BMB Reports 2024; 57(8): 375-380  https://doi.org/10.5483/BMBRep.2024-0055
miR-328-5p functions as a critical negative regulator in early endothelial inflammation and advanced atherosclerosis
Yangxia Zhang1,#, Yingke Li1,#, Zhisheng Han1, Qingyang Huo1, Longkai Ji1, Xuejia Liu2, Han Li2,*, Xinxing Zhu1,3,*  & Zhipeng Hao4,*
1Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, 2Stem Cells and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, 3Department of Respiratory and Critical Care Medicine, Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, First Affiliated Hospital, Bengbu Medical University, Bengbu 233004, 4Department of Thoracic Surgery of Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
Correspondence to: Han Li, Tel: +86-373-3029887; Fax: +86-373-3831322; E-mail: lihanxinyi@126.com; Xinxing Zhu, Tel: +86-373-3831393; Fax: +86-373-3831239; E-mail: zhuxx0105@hotmail.com; Zhipeng Hao, Tel: +86-27-83663217; Fax: +86-27-83662640; E-mail: samhzp@126.com
#These authors contributed equally to this work.
Received: April 10, 2024; Revised: April 26, 2024; Accepted: June 10, 2024; Published online: August 16, 2024.
© Korean Society for Biochemistry and Molecular Biology. All rights reserved.

cc This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
ABSTRACT
Early proatherogenic inflammation constitutes a significant risk factor for atherogenesis development. Despite this, the precise molecular mechanisms driving this pathological progression largely remain elusive. Our study unveils a pivotal role for the microRNA miR-328-5p in dampening endothelial inflammation by modulating the stability of JUNB (JunB proto-oncogene). Perturbation of miR-328-5p levels results in heightened monocyte adhesion to endothelial cells and enhanced transendothelial migration, while its overexpression mitigates these inflammatory processes. Furthermore, miR-328-5p hinders macrophage polarization toward the pro-inflammatory M1 phenotype, and exerts a negative influence on atherosclerotic plaque formation in vivo. By pinpointing JUNB as a direct miR-328-5p target, our research underscores the potential of miR-328-5p as a therapeutic target for inflammatory atherosclerosis. Reintroduction of JUNB effectively counteracts the anti-atherosclerotic effects of miR-328-5p, highlighting the promise of pharmacological miR-328-5p targeting in managing inflammatory atherosclerosis.
Keywords: Atherosclerosis, Inflammation, JUNB, Macrophage polarization, miR-328-5p
INTRODUCTION

Atherosclerosis is a complex process involving endothelial and smooth muscle cell activation, monocyte recruitment, and lipid accumulation in the artery walls. It starts with the retention of certain lipoproteins (LPs) in the artery lining, leading to inflammation and dysfunction (1-4). This early inflammation is a key risk factor for atherosclerosis development (5, 6). Inflammatory cells release molecules that attract immune cells, like macrophages, which contribute to plaque formation. Macrophages can take on different roles (M1 for inflammation, M2 for anti-inflammatory effects), to influence disease progression (7-10). Studies show that macrophages can switch between these roles, potentially impacting the development of atherosclerosis.

The AP-1 protein complex, a widely distributed dimeric structure consisting of Jun (e.g., JUN) and Fos (e.g., FOS) subunits, has been recognized for its pivotal roles in various inflammatory pathways. Growing evidence indicates that AP-1 transcription factors, such as c-Jun, JUNB, and JUND, can modulate the expression of crucial inflammatory molecules, thereby influencing the inflammatory response. A study highlighted the essential role of JUNB in modulating interleukin (IL)1β expression during classical inflammation in macrophages (11). Furthermore, c-Jun has been associated with Sirt6-mediated liver inflammation through the regulation of key pro-inflammatory factors, like IL6 and MCP1 (12).

MicroRNAs (miRNAs), a subset of non-coding RNAs approximately 22 nucleotides (nt) in length, play a crucial role in the regulation of RNA silencing and the post-transcriptional modulation of gene expression by binding to the 3’-UTR regions of specific targets (13-17). Comprehensive studies have shown that miRNAs typically recognize their targets through a (6-8) nt site that aligns with their seed region. Each miRNA has the capacity to silence hundreds of genes, while individual genes may be targeted by multiple miRNAs. Mounting evidence suggests that miRNAs participate in various cellular processes, such as inflammation, apoptosis, and cell cycle regulation, by directly or indirectly influencing key factors (18-22).

In this study, we first identified miR-328-5p as a novel regulator in early inflammation and advanced atherosclerosis, possibly through directly targeting JUNB to control its expression. Inhibition of miR-328-5p augments monocyte adhesion and transendothelial migration. Expressions of central inflammatory factors are significantly enhanced when exposed to miR-328-5p inhibitor treatment. Moreover, we find that miR-328-5p plays an inhibitory role on the M1 polarization of macrophages. Taken together, these findings suggest a suppressing function of miR-328-5p for inflammatory atherosclerosis.

RESULTS

miR-328-5p negatively regulates endothelial inflammation

Previous studies have indicated that miR-328 is involved in regulating endothelial cells injury induced by oxidized low-density lipoprotein (23). However, the specific role of miR-328-5p in inflammatory atherosclerosis has not been fully elucidated. To verify the functional involvement of miR-328-5p in regulating endothelial inflammation, we initially treated human umbilical vein endothelial cells (HUVECs) with tumor necrosis factor α (TNFα), a well-known pro-inflammatory inducer, to assess the expression changes of miR-328-5p in response to inflammatory injury. Notably, the qRT-PCR analysis revealed a down-regulation of miR-328-5p expression after TNFα treatment (Fig. 1A, left panel). Moreover, we established a high-fat diet atherosclerosis model to further confirm the decreased expression of miR-328-5p in response to inflammatory atherosclerosis in vivo (Fig. 1A, right panel). These findings suggest a potential role of miR-328-5p in regulating endothelial inflammation and the advanced progression of atherosclerosis.

Next, we conducted a monocyte adhesion assay to investigate the impact of miR-328-5p on endothelial inflammation using a synthetic inhibitor and mimic of miR-328-5p. Notably, inhibiting miR-328-5p in HUVECs significantly increased the adhesive capacity of monocytes to HUVECs (Fig. 1B). To validate this discovery, we investigated whether enforced miR-328-5p could mitigate the induced monocyte association with HUVECs triggered by TNFα, a well-known pro-inflammatory stimulus that induces the secretion of adhesion molecules (Fig. 1C). Consistently, the overexpression of miR-328-5p mimic markedly impeded TNFα-induced monocyte adhesion with endothelial cells (Fig. 1D). Furthermore, we employed transendothelial migration analysis to validate the negative role of miR-328-5p in endothelial inflammation. The results showed that overexpressing miR-328-5p mimic in HUVECs inhibited monocyte transmigration through endothelial monolayers (Fig. 1E). To explore the downstream signaling pathway mediated by miR-328-5p, we then established expression profiles in HUVECs with or without miR-328-5p mimic overexpression using high-throughput mRNA sequencing. As expected, a variety of inflammatory pathways were enriched after miR-328-5p aberrant overexpression (Fig. 1F). Collectively, these findings demonstrate the possible inhibitory role of miR-328-5p in regulating endothelial inflammation.

miR-328-5p attenuates the expressions of pro-inflammatory factors

Numerous pro-inflammatory factors are induced and play regulatory roles for the formation of an inflammatory microenvironment when endothelial cells are susceptible to pro-inflammatory stimuli. To examine the effect of miR-328-5p on the expression of these inflammatory genes, we analyzed the expression profiles of the mRNA sequencing data above, and found that 238 and 395 genes were up- and down-regulated, respectively, after miR-328-5p mimic overexpression in HUVECs (Fig. 2A). Further analysis suggested that multiple inflammatory factors involved in the regulation of early endothelial inflammation were significantly suppressed in HUVECs with enforced miR-328-5p overexpression (Fig. 2B). Moreover, qRT-PCR assay further confirmed the increased expressions of vascular cell adhesion molecule 1 (VCAM1), IL6, IL8, and IL1β after miR-328-5p inhibitor treatment (Fig. 2C). In parallel, protein expression changes in miR-328-5p-depleted HUVECs were determined by immunoblot analysis that showed that miR-328-5p attenuation significantly augmented the protein levels of these inflammatory genes (Fig. 2D). Consistent with this result, ELISA analysis further verified this inhibitory effect of miR-328-5p on pro-inflammatory factor secretion (Fig. 2E, F). To verify this regulatory role of miR-328-5p for these inflammatory factors, we overexpressed the mimic of miR-328-5p, then examined its effect on the expressions of pro-inflammatory factors. As expected, enforced overexpression of miR-328-5p greatly abrogated the expression levels of these inflammatory factors (Fig. 2G, H). Overall, these observations suggest that miR-328-5p negatively regulates the expressions of canonical pro-inflammatory factors.

Inhibition of miR-328-5p promotes the conversion of inflammatory macrophages to M1 phenotype

It is widely accepted that when suffering from inflammatory injury, macrophages were able to switch between M1 and M2 states. We therefore asked whether miR-328-5p plays a role in the modulation of macrophage polarization. To prove this hypothesis, we silenced miR-328-5p in THP-1 monocytes, which were then subjected to PMA induction for macrophage induction. Of note, miR-328-5p inhibition remarkably augmented the expression levels of M1-type marker genes, such as MCP1 and TNFα (Supplementary Fig. 1A). Conversely, attenuation of miR-328-5p suppressed the levels of M2-type marker genes, like Arg1 and IL10 (Supplementary Fig. 1B). To further validate this inhibitory effect of miR-328-5p deficiency on M2-type macrophage polarization, miR-328-5p mimic was re-expressed in monocytes, and then subjected to qRT-PCR assessment to detect the expression changes of these type-specific markers. In line with the above findings, miR-328-5p mimic re-expression significantly facilitated the macrophage from M1- to M2-like type polarization (Supplementary Fig. 1C, D). In summary, these collective results strongly support that miR-328-5p limits the macrophage polarization toward M1-like type.

miR-328-5p directly targets JUNB to suppress endothelial inflammation

To investigate the precise molecular mechanism by which miR-328-5p regulates inflammatory atherosclerosis, we first analyzed the potential targets of miR-328-5p using the predictor “miRDB”. JUNB, an inflammatory activator, was found to comprise a potential seed region (Fig. 3A). To examine whether this seed region is a specific target site of miR-328-5p, we determined the effect of miR-328-5p on JUNB expression. The qRT-PCR data showed that miR-328-5p inhibition led to a marked increase in both JUNB mRNA and protein expression level (Fig. 3B, C). In line with this, miR-328-5p-mimic overexpression had an obvious inhibitory effect on JUNB expression levels (Fig. 3D, E). To further confirm the direct binding of miR-328-5p with this seed region, we generated a SV40 promoter-driven luciferase reporter carrying this seed region (JUNB-3’UTR-WT) or the seed region-mutated form (JUNB-3’UTR-Mut), then performed luciferase assay. As expected, miR-328-5p mimic overexpression significantly restricted the luciferase activity of JUNB-3’UTR-WT, whereas had no effect on JUNB-3’UTR-Mut activity (Fig. 3F), indicating that JUNB is a direct target of miR-328-5p. Next, we examined the functional involvement of JUNB in the miR-328-5p-mediated suppression of endothelial inflammation. As anticipated, knockdown of JUNB significantly reduced the elevated expression levels of pro-inflammatory factors induced by the absence of miR-328-5p (Fig. 3G-I), as evidenced by qRT-PCR, ELISA, and western blot assays. Conversely, the protein levels of anti-atherosclerotic regulators were notably increased following JUNB depletion (Fig. 3I). Monocyte adhesion assay further validated the critical inhibitory effect of JUNB depletion on endothelial inflammation activated by miR-328-5p deficiency (Fig. 3J). Collectively, these observations suggest that miR-328-5p negatively regulates endothelial inflammation through the post-transcriptional degradation of JUNB.

miR-328-5p negatively regulates atherosclerotic plaque formation

Early endothelial inflammation is a central risk factor for later atherosclerotic plaque development. Hence, we next investigated the role of miR-328-5p in atherogenesis. Intriguingly, silencing miR-328-5p obviously inhibited the levels of the anti-atherosclerotic regulator kruppel-like factor 4 (KLF4), as well as the nitric oxide synthase 3 (NOS3) and the anti-thrombotic regulator thrombomodulin (THBD) (Fig. 4A, B), exhibiting a negative regulatory role of miR-328-5p in the formation of atherogenesis. By contrast, miR-328-5p mimic overexpression dramatically enhanced both the RNA and protein expression levels of these athero-protective regulators (Fig. 4C, D). To further define the regulatory role of miR-328-5p in advanced atherosclerosis, we established the high-fat diet Apoe−/− mice, a widely used atherosclerosis model, which were then transduced with adeno-associated virus (AAV) overexpressing miR-328-5p or its combination with JUNB. Fluorescent microscopy analysis suggested the high infection efficiency of AAV (Fig. 4E). Notably, Oil Red O staining analysis demonstrated that JUNB reintroduction significantly limited the reduced atherosclerotic plaques caused by miR-328-5p overexpression (Fig. 4F). Furthermore, CD68 immunostaining assay further confirmed that the reduced inflammatory infiltration of macrophages triggered by miR-328-5p mimic overexpression was rescued by JUNB reintroduction (Fig. 4G). Taken together, these collective findings lead us to conclude that miR-328-5p acts as a critical repressor in the pathological development of advanced atherosclerosis, at least in part, by regulating JUNB abundance.

DISCUSSION

Previous research has shown that miR-328-5p regulates oxidized low-density lipoprotein-induced endothelial cell injury (23). This study explores the role of miR-328 in inflammatory atherosclerosis and its molecular mechanisms. The findings suggest that miR-328-5p acts as a key negative regulator in atherosclerosis development. Reduced miR-328-5p enhances monocyte adhesion to endothelial cells, while increased miR-328-5p inhibits monocyte migration through endothelial monolayers. These results were confirmed in an Apoe−/− mouse model, showing decreased advanced atherosclerosis with miR-328-5p overexpression. This study establishes miR-328-5p as a critical suppressor of inflammatory atherosclerosis.

Upon exposure to various pro-inflammatory stimuli, endothelial cells can release a plethora of inflammatory factors, including classical adhesion molecules that facilitate macrophage recruitment, as well as cytokines and chemokines that contribute to the inflammatory microenvironment. Disruption of miR-328-5p led to a notable increase in pro-inflammatory factors like VCAM1, IL6, and IL8, underscoring the crucial role of miR-328-5p in regulating inflammatory cascades.

Monocyte-derived macrophages are key drivers of atherosclerosis, able to switch between pro-inflammatory M1 and athero-protective M2 states in response to stimuli (8, 24). M1 macrophages promote inflammation, while M2 macrophages have anti-inflammatory effects, which are crucial in advanced atherosclerosis (8, 25). Macrophages influence the inflammatory microenvironment and atherosclerotic lesion development. Our study identifies miR-328-5p as a pivotal regulator in macrophage polarization. Suppressing miR-328-5p promotes M1 polarization, while overexpressing its mimic enhances M2 formation, underscoring its importance in regulating macrophage polarization.

Numerous crucial factors are involved in the regulation of inflammatory atherosclerosis, including the anti-atherosclerotic regulator KLF4 and NOS3, as well as the anti-thrombotic regulator THBD. These factors are widely recognized as central regulators that negatively regulate the progression of atherogenesis (26-28). To investigate the potential connection between miR-328-5p and atherosclerosis, the impact of miR-328-5p on the expression levels of these atherosclerotic regulators was examined. Interestingly, miR-328-5p triggers a marked increase in the levels of these atherosclerotic repressors, which might partially elucidate the inhibitory effect of miR-328-5p on atherosclerosis. As previously documented, AP-1 tightly associates and collaborates with NF-κB signaling to regulate the inflammatory response (29-31). Activation of NF-κB signaling leads to the transcriptional upregulation of pro-inflammatory factors, such as VCAM1, IL6, and IL8, while simultaneously downregulating the expression of KLF4. The decrease in KLF4, a well-established anti-atherosclerotic regulator, may subsequently inhibit its downstream effectors, THBD and NOS3. Additionally, our RNA-seq data demonstrates the involvement of NF-κB signaling in the miR-328-5p-mediated inflammatory response. It is of significant interest to explore and validate these potential molecular mechanisms that are responsible for the regulation of pro-atherosclerotic events by JUNB.

To elucidate the underlying mechanism by which miR-328-5p mediates inflammatory atherosclerosis, it was found that miR-328-5p directly targets and degrades the RNA level of JUNB, resulting in the downregulation of JUNB protein expression. Reintroducing JUNB effectively reversed the atherosclerotic phenotype induced by miR-328-5p overexpression, underscoring the pivotal role of JUNB in the miR-328-5p-mediated suppression of inflammatory atherosclerosis. This study provides novel insights into the functional importance of the miR-328-5p/JUNB axis in atherosclerosis, and highlights a potential therapeutic target for the clinical treatment of inflammatory atherosclerosis.

FUNDING

This work was supported by grants from the National Natural Science Foundation of China (81900392), Henan Outstanding Youth Science Fund (202300410307), Xinxiang Medical University Doctor Support Foundation (300-505307, XYBSKYZZ201902), Graduate Research Innovation Support Program (YJSCX202206Z).

CONFLICTS OF INTEREST

The authors have no conflicting interests.

DATA AVAILABILITY

Additional data and materials may be requested from the corresponding author on reasonable request.

AUTHOR CONTRIBUTIONS

Z.H., X.Z and H.L. designed and conceived the project. Y.Z. and Y.L. performed most of the experiments. Z.H. and Y.L. performed data analysis. X.Z. and H.L. wrote the manuscript. All authors commented on the manuscript.

FIGURES
Fig. 1. miR-328-5p plays an inhibitory role in endothelial inflammation. (A) The expression of miR-328-5p was detected by qRT-PCR after TNFα treatment (10 ng/mL, 4 h) or high-fat diet induction. (B) Fluorescent imagery showing the effect of miR-328-5p inhibition on THP-1 adhesion to HUVECs. THP-1 cells were pre-treated with CMFDA for 0.5 h before incubation with HUVECs. The HUVECs were identified through anti-CD31 immunostaining, and quantified using ImageJ software. (C) The secretion of VCAM1 was confirmed through both immunofluorescence and western blot assays. (D) The effect of miR-328-5p mimic on monocyte adhesion to endothelial cells. miR-328-5p mimic was first overexpressed in HUVECs, then subjected to TNFα treatment (10 ng/mL, 4 h), before incubation with CMFDA-labeled THP-1 cells. The HUVECs were identified through anti-CD31 immunostaining, and quantified using ImageJ software. (E) Transendothelial migration assay was performed with or without miR-328-5p mimic overexpression. miR-328-5p mimic was overexpressed in HUVECs, which was then treated with TNFα (10 ng/ml, 6 h), and subsequently subjected to incubation with THP-1 cells overnight using a transwell plate. (F) KEGG pathway analysis of the mRNA sequencing data. All values are from biological triplicates, and data shown are the mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 versus PBS or Normal or Ctrl; ##P < 0.01 versus Ctrl + TNFα.
Fig. 2. miR-328-5p regulates the expressions of inflammatory factors. (A) Heatmap of mRNA sequencing in HUVECs with or without miR-328-5p mimic overexpression. (B) Heatmap showing the expression changes of representative pro-inflammatory genes in HUVECs after miR-328-5p enforced overexpression. (C) The mRNA expression levels of inflammatory genes (IL6, IL8, VCAM1, and IL1β) in HUVECs with or without miR-328-5p inhibition were measured by qRT-PCR assay. (D) Immunoblot analysis showing the effect of miR-328-5p inhibitor on changes of the protein expression levels of IL6, VCAM1, and IL1β in HUVECs. (E, F) ELISA analysis showing the effect of miR-328-5p inhibition on IL1β (E) and IL6 (F) production. (G) The effect of miR-328-5p mimic on the expression changes of pro-inflammatory factors (IL6, IL8, VCAM1, and IL1β) in HUVECs was determined by qRT-PCR assay. (H) Immunoblot analysis showing the effect of miR-328-5p mimic on the protein expression levels of IL6, VCAM1, and IL1β in HUVECs with or without miR-328-5p mimic overexpression. All values are from biological triplicates, and data shown are the mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 versus Ctrl.
Fig. 3. miR-328-5p directly targets JUNB for degradation, thereby regulating endothelial inflammation. (A) A predicted seed region of JUNB targeted by miR-328-5p. (B) qRT-PCR analysis showed the effect of miR-328-5p inhibitor on JUNB mRNA levels in HUVECs. (C) Immunoblot analysis showed the effect of miR-328-5p inhibitor on JUNB protein levels in HUVECs. (D) qRT-PCR analysis showed the effect of miR-328-5p mimic on JUNB mRNA levels in HUVECs. (E) Immunoblot analysis showed the effect of miR-328-5p mimics on JUNB protein levels in HUVECs. (F) A luciferase reporter assay showing that miR-328-5p directly targets a JUNB RNA seed region. (G) qRT-PCR assay was performed to test the influence of JUNB knockdown on the increased expression levels of pro-inflammatory factors induced by miR-328-5p inhibition in HUVECs. (H) ELISA analysis showed the effect of JUNB knockdown on the increased IL1β or IL6 production induced by miR-328-5p inhibition. (I) Immunoblot analysis was conducted to investigate the expression changes of pro-inflammatory and anti-atherosclerotic factors following miR-328-5p knockdown, or its combined knockdown with JUNB. (J) Monocyte adhesion assay was performed to examine the effect of JUNB knockdown on the endothelial inflammation activated by miR-328-5p inhibition. HUVEC monolayers were pretreated by miR-328-5p inhibitor or its combination with JUNB siRNA, followed by TNFα induction (10 ng/ml, 4 h), before incubation with CMFDA-labeled THP-1 cells. The HUVECs were identified via anti-CD31 immunostaining, and quantified utilizing ImageJ software. All values are from biological triplicates, and data shown are the mean ± SD. ##P < 0.01 versus miR-328-5p_inh **P < 0.01, ***P < 0.001 versus Ctrl.
Fig. 4. miR-328-5p suppresses advanced atherosclerosis by controlling JUNB stability. (A) Expression changes of anti-atherosclerotic regulators (KLF4, NOS3, and THBD) in HUVECs with or without miR-328-5p inhibitor transfection were measured by qRT-PCR assay. (B) Immunoblot analysis showing the effect of miR-328-5p inhibition on the expression levels of KLF4, NOS3, and THBD in HUVECs. (C) Expression changes of anti-atherosclerotic regulators (KLF4, NOS3, and THBD) in HUVECs with or without miR-328-5p mimic transfection were measured by qRT-PCR assay. (D) Immunoblot analysis showed the effect of miR-328-5p mimic overexpression on the levels of KLF4, NOS3, and THBD in HUVECs. (E) Representative fluorescent imagery of aortic root sections showed the high infection efficiency of AAV-miR-328_mimic. Scale bar, 100 μm. (F) Lesion areas of the thoracoabdominal aortas from the mice infected by the lentivirus expressing miR-328-5p mimic or its combination with Flag-tagged JUNB were examined by Oil-red O staining analysis. (G) Aortic sinus sections from the mice infected by the lentivirus expressing miR-328-5p mimic or its combination with flag-tagged JUNB were subjected to immunohistochemistry staining with an antibody against CD68. (H) A schematic of the molecular mechanism through which miR-328-5p regulates inflammatory atherosclerosis. Scale bar, 500 μm. All values are from biological triplicates, and data shown are the mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001.
REFERENCES
  1. Skålén K, Gustafsson M and Rydberg EK et al (2002) Subendothelial retention of atherogenic lipoproteins in early atherosclerosis. Nature 417, 750-754.
    Pubmed CrossRef
  2. Stachowicz A, Wiśniewska A and Kuś K et al (2019) The Influence of trehalose on atherosclerosis and hepatic steatosis in apolipoprotein E knockout mice. Int J Mol Sci 20, 1552.
    Pubmed KoreaMed CrossRef
  3. Vergallo R and Crea F (2020) Atherosclerotic plaque healing. N Engl J Med 383, 846-857.
    Pubmed CrossRef
  4. Yang ZH, Pryor M and Noguchi A et al (2019) Dietary palmitoleic acid attenuates atherosclerosis progression and hyperlipidemia in low-density lipoprotein receptor-deficient mice. Mol Nutr Food Res 63, e1900120.
    Pubmed KoreaMed CrossRef
  5. Choi BJ, Matsuo Y and Aoki T et al (2014) Coronary endothelial dysfunction is associated with inflammation and vasa vasorum proliferation in patients with early atherosclerosis. Arterioscl Throm Vas 34, 2473-2477.
    Pubmed KoreaMed CrossRef
  6. Feaver RE, Gelfand BD, Wang C, Schwartz MA and Blackman BR (2010) Atheroprone hemodynamics regulate fibronectin deposition to create positive feedback that sustains endothelial inflammation. Circ Res 106, 1703-1711.
    Pubmed KoreaMed CrossRef
  7. Evans TD, Jeong SJ, Zhang X, Sergin I and Razani B (2018) TFEB and trehalose drive the macrophage autophagy-lysosome system to protect against atherosclerosis. Autophagy 14, 724-726.
    Pubmed KoreaMed CrossRef
  8. Peled M and Fisher EA (2014) Dynamic aspects of macrophage polarization during atherosclerosis progression and regression. Front Immunol 5, 579.
    Pubmed KoreaMed CrossRef
  9. Wang Y, Wang GZ, Rabinovitch PS and Tabas I (2014) Macrophage mitochondrial oxidative stress promotes atherosclerosis and nuclear factor-κB-mediated inflammation in macrophages. Circ Res 114, 421-433.
    Pubmed KoreaMed CrossRef
  10. Yurdagul A, Doran AC, Cai B, Fredman G and Tabas IA (2017) Mechanisms and consequences of defective efferocytosis in atherosclerosis. Front Cardiovasc Med 4, 86.
    Pubmed KoreaMed CrossRef
  11. Fontana MF, Baccarella A, Pancholi N, Pufall MA, Herbert DBR and Kim CC (2015) JUNB is a key transcriptional modulator of macrophage activation. J Immunol 194, 177-186.
    Pubmed KoreaMed CrossRef
  12. Xiao C, Wang RH and Lahusen TJ et al (2012) Progression of chronic liver inflammation and fibrosis driven by activation of c-JUN signaling in Sirt6 mutant mice. J Biol Chem 287, 41903-41913.
    Pubmed KoreaMed CrossRef
  13. Hao XZ and Fan HM (2017) Identification of miRNAs as atherosclerosis biomarkers and functional role of miR-126 in atherosclerosis progression through MAPK signalling pathway. Eur Rev Med Pharmacol Sci 21, 2725-2733.
    Pubmed
  14. Hirschberger S, Hinske LC and Kreth S (2018) MiRNAs: dynamic regulators of immune cell functions in inflammation and cancer. Cancer Lett 431, 11-21.
    Pubmed CrossRef
  15. Kim JM, Jung KH and Chu K et al (2015) Atherosclerosis-related circulating MicroRNAs as a predictor of stroke recurrence. Transl Stroke Res 6, 191-197.
    Pubmed CrossRef
  16. Olivieri F, Prattichizzo F and Giuliani A et al (2021) miR-21 and miR-146a: the microRNAs of inflammaging and age-related diseases. Ageing Res Rev 70, 101374.
    Pubmed CrossRef
  17. Quintavalle M, Condorelli G and Elia L (2011) Arterial remodeling and atherosclerosis: miRNAs involvement. Vascul Pharmacol 55, 106-110.
    Pubmed CrossRef
  18. Krol J, Loedige I and Filipowicz W (2010) The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet 11, 597-610.
    Pubmed CrossRef
  19. Lulla AR, Slifker MJ and Zhou Y et al (2017) miR-6883 Family miRNAs target CDK4/6 to induce G(1) phase cell-cycle arrest in colon cancer cells. Cancer Res 77, 6902-6913.
    Pubmed CrossRef
  20. Sherrard R, Luehr S, Holzkamp H, McJunkin K, Memar N and Conradt B (2017) miRNAs cooperate in apoptosis regulation during C. elegans development. Genes Dev 31, 209-222.
    Pubmed KoreaMed CrossRef
  21. Zhao Y, Ponnusamy M, Dong Y, Zhang L, Wang K and Li P (2017) Effects of miRNAs on myocardial apoptosis by modulating mitochondria related proteins. Clin Exp Pharmacol Physiol 44, 431-440.
    Pubmed CrossRef
  22. Bushati N and Cohen SM (2007) microRNA functions. Annu Rev Cell Dev Biol 23, 175-205.
    Pubmed CrossRef
  23. Wu CY, Zhou ZF, Wang B, Ke ZP, Ge ZC and Zhang XJ (2019) MicroRNA-328 ameliorates oxidized low-density lipoprotein-induced endothelial cells injury through targeting HMGB1 in atherosclerosis. J Cell Biochem 120, 1643-1650.
    Pubmed CrossRef
  24. Binesh A, Devaraj SN and Devaraj H (2020) Expression of chemokines in macrophage polarization and downregulation of NFκB in aorta allow macrophage polarization by diosgenin in atherosclerosis. J Biochem Mol Toxic 34, e22422.
    Pubmed CrossRef
  25. Tabas I and Bornfeldt KE (2016) Macrophage phenotype and function in different stages of atherosclerosis. Circ Res 118, 653-667.
    Pubmed KoreaMed CrossRef
  26. Raife TJ, Dwyre DM and Stevens JW et al (2011) Human thrombomodulin knock-in mice reveal differential effects of human thrombomodulin on thrombosis and atherosclerosis. Arterioscler Thromb Vasc Biol 31, 2509-2517.
    Pubmed KoreaMed CrossRef
  27. Shankman LS, Gomez D and Cherepanova OA et al (2015) KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat Med 21, 628-637.
    Pubmed KoreaMed CrossRef
  28. Takaya T, Hirata K and Yamashita T et al (2007) A specific role for eNOS-derived reactive oxygen species in atherosclerosis progression. Arterioscler Thromb Vasc Biol 27, 1632-1637.
    Pubmed CrossRef
  29. Khoyratty TE, Ai Z and Ballesteros I et al (2021) Distinct transcription factor networks control neutrophil-driven inflammation. Nat Immunol 22, 1093-1106.
    Pubmed KoreaMed CrossRef
  30. Mathas S, Hinz M and Anagnostopoulos I et al (2002) Aberrantly expressed c-Jun and JunB are a hallmark of Hodgkin lymphoma cells, stimulate proliferation and synergize with NF-kappa B. EMBO J 21, 4104-4113.
    Pubmed KoreaMed CrossRef
  31. Schmidt D, Textor B and Pein OT et al (2007) Critical role for NF-kappaB-induced JunB in VEGF regulation and tumor angiogenesis. EMBO J 26, 710-719.
    Pubmed KoreaMed CrossRef


This Article


Cited By Articles
  • CrossRef (0)

Author ORCID Information

Funding Information
  • National Natural Science Foundation of China
      10.13039/501100001809
      81900392
  • Henan Outstanding Youth Science Fund
      202300410307
  • Xinxiang Medical University Doctor Support Foundation
      300-505307, XYBSKYZZ201902
  • Graduate Research Innovation Support Program
      YJSCX202206Z

Collections

Services
Social Network Service

e-submission

Archives