Abstract
Background: The purpose of this study was to probe the specific role of long noncoding RNA taurine upregulation 1 (LncRNA TUG1) in viral myocarditis (VMC).
Methods: The mouse model of VMC was induced by Coxsackievirus type B3 (CVB3). LncRNA TUG1 was subsequently silenced, and micro-140-3p (miR-140-3p) was overexpressed in VMC mice. GenePharma synthesized wild-type and mutant LncRNA TUG1 or CXCL8 (C-X-C Motif Chemokine Ligand 8, Interleukin-8) fragments containing the miR-140-3p binding site and cloned them into the pmirGLO luciferase reporter vector. Dual luciferase reporter assays were performed to test the activity of LncRNA TUG1 or CXCL8 fragments containing miR-140-3p mimic and mimic NC. The effects of silencing LncRNA TUG1 on cell proliferation, apoptosis, and inflammation in the VMC mouse model and in vitro were investigated by flow cytometry, enzyme linked immunosorbent assay, and western blot.
Results: In the VMC mouse model, LncRNA TUG1 and CXCL8 were upregulated, while miR-140-3p was downregulated. Suppressing LncRNA TUG1 led to inhibition of CXCL8 by promoting miR-140-3p. Suppressing LncRNA TUG1 or CXCL8 or restoring miR-140-3p were observed to increase cell viability and decrease apoptosis rate of cardiomyocytes.
Conclusion: LncRNA TUG1 knockdown suppresses inflammation and damage of VMC cardiomyocytes via the miR-140-3p/CXCL8 axis.
Highlights
- LncRNA TUG1 is highly expressed in viral myocarditis (VMC).
- LncRNA TUG1 knockdown improves VMC.
- LncRNA TUG1 negatively mediates miR-140-3p levels.
- LncRNA TUG1 is involved in VMC by regulating miR-140-3p.
Introduction
Myocarditis (MC) is an inflammatory disease that has an incidence of less than 10% of the population.1 Myocarditis is usually manifested by chest pain, palpitations, and breathing difficulties, and some cases may develop into heart failure.2 Myocarditis is caused by a variety of infectious and non-infectious factors, myocardial inflammation, and damage.3,
Long noncoding RNAs (LncRNAs) are important regulatory molecules for gene expression in a variety of developmental processes and diseases. In previous studies, in vitro andin vivo experiments have shown that LncRNAs regulate the progression of CVB3-induced MC. Studies to improve high-throughput screening and LncRNA expression profiling could reveal the presence of aberrant LncRNA expression during cardiac remodeling processes.7,
LncRNAs can act as competitive endogenous RNAs (ceRNAs) and are associated with a variety of biological processes and tumorigenesis by competing with the response elements of microRNAs (miRNAs) for binding to messenger RNAs (mRNAs), thereby regulating the expression levels of mRNAs.14 microRNAs are a class of highly conserved noncoding RNAs that regulate gene expression by binding to targeted mRNAs or complementary sequences within the 3'-UTR of targeted binding mRNAs.15 Previous studies have shown that miR-140-3p may be associated with osteoarthritis, liver fibrosis, and tumorigenesis.16,
C-X-C motif chemokine ligand 8 (CXCL8) is a predicted target protein of miR-140-3p. C-X-C motif chemokine ligand 8 is a multicellular chemokine that recruits neutrophils and other immune cells to the inflammatory region.21 Abnormal regulation of CXCL8 has been implicated in inflammation-mediated diseases, including inflammatory bowel disease, rheumatoid arthritis, psoriasis, asthma, and cystic fibrosis.22
This study aimed to investigate the effect of LncRNA TUG1 on VMC by regulating the miR-140-3p/CXCL8 axis.
Methods
Animal Treatment
Thirty-two male BALB/c mice (SPF grade, 4-6 weeks old, 18-22 g) were provided by Shanghai Lab. Animal Research Center (Shanghai, China). Coxsackievirus B3 (CVB3) (Nancy strain) was provided by the State Key Laboratory of Virology (Wuhan, China). The mice were placed in a well-ventilated stainless-steel lattice-roofed pathogen-free cage at room temperature on a light/dark cycle for 12 hours.
Lentiviral vectors were provided by GeneChem (Shanghai, China). All mice were randomly divided into 4 groups: Blank (untreated normal mice), VMC, VMC + sh-TUG1, and VMC + sh-NC, with 8 mice in each group.
Coxsackievirus B3 was subpassaged by Hela cells (ATCC number: CCL-2). Coxsackievirus B3 infection (103 PFU) was given by intraperitoneal injection, while 0.1 mL of normal saline was considered a control. The day of vaccinating CVB3 is defined as day 0. On days 1 and 3, mice were intraperitoneally injected with lentivirus at a dose of 5 × 104 gtu/mouse. On day 7, mice were injected intraperitoneally with chloral hydrate (10%, 4 mg/g) for anesthesia, fixed in the supine position, and euthanized by neck breaking. The heart tissues were collected, rapidly frozen, and kept at −80°C.
Cell Culture
Cardiomyocyte HL-1 cells (ATCC number: CRL-1446; ATCC, USA) were cultured in Dulbecco’s modified eagle medium (DMEM) (ATCC) containing 10% FBS (Gibco, USA) and 1% penicillin/streptomycin (Beyotime, Shanghai, China) at 37°C and 5% CO2. Cells were passed every 48 hours. The HL-1 cells were divided into 2 groups: the control group and the CBV3 group. The CBV3 group was CBV-infected HL-1 cells. Cardiomyocytes were infected with 10 MOI of CVB3 diluted with DMEM. After that, cells were further cultured for 48 hours. Control cells were treated in the same way without CVB3.
Cell Transfection
CXCL8 was overexpressed using pcDNA3.1 (Invitrogen, Thermo Fisher Scientific) to form CXCL8 overexpression plasmid (pcDNA3.1-CXCL8) and its negative control (pcDNA3.1). GenePharma (Shanghai, China) synthesized small interfering RNA (si-TUG1) and its negative control (si-NC), miR-140-3p mimic, miR-140-3p inhibitor, and negative controls (mimic NC; inhibitor NC). To achieve transfection, the above oligonucleotides and plasmids were transfected into HL-1 cells immediately using Lipofectamine 3000 (Invitrogen), according to Lipofectamine 3000 (Invitrogen), at 37°C, and the medium was changed 6 hours later. After 48 hours, cardiomyocytes were assayed for quantitative reverse transcription polymerase chain reaction (RT-qPCR) or western blot to verify the transfection efficiency.
Quantitative Reverse Transcription Polymerase Chain Reaction
Total RNA from mouse heart tissue and HL-1 cells was extracted by TRIzol™ Reagent (Sigma, USA) and then detected by microspectrophotometer (Thermo, USA). cDNA synthesis of miRNA was performed by miRNA reverse transcription kit (Takara, Japan), and that of mRNA and lncRNA by PrimeScript™ RT Reagent kit (Takara). The synthesized cDNA was mixed with SYBR ®Premix Ex Taq™ (Takara) and RT-qPCR was implemented using ABI Prism 7300 system (Applied Biosystems, USA). Polymerase chain reaction (PCR) was amplified by predenaturation 95°C for 10 minutes and 40 cycles of 90°C (10 seconds), 60°C (20 seconds), and 72°C (30 seconds). Polymerase chain reaction primers were synthesized by Beijing Genomics Institute (Beijing, China) and the 2−ΔΔCT method calculated relative gene expression.
Hematoxylin and Eosin Staining
Formalin-fixed heart tissue was paraffin-embedded and sliced (5 μm). The slices were dewaxed and rehydrated, then stained with hematoxylin solution, and re-stained in eosin solution. Imaging was performed using a microscope (DS-Ri2, Nikon, Japan).
TUNEL Staining
Heart tissue sections were dewaxed and stained with terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) kit (Roche, CA, USA). Heart tissue sections were reacted with a TUNEL reaction mixture with terminal deoxynucleotide transferase at 37°C for 1 hour and stained with DAPI (Beyotime). Positive apoptotic cells were marked in green and all nuclei were marked in blue. The results were visualized in the DAB system (Solarbio, Beijing, China) and viewed under a microscope (DS-Ri2, Nikon).
Cell Proliferation Assay
The cell viability for HL-1 cell proliferation was measured by CCK-8 assay (Beyotime). Simply put, treated HL-1 cells were plated into 96-well plates for 24 hours, and CCK-8 solution was incubated at 10 μL/well at 37°C for 1 hour. Optical density values (450 nm) were recorded using a microplate reader.
Flow Cytometry
FITC Annexin V Apoptosis Detection Kit (BD Biosciences, USA) detected HL-1 cell apoptosis. Treated HL-1 cells in the culture medium were obtained by centrifugation, rinsed with pre-cooled PBS (Beyotime), and resuspended in 500 μL 1× binding buffer, and incubated with Annexin V-FITC and PI away from light. BD FACSCanto II flow cytometry (BD Biosciences) was adopted for detection. There were 3 replicates for each group, and the experiments were repeated 3 times.
Enzyme Linked Immunosorbent Assay
HL-1 cell culture supernatant and mouse heart homogenate were collected. Interleukin-6 (IL-6), interleukin 1-beta (IL-β), and interferon‐gamma (IFN-γ) were determined using enzyme linked immunosorbent assay (ELISA) kits (R&D Systems, USA). There were 3 replicates for each group, and the experiments were repeated 3 times.
Oxidative Stress Detection
DCFH-DA (C2938, Thermo Scientific), Lipid Peroxidation MDA Assay Kit (Beyotime), and Total Superoxide Dismutase Assay Kit with WST-8 (Beyotime) were employed to quantitatively determine reactive oxygen species (ROS), Malondialdehyde (MDA), and Superoxide Dismutase (SOD) in HL-1 cells and mouse heart tissue, respectively.
Western Blot Analysis
Mouse heart tissue and HL-1 cells were lysed with RIPA lysis buffer (Solarbio) and the obtained protein was quantified by a BCA kit (Thermo Fisher Scientific). Twelve percentage of SDS-PAGE separation gel and concentrate gel were prepared (Beyotime), and the protein concentration was adjusted to 30 μg with sample buffer (Beyotime). The protein was electroblotted on an electrophoretic apparatus (Bio-Rad, USA) at 80 V for 40 minutes and 120 V for 2 hours, covered with PVDF membrane (Beyotime) at 200 mA for 2 hours, and blocked with TBST (Beyotime) containing 5% skim milk powder for 1 hour. Next, primary antibodies were incubated at 4°C overnight, including p65 (ab32536, Abcam), p-p65 (3031, Cell Signaling Technology), CXCL8 (ab7747, Abcam), Bax (2272, Cell Signaling Technology), Cleaved Caspase-3 (9661, Cell Signaling Technology), Bcl-2 (ab196495, Abcam), and GADPH (5174, Cell Signaling Technology). The secondary antibody was incubated for 2 hours and developed with ECL solution (Beyotime) in a chemiluminescence imager (Image Quant LAS4000 mini, GE Healthcare). Image J software was utilized for gray analysis.
Luciferase Reporter Gene Assay
starBase 3.0 (
RNA Pull-Down
RNA pull-down assay was carried out with an RNA pull-down kit (Pierce, Thermo Fisher Scientific). WT-TUG1 and MUT-TUG1 were labeled and purified using an RNA 3 terminal dethiobiotinization kit (Pierce). After PCR amplification and digestion of the DNA fragments of WT-TUG1 or MUT-TUG1, biotin RNA was used to label WT-TUG1 and MUT-TUG1, which were named Bio-TUG1 WT and Bio-TUG1 MUT, respectively, and Bio-NC was used as a control. Biotinized (Bio)-TUG1 WT and Bio-TUG1 MUT were coupled to streptavidin beads. After lysis, RNA complexes were obtained and purified with TRIzol reagent (Sigma-Aldrich) to determine miR-140-3p by RT-qPCR.
Data Analysis
All data were analyzed using Granpad Prism 8 and expressed as mean ± standard deviation (SD). All experiments were biologically replicated at least 3 times. Two-group datasets were analyzed by Student’s
Statement
No artificial intelligence (AI)—assisted technologies were used in the production of submitted work.
Results
LncRNA TUG1 is Highly Expressed in Viral Myocarditis
After CVB3 injection, pathological changes in heart tissue were observed by hematoxylin and eosin (HE) staining. The heart tissue of VMC mice showed obvious symptoms of injury and inflammation (
LncRNA TUG1 Knockdown Improves Viral Myocarditis
To determine the biological function of LncRNA TUG1 in VMC mice and cardiomyocytes, LncRNA TUG1 shRNA and siRNA were utilized to knock down LncRNA TUG1 in VMC mice and cardiomyocytes, respectively. Quantitative reverse transcription polymerase chain reaction showed that LncRNA TUG1 expression in VMC mice and VMC cells was reduced after LncRNA TUG1 knockdown (
In VMC cells, VMC cell proliferation more than doubled, and apoptosis was alleviated, decreasing by 19.3% after LncRNA TUG1 knockdown (
LncRNA TUG1 Negatively Mediates miR-140-3p Levels
StarBase predicted miR-140-3p as the target of LncRNA TUG1 (
LncRNA TUG1 is Involved in Viral Myocarditis by Regulating miR-140-3p
To explore LncRNA TUG1 and miR-140-3p interaction in VMC, si-TUG1 and miR-140-3p inhibitor were co-transfected into VMC cells. It was observed that si-TUG1 downregulated LncRNA TUG1 and upregulated miR-140-3p (
miR-140-3p Targets CXCL8
As predicted by starBase, CXCL8 was a target of miR-140-3p (
LncRNA TUG1 Influences Viral Myocarditis by Controlling miR-140-3p/CXCL8 Axis
si-TUG1 and pcDNA 3.1-CXCL8 were co-transfected into HL-1 cells, and RT-qPCR results showed that the inhibition effect of si-TUG1 on CXCL8 was reversed by pcDNA 3.1-CXCL8 (
Discussion
Among many viruses that cause VMC, CVB3 is a common strain of VMC, and the pathogenesis of CVB3-induced VMC model consists of direct myocardial injury, in which the virus directly impedes the metabolism of the host cells and leads to cell death, and indirect myocardial injury, in which a large number of inflammatory factors mediated by the immune system contribute to myocardial injury.23 Therefore, this model has been widely applied to study acute infection and the chronic immune phase of human VMC. However, despite extensive research, the pathogenesis of CVB3-induced MC remains unclear, with limited research and treatment options.
Noncoding RNAs play an important role in the viral invasion of the host and in the immune response induced by infection. LncRNAs are universally involved in the regulation of various physiopathological processes such as cancer, tumorigenesis, immunity, inflammation, cell differentiation, and proliferation. Abnormally expressed LncRNAs were identified in the current study of the VMC model.10 Given that some LncRNAs in the blood are highly stable and detectable and also have great potential in diagnosing MC. LncRNA TUG1 is a regulator present in a wide range of physiological or pathological processes, is ubiquitously expressed
At the transcriptional level, LncRNAs can further regulate the expression of mRNAs by competitively binding to miRNAs, thereby affecting target proteins. This mechanism is currently the most widely studied in LncRNAs in various diseases. miRNAs have been identified to play important roles in the regulation of various biological processes, including cell proliferation, apoptosis, and immune responses, and are involved in the regulation of vascular proliferation, cardiogenesis, heart failure, and cardiac hypertrophy.28 LncRNA TUG1 and miR-140-3p interaction was confirmed, suggesting that LncRNA TUG1 negatively mediates miR-140-3p as a ceRNA. In addition, silencing miR-140-3p stimulated myocardial inflammation and injury in MC, but reducing miR-140-3p mitigated the decreased myocardial inflammation and injury in VMC cells induced by silencing LncRNA TUG1, which was characterized by an inflammatory response, oxidative stress, and apoptosis, and impaired proliferation. This is largely consistent with other previous miRNA regulatory mechanisms.29 In virus-induced apoptosis, inhibition of miRNA expression largely reduces changes in the levels of apoptosis-associated proteins such as miR-34a and, in this study, miR-140-3p. In contrast, miRNAs such as miR-222 and miR-21, whose expression is upregulated in the CBV3-induced VMC model, act as anti-apoptotic factors.10 This suggests that there is aberrant expression of miRNAs in VMC and that the extent of VMC could be reduced in the future by targeting these miRNAs.
To date, 17 C-X-C chemokines have been identified in humans, most of which are associated with cardiovascular disease. Among them, CXCL8 is mainly found in macrophages, and it has been shown that CXCL8 promotes the development of atherosclerosis through macrophage recruitment.30,
The present study has some limitations. High expression of LncRNA TUG1 in cardiomyocytes is a damaging factor, and it is closely related to the development of inflammatory response and immune response. However, our study has some shortcomings at present. For the LncRNA TUG1/miR-140-3p/CXCL8 axis to modulate CVB3 infection-induced VMC, this effect does not necessarily translate into meaningful clinical benefits. Moreover, the complexity of the LncRNA/miRNA/mRNAs regulatory network necessitates further investigation of the role of LncRNA TUG1 in clinical practice and detailed physiological mechanisms.
Overall, our results show that high expression of LncRNA TUG1 is observed in VMC mice and cells, and LncRNA TUG1 can regulate CXCL8 by binding to miR-140-3p, and silencing LncRNA TUG1 downregulates CXCL8 by binding to miR-140-3p. This regulation reduces the inflammation and damage of cardiomyocytes, which provides a new idea for developing targeted therapy for VMC.
Supplementary Materials
Footnotes
References
- Basso C. Myocarditis. N Engl J Med. 2022;387(16):1488-1500. https://doi.org/10.1056/NEJMra2114478
- Olejniczak M, Schwartz M, Webber E, Shaffer A, Perry TE. Viral myocarditis-incidence, diagnosis and management. J Cardiothorac Vasc Anesth. 2020;34(6):1591-1601. https://doi.org/10.1053/j.jvca.2019.12.052
- Reyes MP, Lerner AM. Coxsackievirus myocarditis--with special reference to acute and chronic effects. Prog Cardiovasc Dis. 1985;27(6):373-394. https://doi.org/10.1016/0033-0620(85)90001-5
- Fairweather D, Rose NR. Coxsackievirus-induced myocarditis in mice: a model of autoimmune disease for studying immunotoxicity. Methods. 2007;41(1):118-122. https://doi.org/10.1016/j.ymeth.2006.07.009
- Dominguez F, Kühl U, Pieske B, Garcia-Pavia P, Tschöpe C. Update on myocarditis and inflammatory cardiomyopathy: reemergence of endomyocardial biopsy. Rev Esp Cardiol (Engl Ed). 2016;69(2):178-187. https://doi.org/10.1016/j.rec.2015.10.015
- Leonard EG. Viral myocarditis. Pediatr Infect Dis J. 2004;23(7):665-666. https://doi.org/10.1097/01.inf.0000132280.36984.a9
- Wang Z, Zhang XJ, Ji YX. The long noncoding RNA Chaer defines an epigenetic checkpoint in cardiac hypertrophy. Nat Med. 2016;22(10):1131-1139. https://doi.org/10.1038/nm.4179
- Viereck J, Kumarswamy R, Foinquinos A. Long noncoding RNA Chast promotes cardiac remodeling. Sci Transl Med. 2016;8(326):326ra22-. https://doi.org/10.1126/scitranslmed.aaf1475
- Chen L, Zhou Y, Li H. LncRNA, miRNA and lncRNA-miRNA interaction in viral infection. Virus Res. 2018;257():25-32. https://doi.org/10.1016/j.virusres.2018.08.018
- Zhang C, Xiong Y, Zeng L. The role of non-coding RNAs in viral myocarditis. Front Cell Infect Microbiol. 2020;10():312-. https://doi.org/10.3389/fcimb.2020.00312
- Liu Q, Kong Y, Han B, Jiang D, Jia H, Zhang L. Long non-coding RNA expression profile and functional analysis in children with acute fulminant myocarditis. Front Pediatr. 2019;7():283-. https://doi.org/10.3389/fped.2019.00283
- Han Y, Liu Y, Gui Y, Cai Z. Long intergenic non-coding RNA TUG1 is overexpressed in urothelial carcinoma of the bladder. J Surg Oncol. 2013;107(5):555-559. https://doi.org/10.1002/jso.23264
- Xu Y, Wang J, Qiu M. Upregulation of the long noncoding RNA TUG1 promotes proliferation and migration of esophageal squamous cell carcinoma. Tumour Biol. 2015;36(3):1643-1651. https://doi.org/10.1007/s13277-014-2763-6
- Khashkhashi Moghadam S, Bakhshinejad B, Khalafizadeh A, Mahmud Hussen B, Babashah S. Non‐coding RNA‐associated competitive endogenous RNA regulatory networks: novel diagnostic and therapeutic opportunities for hepatocellular carcinoma. J Cell Mol Med. 2022;26(2):287-305. https://doi.org/10.1111/jcmm.17126
- Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281-297. https://doi.org/10.1016/s0092-8674(04)00045-5
- Ntoumou E, Tzetis M, Braoudaki M. Serum microRNA array analysis identifies miR-140-3p, miR-33b-3p and miR-671-3p as potential osteoarthritis biomarkers involved in metabolic processes. Clin Epigenet. 2017;9():127-. https://doi.org/10.1186/s13148-017-0428-1
- Wu SM, Li TH, Yun H, Ai HW, Zhang KH. miR-140-3p knockdown suppresses cell proliferation and fibrogenesis in hepatic stellate cells via PTEN-mediated AKT/mTOR signaling. Yonsei Med J. 2019;60(6):561-569. https://doi.org/10.3349/ymj.2019.60.6.561
- Karakas M, Schulte C, Appelbaum S. Circulating microRNAs strongly predict cardiovascular death in patients with coronary artery disease-results from the large AtheroGene study. Eur Heart J. 2017;38(7):516-523. https://doi.org/10.1093/eurheartj/ehw250
- Vavassori C, Cipriani E, Colombo GI. Circulating microRNAs as novel biomarkers in risk assessment and prognosis of coronary artery disease. Eur Cardiol. 2022;17():e06-. https://doi.org/10.15420/ecr.2021.47
- Yang D, Wang M, Hu Z. Extracorporeal cardiac shock wave-induced exosome derived from endothelial colony-forming cells carrying miR-140-3p alleviate cardiomyocyte hypoxia/reoxygenation injury via the PTEN/PI3K/AKT pathway. Front Cell Dev Biol. 2021;9():779936-. https://doi.org/10.3389/fcell.2021.779936
- Sokol CL, Luster AD. The chemokine system in innate immunity. Cold Spring Harb Perspect Biol. 2015;7(5):a016303-. https://doi.org/10.1101/cshperspect.a016303
- Cambier S, Gouwy M, Proost P. The chemokines CXCL8 and CXCL12: molecular and functional properties, role in disease and efforts towards pharmacological intervention. Cell Mol Immunol. 2023;20(3):217-251. https://doi.org/10.1038/s41423-023-00974-6
- Yip F, Lai B, Yang D. Role of coxsackievirus B3-induced immune responses in the transition from myocarditis to dilated cardiomyopathy and heart failure. Int J Mol Sci. 2023;24(9):7717-. https://doi.org/10.3390/ijms24097717
- Tam C, Wong JH, Tsui SKW, Zuo T, Chan TF, Ng TB. LncRNAs with miRNAs in regulation of gastric, liver, and colorectal cancers: updates in recent years. Appl Microbiol Biotechnol. 2019;103(12):4649-4677. https://doi.org/10.1007/s00253-019-09837-5
- Haybar H, Sadati NS, Purrahman D, Mahmoudian-Sani MR, Saki N. lncRNA TUG1 as potential novel biomarker for prognosis of cardiovascular diseases. Epigenomics. 2023;15(23):1273-1290. https://doi.org/10.2217/epi-2023-0242
- Gou W, Zhang Z, Yang C, Li Y. MiR-223/Pknox1 axis protects mice from CVB3-induced viral myocarditis by modulating macrophage polarization. Exp Cell Res. 2018;366(1):41-48. https://doi.org/10.1016/j.yexcr.2018.03.004
- Bao J, Sun T, Yue Y, Xiong S. Macrophage NLRP3 inflammasome activated by CVB3 capsid proteins contributes to the development of viral myocarditis. Mol Immunol. 2019;114():41-48. https://doi.org/10.1016/j.molimm.2019.07.012
- Lu P, Ding F, Xiang YK, Hao L, Zhao M. Noncoding RNAs in cardiac hypertrophy and heart failure. Cells. 2022;11(5):777-. https://doi.org/10.3390/cells11050777
- Marketou M, Kontaraki J, Patrianakos A. Peripheral blood microRNAs as potential biomarkers of myocardial damage in acute viral myocarditis. Genes. 2021;12(3):420-. https://doi.org/10.3390/genes12030420
- Domschke G, Gleissner CA. CXCL4-induced macrophages in human atherosclerosis. Cytokine. 2019;122():154141-. https://doi.org/10.1016/j.cyto.2017.08.021
- Apostolakis S, Spandidos D. Chemokines and atherosclerosis: focus on the CX3CL1/CX3CR1 pathway. Acta Pharmacol Sin. 2013;34(10):1251-1256. https://doi.org/10.1038/aps.2013.92
- Lu X, Wang Z, Ye D. The role of CXC chemokines in cardiovascular diseases. Front Pharmacol. 2021;12():765768-. https://doi.org/10.3389/fphar.2021.765768
- Wang J. Neutrophils in tissue injury and repair. Cell Tissue Res. 2018;371(3):531-539. https://doi.org/10.1007/s00441-017-2785-7