1
|
Lau C, Primus CP, Shabbir A, Chhetri I, Ono M, Masucci M, Bin Noorany Aubdool MA, Amarin J, Hamers AJ, Khan Z, Kumar NA, Montalvo Moreira SA, Nuredini G, Osman M, Whitear C, Godec T, Kapil V, Massimo G, Khambata RS, Rathod KS, Ahluwalia A. Accelerating inflammatory resolution in humans to improve endothelial function and vascular health: Targeting the non-canonical pathway for NO. Redox Biol 2025; 82:103592. [PMID: 40209616 PMCID: PMC12005330 DOI: 10.1016/j.redox.2025.103592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Chronic cardiovascular diseases (CVD) are characterised by low-grade systemic inflammation in part due to reduced nitric oxide (NO) bioavailability associated with endothelial dysfunction. Bioavailability of NO can be enhanced by activation of the non-canonical pathway, through increased dietary inorganic nitrate consumption with the potential to attenuate inflammation. METHODS We sought to determine whether dietary inorganic nitrate influences the inflammatory response in models of localised (cantharidin-induced blisters) and systemic inflammation (typhoid vaccine), in healthy male volunteers and conducted two clinical trials; Blister-NITRATE and Typhoid-NITRATE respectively. RESULTS We show that dietary nitrate attenuates endothelial dysfunction following typhoid vaccine administration and accelerates resolution of cantharidin-induced blisters. Both phenomena were associated with an increased level of pro-resolving mediators consequent to a reduction in the expression and activity of pro-inflammatory monocytes. Moreover, we show that leukocytes of the monocyte lineage express the nitrite reductase XOR, that may drive localised nitrite reduction to elevate NO (and cGMP) to drive the protective phenotype. CONCLUSIONS Inorganic nitrate improves endothelial function in the setting of systemic inflammation. Whilst the immediate inflammatory response appeared unaffected by inorganic nitrate treatment, during the resolution phase of the acute inflammatory response lower levels of pro-inflammatory classical inflammatory and intermediate monocytes and attenuated levels of inflammatory cytokines and chemokines were evident. We propose that this reflects a pro-resolution phenotype that may be of potential therapeutic benefit in patients with established CVD. CLINICAL TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov; unique identifiers NCT02715635, NCT03183830.
Collapse
Affiliation(s)
- Clement Lau
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Christopher P Primus
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Asad Shabbir
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ismita Chhetri
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mutsumi Ono
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Masucci
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muhammad Aadil Bin Noorany Aubdool
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Amarin
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexander Jp Hamers
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Zara Khan
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nitin Ajit Kumar
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Gani Nuredini
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Miski Osman
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Charlotte Whitear
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tom Godec
- Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vikas Kapil
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gianmichele Massimo
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Krishnaraj S Rathod
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Cardiology, Barts Heart Centre, 2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Amrita Ahluwalia
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
2
|
Li K, Feng J, Li M, Han L, Wu Y. Systematic Review of Interleukin-35 in Endothelial Dysfunction: A New Target for Therapeutic Intervention. Mediators Inflamm 2025; 2025:2003124. [PMID: 39974277 PMCID: PMC11839265 DOI: 10.1155/mi/2003124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial dysfunction is a significant factor in the pathogenesis of various diseases. In pathological states, endothelial cells (ECs) undergo activation, resulting in dysfunction characterized by the stimulation of inflammatory responses, oxidative stress, cell proliferation, blood coagulation, and vascular adhesions. Interleukin-35 (IL-35), a novel member of the IL-12 family, is primarily secreted by regulatory T cells (Tregs) and regulatory B cells (Bregs). The role of IL-35 in immunomodulation, antioxidative stress, resistance to apoptosis, control of EC activation, adhesion, and angiogenesis in ECs remains incompletely understood, as the specific mechanisms of IL-35 action and its regulation have yet to be fully elucidated. Therefore, this systematic review aims to comprehensively investigate the impact of IL-35 on ECs and their physiological roles in a range of conditions, including cardiovascular diseases, tumors, sepsis, and rheumatoid arthritis (RA), with the objective of elucidating the potential of IL-35 as a therapeutic target for these ailments.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
3
|
Yang O, Teng Y, Zhang R, Qu J. Long-Term Clinical Outcomes of Polymer-Free Sirolimus-Eluting Stent and Polymer-Coated Sirolimus-Eluting Stent in Patients with Type 2 Diabetes. Int J Nanomedicine 2024; 19:11689-11700. [PMID: 39553456 PMCID: PMC11566208 DOI: 10.2147/ijn.s482608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Polymer-free sirolimus-eluting stent (PF-SES) possess multiple properties improving targeted drug elution and in-stent reendothelialization without the presence of polymers. The long-term clinical performance comparison between PF-SES and the latest generation polymer-coated sirolimus-eluting stents (SES), particularly regarding intravascular imaging assessment and in the type 2 diabetes mellitus (DM) population, remains unexplored. Methods We conducted a retrospective study involving 2646 diabetes patients meeting coronary artery disease (CAD) criteria underwent coronary stents in the real-world. All patients were divided into the PF-SES group and the SES group. Optical coherence tomography (OCT) was used to evaluate the imaging characteristics of in-stent reendothelialization. Patient information between the two groups was systematically compared in hospital and at 5-year follow-up. Results In terms of basic characteristics, the proportion of current smoker and stable angina patients in the PF-SES group was significantly higher than that in the SES group. The PF-SES group exhibited significantly higher rate of left anterior descending (LAD) lesion and more stents per patients compared to the SES group. The value of minimum lumen area (MLA), neointimal area (NA) and neointimal thickness (NT) were higher in the PF-SES group. Additionally, the occurrence rates of heterogeneous, lipid layer, intimal tears, thrombi, and micro-vessels were notably lower in the PF-SES group compared to the SES group. A higher all-cause mortality was observed in the SES cohort. Discussion PF-SES could effectively improve in-stent reendothelialization in patients with type 2 DM, with positive effects on survival rate and may, therefore, be considered as an alternative treatment option for improving clinical long-term outcomes.
Collapse
Affiliation(s)
- Ou Yang
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
| | - Yuhuan Teng
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
| | - Ruoxi Zhang
- Department of Cardiology, Harbin Yinghua Hospital, Harbin, Heilongjiang, 150199, People’s Republic of China
| | - Jie Qu
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
| |
Collapse
|
4
|
Li L, Yu Y, Sun X, Wang X, Yang X, Yu Q, Kang K, Wu Y, Yi Q. Pro-endothelialization of nitinol alloy cardiovascular stents enhanced by the programmed assembly of exosomes and endothelial affinity peptide. J Mater Chem B 2024; 12:4184-4196. [PMID: 38592788 DOI: 10.1039/d4tb00363b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Stent implantation is one of the most effective methods for the treatment of atherosclerosis. Nitinol stent is a type of stent with good biocompatibility and relatively mature development; however, it cannot effectively achieve long-term anticoagulation and early endothelialization. In this study, nitinol surfaces with the programmed assembly of heparin, exosomes from endothelial cells, and endothelial affinity peptide (REDV) were fabricated through layer-by-layer assembly technology and click-chemistry, and then exosomes/REDV-modified nitinol interface (ACC-Exo-REDV) was prepared. ACC-Exo-REDV could promote the rapid proliferation and adhesion of endothelial cells and achieve anticoagulant function in the blood. Besides, ACC-Exo-REDV had excellent anti-inflammatory properties and played a positive role in the transformation of macrophage from the pro-inflammatory to anti-inflammatory phenotype. Ex vivo and in vivo experiments demonstrated the effectiveness of ACC-Exo-REDV in preventing thrombosis and hyperplasia formation. Hence, the programmed assembly of exosome interface could contribute to endothelialization and have potential application on the cardiovascular surface modification to prevent stent thrombosis and restenosis.
Collapse
Affiliation(s)
- Linsen Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Yue Yu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Xiaoqing Sun
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Xingyou Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Xiayan Yang
- Chengdu NewMed Biotechnology Co., Ltd, Chengdu 611139, P. R. China
| | - Qifeng Yu
- Chengdu NewMed Biotechnology Co., Ltd, Chengdu 611139, P. R. China
| | - Ke Kang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
- Institute of Regulatory Science for Medical Device, Sichuan University, Chengdu, Sichuan Province, 610065, P. R. China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China.
| |
Collapse
|
5
|
Merinopoulos I, Gunawardena T, Corballis N, Tsampasian V, Eccleshall SC, Smith J, Vassiliou VS. The role of inflammation in percutaneous coronary intervention, from balloon angioplasty to drug eluting stents. Minerva Cardiol Angiol 2023; 71:631-642. [PMID: 35785928 DOI: 10.23736/s2724-5683.22.06091-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
The role of inflammation in percutaneous coronary intervention (PCI) has been investigated in numerous studies. Both pre-PCI and post-PCI inflammatory status have been demonstrated to be linked with patient outcomes. C-reactive protein continues to be the most studied inflammatory biomarker, while a growing number of additional biomarkers, including cytokines and immune cells, are being assessed. As insights are gained into the complexities of the inflammatory response to PCI, it becomes evident that a targeted approach is necessary to ensure optimal patient outcomes. Here, we review the biomarkers that can predict patient outcomes following PCI and specifically how they differ for balloon angioplasty, bare metal stents and drug eluting stents. A specific focus is given to human studies and periprocedural inflammation rather than inflammation associated with myocardial infarction.
Collapse
Affiliation(s)
- Ioannis Merinopoulos
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Tharusha Gunawardena
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Natasha Corballis
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Vassiliki Tsampasian
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Simon C Eccleshall
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK
| | - James Smith
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Vassilios S Vassiliou
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, UK -
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
6
|
Li Y, Yao L, Lu J. IL-35 inhibits adipogenesis via PPARγ-Wnt/β-catenin signaling pathway by targeting Axin2. Int Immunopharmacol 2023; 122:110615. [PMID: 37429144 DOI: 10.1016/j.intimp.2023.110615] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 07/12/2023]
Abstract
Interleukin (IL)-35, a member of the IL-12 family, functions as an immunosuppressive cytokine that plays a crucial role in the regulation of immune-related disorders and inflammatory diseases. Adipose tissue, which is now recognized as an immune organ, is regulated by immunocytes through various signaling pathways, including the peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα) pathway and the Wnt/β-actin pathway. However, there is limited research regarding the effects of IL-35 on adipogenesis. Our current findings indicated that IL-35 impedes the proliferation and promotes the cytotoxicity of 3T3-L1 preadipocytes. Furthermore, IL-35 inhibited the adipogenic differentiation, as well as suppressed triglyceride and lipid accumulation. Additionally, the expression of PPARγ and C/EBPα, two key regulators of adipogenesis, were both down-regulated with IL-35 treatment. In order to explicate the mechanisms underlying the effects of IL-35, we conducted an investigation into the expression of Axin2, an intracellular inhibitor of Wnt/β-catenin signaling, in 3T3-L1 preadipocyte cells. Gene silencing of Axin2 through small interfering RNAs (siRNAs) enhanced PPARγ and C/EBPα expression while decreasing nuclear β-catenin levels in the presence of IL-35. Furthermore, in IL-35-treated cells, Axin2 knockdown boosted adipogenic differentiation (as measured by increased Oil Red O staining). These findings imply that IL-35 regulates Axin2 expression and thereby plays an important role in adipocyte development.
Collapse
Affiliation(s)
- Yuxuan Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, No. 36 San Hao Street, Heping District, Shenyang, 110004, PR China
| | - Lutian Yao
- Department of Orthopedics, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, PR China.
| | - Jing Lu
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, PR China.
| |
Collapse
|
7
|
Wu M, Xun M, Chen Y. Adaptation of Vascular Smooth Muscle Cell to Degradable Metal Stent Implantation. ACS Biomater Sci Eng 2023. [PMID: 37364226 DOI: 10.1021/acsbiomaterials.3c00637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Iron-, magnesium-, or zinc-based metal vessel stents support vessel expansion at the period early after implantation and degrade away after vascular reconstruction, eliminating the side effects due to the long stay of stent implants in the body and the risks of restenosis and neoatherosclerosis. However, emerging evidence has indicated that their degradation alters the vascular microenvironment and induces adaptive responses of surrounding vessel cells, especially vascular smooth muscle cells (VSMCs). VSMCs are highly flexible cells that actively alter their phenotype in response to the stenting, similarly to what they do during all stages of atherosclerosis pathology, which significantly influences stent performance. This Review discusses how biodegradable metal stents modify vascular conditions and how VSMCs respond to various chemical, biological, and physical signals attributable to stent implantation. The focus is placed on the phenotypic adaptation of VSMCs and the clinical complications, which highlight the importance of VSMC transformation in future stent design.
Collapse
Affiliation(s)
- Meichun Wu
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- School of Nursing, University of South China, Hengyang, Hunan 410001, China
| | - Min Xun
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, Hunan 410001, China
| | - Yuping Chen
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, Hunan 410001, China
| |
Collapse
|
8
|
Zhou J, Weng J, Huang X, Sun S, Yang Q, Lin H, Yang J, Guo H, Chi J. Repair effect of the poly (D,L-lactic acid) nanoparticle containing tauroursodeoxycholic acid-eluting stents on endothelial injury after stent implantation. Front Cardiovasc Med 2022; 9:1025558. [PMID: 36426231 PMCID: PMC9678935 DOI: 10.3389/fcvm.2022.1025558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/24/2022] [Indexed: 11/10/2022] Open
Abstract
Background Chronic endoplasmic reticulum stress (ERS) plays a crucial role in cardiovascular diseases. Thus, it can be considered a therapeutic target for these diseases. In this study, poly (D,L-lactic acid) (PDLLA) nanoparticle-eluting stents loaded with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor, was fabricated to assess their ability to reduce endothelial cell apoptosis and promote re-endothelialization after stent implantation. Materials and methods PDLLA nanoparticles loaded with TUDCA were prepared via the emulsification-solvent evaporation method. The cumulative release rates of TUDCA were measured in vitro via high-performance liquid chromatography. The carotid arteries of rabbits were subsequently implanted with stents in vivo. The rabbits were then sacrificed after 4 weeks for scanning electron microscopy. Meanwhile, TUDCA concentration in the homogenate of the peripheral blood and distal vascular tissue after stent implantation was measured. The effect of TUDCA on ERS, apoptosis, and human umbilical vein endothelial cell (HUVEC) function was investigated in vitro by performing cell migration assay, wound healing assay, cell proliferation assays, endoplasmic reticulum (ER)-specific fluorescence staining, immunofluorescence, and western blotting. Results TUDCA nanoparticles were released slowly over 28 days. In addition, TUDCA-eluting stents enhanced re-endothelialization and accelerated the recovery of endotheliocytes in vivo. ERS and apoptosis significantly increased in H2O2-treated HUVECs in vitro. Meanwhile, TUDCA reduced apoptosis and improved function by inhibiting ERS in H2O2-treated HUVECs. Decreased rates of apoptosis and ERS were observed after silencing XBP-1s in H2O2-treated HUVECs. Conclusion TUDCA can inhibit apoptosis and promote re-endothelialization after stent implantation by inhibiting IRE/XBP1s-related ERS. These results indicate the potential therapeutic application of TUDCA as a drug-coated stent.
Collapse
Affiliation(s)
- Jiedong Zhou
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Jingfan Weng
- Zhejiang Hospital Affiliated to Medical College of Zhejiang University, Hangzhou, China
| | - Xingxiao Huang
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Shimin Sun
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Qi Yang
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Jinjin Yang
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Hangyuan Guo
- Shaoxing University School of Medicine, Shaoxing, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
- *Correspondence: Jufang Chi,
| |
Collapse
|
9
|
Chen S, Xia J, Zhang Y, Zhan Q. IL35 attenuated LPS-induced acute lung injury by regulating macrophage polarization. Mol Biol Rep 2022; 49:5811-5820. [PMID: 35748972 PMCID: PMC9244303 DOI: 10.1007/s11033-022-07293-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/27/2022] [Accepted: 02/23/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Interleukin 35 (IL35) has been reported to play a role in acute lung injury (ALI); however, the current results regarding the relationship between IL35 and ALI are inconsistent. Therefore, we aimed to further determine the function of IL35 in ALI in mice and the potential mechanism in this paper. MATERIALS AND METHODS Hematoxylin-eosin (HE) staining and Masson staining were used to evaluate lung injury in mice. Immunohistochemical staining was used to evaluate the expression of IL35 p35, TLR4 and MD2 and the Bax/Bcl2 and p-P65/P65 ratios. The expression levels of IL35 EBi3, CD68, CD206 and MPO were assessed by immunofluorescence staining. RT-PCR was used to examine the expression levels of IL1β and IL6. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining was performed to detect apoptotic cells. RESULTS Overexpression of IL35 alleviated LPS-induced ALI in mice. IL35 overexpression decreased the expression of CD68 and increased the expression of CD206 in mice with ALI. Furthermore, upregulation of IL35 expression obviously reduced the expression of MPO, IL1β and IL6 in the lung tissues of mice with ALI. Mechanistically, IL35 suppressed the TLR4/NFκB-P65 pathway, leading to the promotion of the M1 to M2 macrophage transition and alleviation of inflammation in mice with ALI. CONCLUSIONS IL35 relieved LPS-induced inflammation and ALI in mice by regulating M1/M2 macrophage polarization and inhibiting the activation of the TLR4/NFκB-P65 pathway.
Collapse
Affiliation(s)
- Shengsong Chen
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, No 9, Dongdan Santao, Dongcheng District, 100730, Beijing, P. R. China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
| | - Jingen Xia
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China
| | - Yi Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, No 9, Dongdan Santao, Dongcheng District, 100730, Beijing, P. R. China.
- National Center for Respiratory Medicine, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- National Clinical Research Center for Respiratory Diseases, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
- WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, No 2, East Yinghua Road, Chaoyang District, 100029, Beijing, P. R. China.
| |
Collapse
|
10
|
Feng J, Wu Y. Interleukin-35 ameliorates cardiovascular disease by suppressing inflammatory responses and regulating immune homeostasis. Int Immunopharmacol 2022; 110:108938. [PMID: 35759811 DOI: 10.1016/j.intimp.2022.108938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022]
Abstract
The immune response is of great significance in the initiation and progression of a diversity of cardiovascular diseases involving pro-and anti-inflammatory cytokines. Interleukin-35 (IL-35), a cytokine of the interleukin-12 family, is a novel anti-inflammation and immunosuppressive cytokine, maintaining inflammatory suppression and regulating immune homeostasis. The role of IL-35 in cardiovascular diseases (CVDs) has aroused enthusiastic attention, a diversity of experimental or clinical evidence has indicated that IL-35 potentially has a pivot role in protecting against cardiovascular diseases, especially atherosclerosis and myocarditis. In this review, we initiate an overview of the relationship between Interleukin-35 and cardiovascular diseases, including atherosclerosis, acute coronary syndrome, pulmonary hypertension, abdominal aortic aneurysm, heart failure, myocardial ischemia-reperfusion, aortic dissection and myocarditis. Although the specific molecular mechanisms entailing the protective effects of IL-35 remain an unsolved issue, targeted therapies with IL-35 might provide a promising and effective solution to prevent and cure cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
11
|
Feitosa GS. Interleukin 35 (IL35): A New Biomarker for Coronary Artery Disease? Arq Bras Cardiol 2022; 118:409-410. [PMID: 35262573 PMCID: PMC8856691 DOI: 10.36660/abc.20210991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Gilson Soares Feitosa
- Escola Bahiana de Medicina e Saúde PúblicaSalvadorBABrasilEscola Bahiana de Medicina e Saúde Pública,Salvador, BA – Brasil
- Hospital Santa Izabel da Santa Casa da BahiaSalvadorBABrasilHospital Santa Izabel da Santa Casa da Bahia,Salvador, BA – Brasil
| |
Collapse
|
12
|
Zhang XY, Cui ZW, Zhou YY, Chen DD, Zhang YA. Neutrophil functions can be regulated by IL-35, which is mainly expressed in IL-15Rα + cells in grass carp (Ctenopharyngodon idella). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 121:104103. [PMID: 33857470 DOI: 10.1016/j.dci.2021.104103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 06/12/2023]
Abstract
IL-35 plays a key role in regulatory T (Treg) and regulatory B (Breg) cell functions in mammals. CD25 has been demonstrated as one of the markers of Treg cells, and CD19+CD25hiCD71hi cells have been verified as a type of Breg cells in humans. These results indicate that there is a close relationship between IL-35 and CD25+ cells. In mammals, CD25 (alias IL-2Rα) has been identified as having high affinity and specificity for IL-2 binding, and is closely linked and structurally related to IL-15Rα, which having high affinity for IL-15 binding. In teleost, IL-15Rα can bind to both IL-2 and IL-15, with higher affinity to IL-15 than IL-2, and has been termed a CD25-like molecule in some research studies. To date, no studies of IL-35 and IL-15Rα have been documented in fish. In this work, five isoforms of IL-15Rα were cloned from grass carp, and a monoclonal antibody to the protein was developed. The results of flow cytometry and quantitative real-time PCR analyses demonstrated that grass carp IL-35 subunit genes EBI3a and IL-12p35 were mainly expressed in IL-15Rα+ cells, while the expression levels of IL-10 and TGF-β in IL-15Rα+ and IL-15Rα- cells were insignificant. Recombinant grass carp IL-35 (rgcIL-35) could increase the proportion of IL-15Rα+ cells in leukocytes, and a certain proportion of IL-15Rα+ cells also appeared in myeloid cell subset II after stimulation with rgcIL-35. Meanwhile, the migration, phagocytic ability, and bactericidal ability of grass carp neutrophils were significantly decreased after stimulation with certain concentrations of rgcIL-35. Moreover, neutrophil apoptosis could be significantly inhibited by rgcIL-35.
Collapse
Affiliation(s)
- Xiang-Yang Zhang
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zheng-Wei Cui
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Yuan-Yuan Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Yong-An Zhang
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China; State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China.
| |
Collapse
|
13
|
Chen D, Liang M, Jin C, Sun Y, Xu D, Lin Y. Expression of inflammatory factors and oxidative stress markers in serum of patients with coronary heart disease and correlation with coronary artery calcium score. Exp Ther Med 2020; 20:2127-2133. [PMID: 32765687 PMCID: PMC7401708 DOI: 10.3892/etm.2020.8958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/05/2020] [Indexed: 12/21/2022] Open
Abstract
Expression characteristics of inflammatory factors interleukin-23 and interleukin-35; oxidative stress markers of malondialdehyde, which is a final product of lipid peroxidation; superoxide dismutase; microRNA-126 and microRNA-146a in serum of patients with coronary heart disease were investigated. Correlation between these biomarkers and CACS (calcification score), as well as the underlying clinical significance were evaluated. A total of 192 patients diagnosed with coronary heart disease were recruited as the observation group, and 69 healthy adults who provided their blood samples were selected as the control group. Enzyme linked immunosorbent assay was carried out to measure the levels of inflammatory factors interleukin-23 and interleukin-35, and the levels of oxidative stress markers of malondialdehyde and superoxide dismutase in serum of the patients and healthy subjects. Real-time fluorescence-based quantitative PCR was performed to measure the expression levels of microRNA-126 and microRNA-146a in serum. The differences in expression of these biomarkers were analyzed, and correlation between these biomarkers and coronary artery calcium score were assessed. The differences in expression levels of interleukin-23, interleukin-35, malondialdehyde, superoxide dismutase, microRNA-126 and microRNA-146a were statistically significant in both groups. The expression levels of interleukin-23, interleukin-35, malondialdehyde, superoxide dismutase, microRNA-126 and microRNA-146a in the observation group were closely associated with severity of the disease. There were positive correlations between coronary artery calcium score and interleukin-23, interleukin-35, malondialdehyde, microRNA-126 and microRNA-146a, respectively; while a negative correlation existed between coronary artery calcium score and superoxide dismutase in the observation group. In conclusion, biomarkers interleukin-23, interleukin-35, malondialdehyde, superoxide dismutase, microRNA-126 and microRNA-146a were abnormally expressed in serum of patients with coronary heart disease, implicating their association with onset and progression of the disease. The biomarkers were found to be correlated with coronary artery calcium score. Detection of changes of related biomarkers in serum may have certain value in diagnosis of disease formation, as well as assessment of disease severity.
Collapse
Affiliation(s)
- Datong Chen
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Minghui Liang
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Cheng Jin
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yue Sun
- CT Room, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Dongbin Xu
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yueming Lin
- School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
14
|
IL-35 promotes microglial M2 polarization in a rat model of diabetic neuropathic pain. Arch Biochem Biophys 2020; 685:108330. [DOI: 10.1016/j.abb.2020.108330] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/19/2020] [Accepted: 03/05/2020] [Indexed: 01/31/2023]
|
15
|
Ye J, Wang Y, Wang Z, Liu L, Yang Z, Wang M, Xu Y, Ye D, Zhang J, Lin Y, Ji Q, Wan J. Roles and Mechanisms of Interleukin-12 Family Members in Cardiovascular Diseases: Opportunities and Challenges. Front Pharmacol 2020; 11:129. [PMID: 32194399 PMCID: PMC7064549 DOI: 10.3389/fphar.2020.00129] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/30/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases represent a complex group of clinical syndromes caused by a variety of interacting pathological factors. They include the most extensive disease population and rank first in all-cause mortality worldwide. Accumulating evidence demonstrates that cytokines play critical roles in the presence and development of cardiovascular diseases. Interleukin-12 family members, including IL-12, IL-23, IL-27 and IL-35, are a class of cytokines that regulate a variety of biological effects; they are closely related to the progression of various cardiovascular diseases, including atherosclerosis, hypertension, aortic dissection, cardiac hypertrophy, myocardial infarction, and acute cardiac injury. This paper mainly discusses the role of IL-12 family members in cardiovascular diseases, and the molecular and cellular mechanisms potentially involved in their action in order to identify possible intervention targets for the prevention and clinical treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jing Ye
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen Wang
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Ling Liu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zicong Yang
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Menglong Wang
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yao Xu
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Di Ye
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Jishou Zhang
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yingzhong Lin
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qingwei Ji
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jun Wan
- Hubei Key Laboratory of Cardiology, Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Wang D, Bi K, Yang X, Chen T. Letter by Wang et al Regarding Article, "Interleukin-35 Promotes Macrophage Survival and Improves Wound Healing After Myocardial Infarction in Mice". Circ Res 2019; 125:e5-e6. [PMID: 31219751 DOI: 10.1161/circresaha.119.315309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Daxin Wang
- Clinical Medical College, Yangzhou University, Jiangsu, China
| | - Keying Bi
- Clinical Medical College, Dalian Medical University, Liaoning Province, China
| | - Xinquan Yang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tian Chen
- Clinical Medical College, Dalian Medical University, Liaoning Province, China
| |
Collapse
|