then treated with ox-LDL, 3-13 and its analogs for an additional 24 h. RSG (20 μM) was added to the PPARγ agonist group. The total RNA of aortic arch tissue and macrophages were extracted with TRIzol reagent (15596018CN; Invitrogen, Shanghai, China). Reverse transcription was performed via a Super Script™ III kit (18080400, Invitrogen, Shanghai, China). cDNA amplification was performed in an ABI Prism 7500 Fast sequence detection system (Applied Biosystems, San Francisco, CA, USA) using a TB Green Premix Ex Taq II kit (RR820Q, TaKaRa, Dalian, China). The primer sequences are summarized in
Table S2. The relative mRNA expression levels were normalized to that of GAPDH (internal control) by using the Ct values calculated according to the manufacturer’s instructions.
2.9. Western Blot Analysis
The samples of aortic arch tissue in each group were the same as before. Macrophages were seeded in 6-well plates and then treated with drugs in each group. The aortic arch tissue and macrophages in each group were lysed in RIPA lysis buffer. After protein quantification using a BCA kit, the protein in the cell lysates was separated via 10–12% SDS–PAGE before being electrotransferred to a PVDF membrane via standard procedures. After being blocked with 5% skim milk in TBST for 1 h at room temperature, the membranes were incubated with specific primary antibodies at 4 °C overnight. The primary antibodies used are summarized in
Table S3. The relative protein expression levels were normalized to those of histone H3 (nuclear protein) or ATP1A1 (membrane protein). Then, the samples were incubated with secondary antibodies conjugated to HRP. Bands were visualized with an Azure Biosystems c500 instrument using ECL-Plus detection reagents (Santa Cruz, CA, USA). Densitometric quantification of the protein concentration was performed via Image-Pro Plus software (Version 6.0.0.260).
2.10. Immunohistochemical
The aortic vessel sections were described as before. After hydration, sodium citrate antigen repair solution was added to the sections in each group, which were then microwaved at 700 W for 5 min and 250 W for 10 min and cooled to room temperature. Endogenous peroxidase was removed by adding 3% H
2O
2, blocking with 5% BSA at room temperature for 1 h, adding primary antibodies and incubating overnight in a wet box at 4 °C. The primary antibodies used are summarized in
Table S3. HRP-labeled secondary antibodies were added, and the samples were incubated for 1 h at 37 °C. DAB was added for staining in the dark for 3–10 min. Finally, photographs were taken with a microscope.
2.11. Cell Proliferation Assay
The growth rate of the THP-1 cells and the concentration of ox-LDL were determined via real-time cellular analysis (RTCA). Fifty microliters of medium were added to the RTCA plate to determine the response baseline, and then 100 μL of cell suspensions with densities of 5 × 104, 1 × 105, 1.5 × 105 and 2 × 105 cells/mL were added. After 48 h, serum-free 1640 medium was added to the well plate, and the plate was treated for 24 h. ox-LDL concentrations of 50, 100, and 200 μg/mL were added, and the proliferation curves of each group at 24 h were generated via RTCA. In accordance with the results of the RTCA and ox-LDL concentration experiments, 1 × 105 cells/mL was selected as the density of THP-1 cells and 100 μg/mL was selected as the concentration of ox-LDL for the subsequent experiments. THP-1 cells were seeded in 96-well plates at a density of 1 × 105 cells/mL. After differentiation and synchronization, 100 μg/mL ox-LDL was added to each group of cells (ox-LDL was not added to the AMP-only treatment groups). The frog skin peptides chensinin-1b, W3R6 and 3-13 were subsequently added at concentrations of 3.125, 6.25, 12.5, 25, 50 and 100 μM, respectively. After 24 h, 10% final concentration of CCK8 was added to each well, and the samples were incubated for 4 h in the dark. The absorbance of each well was measured with a Multiskan FC microplate reader (Thermo Fisher Scientific, Waltham, MA, USA) at 490 nm.
2.12. Cholesterol Detection
Macrophages were seeded in 96-well plates, and the cells in each group were treated with drugs. In accordance with the manufacturer’s instructions (BC1985/BC1895, Solarbio, Shanghai, China), total cholesterol (TC) or free cholesterol (FC) working solution was added, and 20 μL of blank control solution, standard solution or sample mixture was added. Each group of samples was incubated for 15 min at 37 °C. The absorbance of each well was measured with a Multiskan FC microplate reader (Thermo Fisher Scientific, Waltham, MA, USA) at 500 nm. Cellular protein was measured with a BCA protein assay kit according to the manufacturer’s instructions (MA0082, Meilunbio, Dalian, China). TC and FC were normalized to the cellular protein levels. Cholesterol ester (CE) levels were calculated via the following formula: CE = TC − FC.
2.13. ORO Staining
Macrophages were seeded in 24-well plates and then treated with ox-LDL and/or 3-13 and its analogs for an additional 24 h. Lipid droplets in ox-LDL-stimulated macrophages were observed via ORO staining (G1262, Solarbio, Shanghai, China). In brief, ORO fixative solution was added, and the samples were fixed for 30 min at room temperature. The cells were then incubated in 60% isopropyl alcohol for 5 min. ORO was added to the wells and incubated for 15 min at room temperature. The cells were then counterstained with Mayer’s hematoxylin. Images were acquired via a light microscope (Leica, Wetzlar, Germany).
2.14. Nitrobenzoxadiazole (NBD)-Labeled Cholesterol Detection
After drug treatment, NBD-labeled cholesterol (5 μM), 3-13 and its analogs prepared in RPMI 1640 medium without phenol red were added to the THP-1 cells. After 3 h, the cells were washed with PBS, apolipoprotein A-1 (apoA-1) and HDL at concentrations of 10 μg/mL prepared in phenol red-free RPMI 1640 medium were added, and the cells were cultured for another 4 h. The supernatant of each group was collected, which was the cholesterol effluent. After the THP-1 cells were lysed by the addition of 0.1% Triton X-100, the cell lysates from each group were collected. The fluorescence intensity of each sample was measured by a Multiskan FC microplate reader (Thermo Fisher Scientific, Waltham, MA, USA), the excitation wavelength was 469 nm, and the emission wavelength was 537 nm. The cholesterol efflux rates of each group were calculated.
2.15. Dil-ox-LDL Uptake Detection
THP-1 cells were seeded in 12-well plates at a density of 1 × 105 cells/mL. After the differentiation and synchronization of the cells in each group, 40 μg/mL (Dil dye-labeled ox-LDL) Dil-ox-LDL prepared in phenol red-free RPMI 1640 medium was added, and AMPs were added at the same time. After 24 h, the cells in each group were washed three times with PBS, and then Hoechst 33342 was added and incubated for 1 h. The slides were removed and sealed by the addition of 5 μL of anti-fluorescence quenching agent. The slides were photographed under a laser confocal microscope at an excitation wavelength of 554 nm and an emission wavelength of 571 nm for Dil-ox-LDL and at an excitation wavelength of 350 nm and an emission wavelength of 461 nm for Hoechst 33342.
2.16. Cellular Localization of AMPs
THP-1 cells were seeded in 12-well plates at a density of 1 × 105 cells/mL. After the differentiation and synchronization of the cells in each group, the FITC-labeled antimicrobial peptide 3-13 and its analogs prepared in phenol red-free RPMI 1640 medium were added. After 6 h, the cells in each group were washed three times with PBS, and then Hoechst 33342 was added and incubated for 1 h. The slides were removed and sealed with 5 μL of anti-fluorescence quenching agent. The slides were photographed under a laser confocal microscope at an excitation between of 490–495 nm and emission wavelength between 520 and 530 nm for FITC-AMPs and at an excitation wavelength of 350 nm and an emission wavelength of 461 nm for Hoechst 33342.
2.17. Dual-Luciferase Reporter Gene Assay
Total mRNA was extracted from THP-1 cells, and the PPARγ gene was amplified via high-fidelity PCR. The target fragment and vector were digested by a double enzyme, and the gel was recovered, ligated, and transformed into E.coli DH5α, which was subsequently seeded in plates with lysogeny broth (LB). Positive clones were screened, and gene sequencing was performed to obtain the PPARγ overexpression plasmid. HEK293T cells were used for plasmid transfection, and the cells were divided into a vector group, a ctrl group and an AMP group. First, the PPARγ overexpression plasmid was transfected. After the transfection efficiency was determined, the transfected cells were subsequently transfected with a CD36 promoter reporter gene plasmid and an internal control reporter gene plasmid. After 6 h, AMP 3-13 were added and detected with a TransDetect® Double-Luciferase Reporter Assay Kit (FR201, Trans, Beijing, China), and after 12 h, the fluorescence intensities of each group were calculated.
2.18. ChIP Assay
Studies have shown that PPARγ forms a heterodimer with RXRα, which regulates the expression of target genes by acting on the PPRE sequence of the target gene promoter region. The sequence feature was TGACCT X TGACCT (X is 1 to 2 bases). First, the promoter region of the human
CD36 gene was searched using the UCSC website (
https://genome.ucsc.edu/, accessed on 15 August 2023). Then, using the JASPAR website (
https://jaspar.elixir.no/analysis, accessed on 15 August 2023), the PPARγ control
CD36 promoter region locus was determined to be TATGACCTAATGAACTAA. The following ChIP primers were designed according to the PPRE sequence of the
CD36 gene promoter region: F: 5′-CAAAAAGGACAGCACGAGCA-3′, R: 5′-ATGCATTCAAACAACCTTAGAAGT-3′. A ChIP kit was used to detect the effects of 3-13 and its analogs on the binding of PPARγ to the
CD36 promoter in THP-1 foam cells. THP-1 cells were seeded in culture flasks at a density of 1 × 10
5 cells/mL. After the differentiation and synchronization of the THP-1 cells in each group, ox-LDL, 3-13 and its analogs were added. The cells were divided into negative control, positive control, target protein and input groups according to the instructions of the ChIP kit. The binding of PPARγ to the
CD36 promoter in each group was detected by agarose gel electrophoresis.
2.19. Electrophoretic Mobility Shift Assay (EMSA)
The 3′ terminal biotin-labeled PPRE sequence probes were purchased from Sangon Biotech (GS009, Shanghai, China), and the EMSA experiment was performed using the Chemiluminescent EMSA Kit (Beyotime Biotechnology, Shanghai, China) according to the manufacturer’s instructions. The AMPs were incubated with PPRE sequence probes at room temperature. Loading buffer was then added to the reaction mixture, and PAGE electrophoresis was performed at 120 V on a 6% TBE gel. Then, the membrane transfer, ultraviolet crosslinking and blocking reaction of the EMSA gel were carried out successively. Streptavidin-HRP conjugate was added, and the mixture was incubated for 15 min. After washing with buffer, the membrane was incubated in substrate balance buffer for 5 min before ECL development. Bands were visualized with an Azure Biosystems c500 instrument using ECL-Plus detection reagents (Santa Cruz, CA, USA). Densitometric quantification of the protein concentration was performed via Image-Pro Plus software (Version 6.0.0.260).
2.20. Statistical Analysis
SPSS 18.0 software (SPSS Inc., Chicago, IL, USA) was used to analyze all the experimental data. The experimental results were analyzed by one-way univariate analysis of variance (ANOVA) by Tukey’s test for multiple corrections between groups. sample sizes and statistical method were described in each figure legend. p < 0.05 was considered statistically significant, and the experimental data are presented as the means ± standard deviations.
4. Discussion
Atherosclerosis is the primary risk factor for cardiovascular disease, which is the leading cause of morbidity and mortality in developed countries, and is becoming increasingly widespread in developing countries [
1,
43,
44]. Foam cell formation is a critical event in the early stages of AS, which are characterized by abnormally high levels of cholesterol and cholesteryl esters [
2,
3,
45]. Inhibiting foam cell formation is crucial for developing therapeutic strategies to prevent and treat AS [
13]. For instance, LL-37 has been shown to mitigate cholesterol accumulation by downregulating CD36 expression through the ERK signaling pathway [
29]. Additionally, PR-39 exerts cardioprotective effects by inhibiting proteasome-mediated degradation of IκBα, thereby suppressing inflammatory responses [
31]. Our study previously demonstrated that the frog skin peptide temporin-1CEa and its analogs inhibited ox-LDL uptake in foam cells by downregulating CD36 expression, consequently ameliorating the accumulation of cholesterol within cells [
32]. In this study, three frog skin AMPs 3-13, W3R6 and chensinin-1b, were designed and synthesized on the basis of the parent peptide chensinin-1, a natural peptide that was isolated from
Rana chensinensis. To investigate the effects of these AMPs on AS, we utilized
ApoE-/- mice fed a high-fat diet (HFD) to establish an AS mouse model and induced THP-1 cells with ox-LDL to create a foam cell model. Here, we revealed that AMP 3-13 and its analogs alleviate AS through a dual mechanism: activating the PPARγ-ABCA1/ABCG1 signaling pathway to promote cholesterol efflux, and inhibiting CD36-dependent ox-LDL uptake by competitively binding to the
CD36 promoter, thereby disrupting PPARγ-mediated transcriptional activation.
Atherosclerotic plaque formation is primarily driven by dysregulated cholesterol metabolism. To evaluate the therapeutic potential of AMP 3-13, we first examined its effects in
ApoE-/- AS mice. The results demonstrated that AMP 3-13 significantly decreased weight gain, aortic root plaque formation, and the plasma cholesterol levels in
ApoE-/- AS mice, suggesting that the AMP might alleviate AS by regulating cholesterol metabolism in
ApoE-/- mice. To investigate the effect of AMP 3-13 on the regulation of AS, transcriptome was performed to analyze the differentially expressed genes in the peritoneal macrophages, which were isolated from
ApoE-/- AS mice following treatment with or without AMP 3-13. Wikipathways enrichment analysis revealed that AMP 3-13 treatment regulated the gene expression related with cholesterol metabolism and the PPAR signaling pathway, with notable changes in the expression of PPARγ, ABCA1, ABCG1 and CD36. Notably, PPARγ is a critical transcription factor involved in maintaining cellular cholesterol homeostasis and foam cell formation. Previous studies have suggested an important role of the PPARγ-LXRα-ABCA1/ABCG1 pathway in regulating cholesterol efflux, which could be a promising target for treating AS [
17,
46]. For example, mangiferin, a xanthonoid from
Salacia oblonga, promotes macrophage cholesterol efflux and protects against AS by upregulating ABCA1 and ABCG1 expression [
17]. Additionally, Bu1 and Bu2 peptides inhibit foam cell formation by activating the PPARγ/LXRα signaling pathway and promoting cholesterol efflux [
46]. In this study, AMP 3-13 and its analogs increased the expression of PPARγ, ABCA1 and ABCG1 in foam cells. In addition, a PPARγ inhibitor (T0070907) attenuated the increased expression of ABCA1 and ABCG1 induced by AMP 3-13 and its analogs, indicating that these AMPs increased the expression of ABCA1 and ABCG1 via the PPARγ-mediated pathway to promote cholesterol efflux.
It has been reported that PPARγ plays a crucial role in regulating ox-LDL uptake via CD36 [
42]. Mechanically, PPARγ binds with RXRα to form heterodimers, which interact with the PPRE motif in the
CD36 promoter [
11,
12]. Inactivation of the PPARγ/CD36 signaling pathway is a potential strategy for treating AS [
47,
48,
49]. Tamoxifen, a selective estrogen receptor modulator, inhibits CD36 expression and cellular ox-LDL accumulation by downregulating PPARγ and disrupting the interaction between PPARγ and PPRE in the
CD36 promoter [
47]. In addition, numerous natural products, such as black mulberry ethanol extract and spiromastixones, exert anti-AS effects by inhibiting PPARγ and CD36 expression [
48,
49]. Our results revealed that AMP 3-13 and its analogs upregulated PPARγ expression but downregulated CD36 expression. We proposed that AMPs might disrupt the binding ability of PPARγ to the
CD36 promoter. This hypothesis was verified via dual-luciferase reporter gene and ChIP assays. The results demonstrated that AMP 3-13 and its analogs were localized in the nucleus, particularly in the nucleolus, suggesting their potential to interfere with the binding of PPARγ to the
CD36 promoter. The dual-luciferase reporter gene results revealed that AMP 3-13 and its analogs reduced the activity of the
CD36 promoter. The ChIP results indicated that the AMPs decreased the binding ability of PPARγ to the
CD36 promoter. The EMSA results further demonstrated that the AMPs bound directly to the PPRE motif.
The mechanisms of canonical AMPs primarily involve membrane disruption or intracellular targeting to disrupt protein–protein interactions. Direct nuclear localization and competitive interference with specific transcription factor–DNA interactions represent a less conventional mechanism for AMPs. However, there are some examples of stapled peptides that disrupt protein–DNA interactions. For instance, hydrocarbon-stapled peptides competitively bind to Nrf2 response elements, thereby inhibiting Nrf2–DNA interactions and suppressing Nrf2 transcriptional activation in cancer cells [
50,
51]. Similarly, a stapled peptide based on GCN4 retains DNA-binding ability and enhancing cellular uptake [
52,
53]. Although AMP 3-13 and stapled peptides demonstrate structural differences, they share common physicochemical properties including a high density of cationic amino acids and a strong propensity for α-helix formation. These conserved features likely contribute to competitively inhibit DNA binding to transcription factor domains. This suggests a novel peptide mechanism involving direct modulation of transcriptional complexes at genomic regulatory elements.
However, several important limitations must be acknowledged. Firstly, despite our results demonstrated that AMP 3-13 and its analogs bound directly to the PPRE motif and decreased the binding ability of PPARγ to the
CD36 promoter, we could not definitively conclude whether the observed AMP-PPRE interaction represents a sequence-specific recognition event or stems from non-specific electrostatic interactions. Electrostatic interactions between the negatively charged DNA and positively charged regions of the protein play a role in these non-specific DNA–protein interactions [
54]. Due to its charge properties, the binding of AMPs to DNA probably represents non-specific DNA–protein interactions. However, it cannot totally explain for the results in our study for the following reasons: (a) AMP 3-13 and its analogs treatment led to a significant upregulation of PPARγ expression while significantly downregulation of CD36 expression. If AMPs bound with DNA through non-specific manner, a broader suppression of gene expression would be observed. Notably, we found that the specific suppression of CD36, while the expression levels of ABCA1 and ABCG1, which are also downstream targets of PPARγ, are upregulated. These results suggested a mechanism involving intervention at specific promoter responsive element rather than a global, non-specific block. (b) AMP 3-13 and its analogs demonstrated strictly concentration-dependent effects on PPRE-binding affinity (EMSA,
Figure 6F), and the interaction between PPARγ and the
CD36 promoter (ChIP,
Figure 6D,E). Although non-specific binding interactions may exhibit limited concentration dependence, they generally fail to produce the precisely calibrated, target-specific dose–response relationships observed in our study. (c) Furthermore, EMSA results (
Figure 6F) showed that AMP 3-13 and its analogs binding to the target promoter probe formed discrete bands with specific mobility shifts, rather than diffuse smearing or precipitation. Although this did not entirely rule out non-specificity, the presence of discrete bands was generally more indicative of ordered binding or complex formation, rather than purely random, non-specific binding. Collectively, while non-specific DNA binding may contribute to initial AMPs interactions, the ChIP and EMSA results indicated a model of specific competitive interference at PPARγ/CD36 promoter regions. The definitive proof of sequence-specific recognition between AMPs and PPRE would require structural studies (e.g., crystallography) or binding affinity assays comparing mutated PPRE sequences. Elucidating the precise mode of action will be a key focus of our future research. Secondly, the therapeutic potential of AMPs is currently constrained by limited safety assessment. Preliminary hepatic histology showed preserved liver architecture without necrosis or inflammation at the tested dose (10 mg/kg, 2 weeks;
Figure S6). However, comprehensive safety assessment including renal histopathology and serum biochemistry was not performed; the complete toxicity profile is to be established in the future.