1
|
Wang Z, Han L, Dong M, Liu Y, Hu X, Huang L, Zhang C, Guo L, Liu S, Liao L. IL-35 Ameliorates Myocardial Strain in Mice with T2DM-Induced Cardiac Injury: Assessment by Layer-Specific Strain. Diabetes Metab Syndr Obes 2025; 18:1551-1562. [PMID: 40375942 PMCID: PMC12079041 DOI: 10.2147/dmso.s510594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
Purpose The established association between endothelial dysfunction and the pathogenesis of cardiovascular disease in diabetic individuals has been well-documented. Interleukin-35 (IL-35) can suppress inflammatory processes and ameliorate endothelial dysfunction. This study aimed to evaluate the effect of IL-35 treatment on diabetic mice with diabetes-induced cardiac injury using layer-specific strain analysis. Patients and Methods Twenty-six mice were allocated into three groups: the control group (CON, n=10), the diabetic group (DM, n=10), and the diabetic group treated with IL-35 (DMIL, n=6). The DM and DMIL groups were subjected to a high-fat diet and streptozotocin to induce diabetes, with the DMIL group receiving an additional 6 weeks of IL-35 treatment. Measurements of body weight, blood glucose levels, routine echocardiographic parameters, and layer-specific strain were conducted at baseline, post-diabetes induction, and post-treatment. Morphological changes in cardiomyocytes were examined in pathological heart sections, and cardiac inflammation was detected by protein immunoblotting. Results After inducing diabetes, diabetic mice exhibited notable systolic and diastolic dysfunction. IL-35 treatment significantly reduced myocardial inflammatory infiltration and improved myocardial fibrosis in the DMIL group in comparison to the DM group. Only diastolic function E/e' showed a significant improvement when comparing conventional echocardiograms between the DMIL and DM groups. In the context of layered strain analysis, the DMIL group exhibited a notable enhancement in middle and epicardial global longitudinal strain and global radial strain when compared to the DM group. Conclusion IL-35 can enhance myocardial function in diabetic mice. Layer-specific strain could serve as a valuable tool for evaluating interventions in diabetes.
Collapse
Affiliation(s)
- Ziying Wang
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Mingyi Dong
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yunman Liu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Xiangsui Hu
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Long Huang
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Chunquan Zhang
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Liangyun Guo
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Shengbo Liu
- GE Healthcare Ultrasound Application Specialist, Nanchang, People’s Republic of China
| | - Lingmin Liao
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
2
|
He Z, Dang J, Cui X, Li B, Bao S, Fan J. The distinct role of IL-34 and IL-35 in gastric cancer. Front Immunol 2025; 16:1559508. [PMID: 40416985 PMCID: PMC12098620 DOI: 10.3389/fimmu.2025.1559508] [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: 01/12/2025] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Gastric cancer (GC) remains a major challenge due to its high mortality and morbidity, despite extensive research. Dysregulated host immunity plays a critical role in carcinogenesis, particularly among susceptible cohorts. In the gastric mucosa of GC patients, a reduction in IL-34 and TAM1, accompanied by an increase in TAM2 via M-CSF, enhances Th2 cell function, reduces pro-inflammatory activity, and elevates anti-inflammatory responses. Consequently, TAM2 acts in both paracrine and autocrine manners to polarize and boost TAM2, creating a tumour-favourable microenvironment that supports GC progression. High levels of TAM2, observed during advanced GC stages, suppress gastric IL-34 production, further promoting GC development. In contrast, IL-35, a cytokine involved in immune regulation and suppression, is produced by activated T cells and/or B cells in the affected gastric mucosa. Persistent H. pylori infection in GC tissues is associated with significant infiltration of IL-35-producing B cells and regulatory T cells (Tregs), which enhance the immunosuppressive and pro-tumour microenvironment by disrupting the local immune balance. Upregulated mucosal IL-35 promotes the polarization of TAM2 and Tregs while suppressing TAM1 cells, fostering a tumour-friendly environment that allows transformed gastric mucosal cells to evade immune surveillance, particularly in chronic H. pylori-infected patients. This cascade enhances proliferation and invasion while suppressing differentiation and apoptosis of GC cells. Together, the differential regulation of these cytokines creates an environment that supports cancer progression and resistance to therapy. Targeting the IL-34 and IL-35 pathways may offer a novel therapeutic strategy for improving outcomes in GC patients.
Collapse
Affiliation(s)
- Zhiyun He
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jie Dang
- Central Sterile Supply Department, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiang Cui
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Bo Li
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Shisan Bao
- Scientific Research Division, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, China
- Scientific Research Division, The First People’s Hospital of Baiyin, Baiyin, Gansu, China
| | - Jingchun Fan
- School of Public Health, Centre for Evidence-based Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Wang P, Li Y, Zhao L, Liu B, Cai Z, Zhang P, Li P, Gao X, Zhan Y. High interleukin-35 expression is associated with the severity of rheumatic mitral stenosis. Front Immunol 2025; 16:1537497. [PMID: 40264784 PMCID: PMC12011801 DOI: 10.3389/fimmu.2025.1537497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Background Rheumatic mitral stenosis (RMS) is the most common manifestation of rheumatic heart disease, with high morbidity and mortality. Interleukin-35 (IL-35) is a novel anti-inflammatory cytokine associated with many autoimmune diseases. However, the relation between IL-35 expression and RMS remains unknown. We aimed to study IL-35 expression in RMS and its association with disease progression. Methods IL-35 concentration was analyzed in blood samples from 40 patients, including 20 moderate, 20 severe RMS, and 20 healthy controls by ELISA. Mitral valve (MV) IL-35 expression was determined by western blot and immunohistochemistry in patients with RMS (22 and 29 cases, respectively) in comparison to control specimens with mitral valve prolapsed (5 cases, respectively). Results IL-35 levels were significantly elevated in the blood of the RMS patients compared to those from healthy subjects(p<0.05) and positively correlated with the severity of RMS (r=0.317, p<0.05). The expression of IL-35 and its subunits (p35 and EBI3) was also detected in MV tissues of patients with moderate or severe RMS. The expression of IL-35 and its subunits (p35 and EBI3) had a positive association with the severity of RMS in MV tissues (r=0.528, p<0.01; r=0.561, p<0.001; r=0.456, p<0.01). Co-localization of p35 and EBI3 was seen in MV tissues of RMS patients in a predominantly perivascular pattern. Conclusion We show for the first time an increase of IL-35 level in the blood and MV tissues of RMS patients, which is strongly correlated with the severity of RMS. These results suggest that IL-35 plays an important regulatory role in the progression of RMS.
Collapse
Affiliation(s)
- Ping Wang
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yaxiong Li
- Department of Cardiovascular Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Li Zhao
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Bin Liu
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Zhibin Cai
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Peng Zhang
- Key Laboratory of Cardiovascular Research of Yunnan Province, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Peng Li
- Department of Cardiovascular Surgery, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Xuezhen Gao
- Health Examination Center, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yong Zhan
- Division of Cardiac Surgery, Cardiovascular Center, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
4
|
Jie F, Dong F, Xu L, Deng S, Wang Q, Wu Q. Cytokine Expression and Cytolytic Effect of Natural Killer Cells are Suppressed in Septic Shock. Scand J Immunol 2025; 101:e70023. [PMID: 40254928 DOI: 10.1111/sji.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 02/21/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
Septic shock is the most severe stage of sepsis. How immune dysregulation contributes to the pathogenesis of septic shock has not been thoroughly understood. In the current research, the phenotype and function of circulating natural killer (NK) cells of septic patients were characterised. The absolute number of NK cells was comparably reduced in septic shock survivors and non-survivors, probably owing to elevated NK cell apoptosis. Activating receptors including signalling lymphocytic activation molecule 4 (SLAMF4), natural killer cell p30-related protein (NKp30), natural killer group 2, member D (NKG2D), and DNAX accessory molecule 1 (DNAM-1) were significantly downregulated on NK cell surface in septic shock patients, especially non-survivors. Furthermore, the patients' NK cells exhibited lower expression of granzyme B and perforin, weaker target cell-induced degranulation and cytokine expression, as well as incompetent cytolytic effect. These alterations were more profound in septic shock non-survivors. Importantly, serum interleukin-35 (IL-35), which is an immunosuppressive cytokine, was remarkably elevated in septic shock patients. Besides, serum interleukin-35 concentration was positively correlated with disease scores but negatively correlated with NK cell activating receptor expression. In vitro assays indicated IL-35-induced strong suppression of NK cell activity, as evidenced by concomitant downregulation of cytokines and activating receptors along with inhibition of cytolytic capacity. Therefore, we uncovered for the first time the contributing role of IL-35 in septic shock-related human NK cell dysfunction.
Collapse
Affiliation(s)
- Fengying Jie
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Fang Dong
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Lingwen Xu
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Shuping Deng
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Qian Wang
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Qun Wu
- The Department of Intensive Care Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| |
Collapse
|
5
|
Wu YY, Chen MS, Chen IC, Wu FH, Liao TL, Wen HW, Nielsen BL, Liu HJ. Lidocaine Modulates Cytokine Production and Reprograms the Tumor Immune Microenvironment to Enhance Anti-Tumor Immune Responses in Gastric Cancer. Int J Mol Sci 2025; 26:3236. [PMID: 40244064 PMCID: PMC11989700 DOI: 10.3390/ijms26073236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Lidocaine, a local anesthetic, has been shown to modulate immune responses. This study examines its effects on cytokine production in peripheral blood mononuclear cells (PBMCs) from healthy donors and tumor-infiltrating immune cells (TIICs) from gastric cancer patients. PBMCs from healthy donors and TIICs from gastric cancer patients were treated with lidocaine. Cytokine production was assessed using flow cytometry and cytokine assays, with a focus on IFN-γ, IL-12, IL-10, TGF-β, and IL-35 levels. Cytotoxicity against primary gastric cancer cells (PGCCs) was also evaluated. Lidocaine inhibited IFN-γ production in CD8+ PBMCs and IL-12 in CD14+ PBMCs while increasing anti-inflammatory cytokines (IL-10, TGF-β, IL-35) in CD4+CD25+ and CD14+ cells. In TIICs, lidocaine enhanced IFN-γ and IL-12 production in CD8+ and CD14+ cells while reducing IL-10, TGF-β, and IL-35 levels, promoting an M1-like phenotype in macrophages. Mechanistically, lidocaine enhanced IFN-γ production in sorted CD8+ TIICs through G-protein-coupled receptor (GPCR) signaling and increased IL-12 production in sorted CD14+ TIICs via the toll-like receptor 4 (TLR4) signaling pathway. Lidocaine also increased IFN-γ production and cytotoxicity in CD8+ TIICs via NF-κB activation. Importantly, lidocaine did not affect the viability of PBMCs, TIICs, or PGCCs at concentrations up to 1.5 mM. Lidocaine reprogrammed the tumor immune microenvironment, enhancing anti-tumor immune responses, suggesting its potential to modulate immune functions in gastric cancer.
Collapse
Affiliation(s)
- Yi-Ying Wu
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan;
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi City 600, Taiwan;
| | - I-Chun Chen
- Department of Psychiatry, Taichung Veterans General Hospital, Taichung 407, Taiwan;
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Feng-Hsu Wu
- Division of General Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung 407, Taiwan;
- Department of Critical Care, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Department of Nursing, Hung Kuang University, Taichung 433, Taiwan
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan;
| | - Hsiao-Wei Wen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan;
| | - Brent L. Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA;
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan;
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
6
|
Duan Y, Qiu M, Na K, Liu D, Zhou S, Xu Y, Qi Z, Liu H, Xu K, Wang X, Li J, Li Y, Han Y. Inflammatory and Bleeding Risks on Clinical Outcomes in Acute Coronary Syndrome Patients Undergoing Percutaneous Coronary Intervention. Thromb Haemost 2025. [PMID: 39904360 DOI: 10.1055/a-2531-3268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
This study aimed to evaluate the impact of systemic inflammation burden using high-sensitivity C-reactive protein (hsCRP) and long-term prognosis in acute coronary syndrome (ACS) patients undergoing percutaneous coronary intervention (PCI) stratified by bleeding risk status.Consecutive patients admitted for ACS and who received PCI between March 2016 and March 2022 were enrolled in the analysis. Elevated systemic inflammation was defined as hsCRP >2 mg/L, and high bleeding risk (HBR) was defined the Academic Research Consortium (ARC)-HBR criteria. The primary outcome was ischemic events at 12 months, composed of cardiac death, myocardial infarction, and/or stroke. The main secondary outcomes included all-cause death, and Bleeding Academic Research Consortium (BARC) types 2, 3, and 5 bleeding and types 3 and 5 bleeding.Of 15,013 patients, 4,606 (30.7%) were qualified as HBR and 8,395 (55.9%) had hsCRP >2 mg/L. Elevated hsCRP was consistently associated with higher risk of ischemic events in both HBR (adjusted hazard ratio [aHR]: 1.20; 95% confidence interval [CI]: 0.91-1.58) and non-HBR (aHR: 1.34; 95% CI: 1.01-1.78) subgroups (P interaction = 0.755). Although the incidence of bleeding events was higher in HBR patients, an elevated hsCRP level was not associated with bleeding events regardless of HBR status. Restricted cubic spline regression represented an inverse J-shaped relation between hsCRP and non-HBR for ischemic events (P nonlinearity <0.001) and all-cause death (P nonlinearity = 0.003).Regardless of HBR status, high levels of hsCRP were associated with an increased risk of ischemic events and all-cause death in ACS patients following PCI, but not for bleeding.
Collapse
Affiliation(s)
- Yixuan Duan
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
- The Department of Cardiology, Air Force Medical University, Xijing Hospital, Xi'an, Shaanxi, China
| | - Miaohan Qiu
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Kun Na
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Daoshen Liu
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
- The Department of Cardiology, Air Force Medical University, Xijing Hospital, Xi'an, Shaanxi, China
| | - Shangxun Zhou
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
- The Department of Cardiology, Air Force Medical University, Xijing Hospital, Xi'an, Shaanxi, China
| | - Ying Xu
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Zizhao Qi
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Haiwei Liu
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Kai Xu
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaozeng Wang
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing Li
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Yi Li
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- Department of Cardiology, State Key Laboratory of Frigid Zone Cardiovascular Disease, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
7
|
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
|
8
|
Zhang Y, Tang Q, Cui J, Li Y, Xu H, Qiu Z, Lei S, Xue R, Sun Q, Xia Z. Bioinformatics analysis of ferroptosis-related hub genes and immunoinfiltration in myocardial ischemia/reperfusion following heart transplantation. BMC Cardiovasc Disord 2025; 25:16. [PMID: 39794696 PMCID: PMC11724544 DOI: 10.1186/s12872-024-04462-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Ischemia/reperfusion (I/R) is an inevitable pathophysiological process during heart transplantation, and ferroptosis is an important pathogenic mechanism. Unlike other modes of cell death, ferroptosis depends on the accumulation of iron within the cell and the oxidative degradation of polyunsaturated fatty acids. Dysregulation of this pathway has been linked to the progression of multiple pathological conditions, making it an attractive target for therapeutic intervention. Therefore, this study aims to explore the effect of ferroptosis on I/R during heart transplantation. METHODS GEO2R was applied to identify differentially expressed genes (DEGs) obtained from GSE50884 data, which was involved in I/R and heart transplantation. And ferroptosis-related DEGs (FRDEGs) were screened by venn diagram with ferroptosis-related genes downloaded from FerDb database. FRDEGs was enriched and analyzed by GO and KEGG, and hub genes related to ferroptosis were screened by Cytoscape software and database STRING. Additionally, considering the relationship between ferroptosis and immunity, CIBERSORTx was to analyze the infiltration of 22 kinds of immune cells in I/R during heart transplantation, and the correlation between each immune cell and the expression of FRDEGs was also discussed. Finally, the mouse model of heart transplantation with I/R was constructed, and the hub genes was verified by RT-qPCR and western blot. RESULTS 12 FRDEGs were identified out of 327 DEGs in GSE50844, which were mainly involved in ferroptosis and other pathways. Three hub genes (SLC7A11, PSAT1, ASNS) were obtained by the degree algorithm of cytohubba plug-in. Immunoinfiltration analysis showed that 16 of 22 immune cells changed, and the immune score of heart transplantation with I/R was higher than that without I/R. In addition, hub genes exhibited significant correlation with Eosinophils, NK cells resting, Dendritic cells resting, NK cells activated and T cells CD4 memory activated. We verified the expression of SLC7A11, PSAT1 and ASNS was higher than that in normal tissues using RT-qPCR and western blot in mouse models of heart transplantation with I/R, companied by ferroptosis aggravated is involved. CONCLUSIONS In short, ferroptosis is involved in I/R injury during heart transplantation, which is related to immune cell infiltration. Three hub genes (SLC7A11, PSAT1 and ASNS) identified in this study provide therapeutic targets for ameliorating I/R injury in heart transplantation.
Collapse
Affiliation(s)
- Yuxi Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China
| | - Qiao Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China
| | - Jiahui Cui
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China
| | - Yanan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China
| | - Heng Xu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China
| | - Rui Xue
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Qian Sun
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430061, China.
| |
Collapse
|
9
|
Huang X, Hu L, Li J, Tao S, Xue T. The relationship between inflammatory factors and heart failure: evidence based on bidirectional Mendelian randomization analysis. Front Cardiovasc Med 2024; 11:1378327. [PMID: 39726944 PMCID: PMC11669679 DOI: 10.3389/fcvm.2024.1378327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 11/30/2024] [Indexed: 12/28/2024] Open
Abstract
Objective Inflammatory factors play a crucial role in the onset and progression of heart failure. To further explore the causal relationship between inflammatory factors and heart failure, we employed bidirectional Mendelian randomization analysis to investigate the causal links between 91 inflammatory cytokines and heart failure. Methods We conducted our study using the bidirectional Mendelian randomization approach. Data on 91 inflammatory factors were sourced from large-scale public genome-wide association study databases, while heart failure data were obtained from the FINNGEN database. The relationships between inflammatory factors and heart failure were evaluated using five methods: MR-Egger regression model, Inverse Variance Weighted method, Simple mode model, Weighted mode model, and Weighted median. Results were subjected to FDR multiple testing correction, and significant findings were discussed in detail. To enhance the robustness of our findings, various sensitivity analyses were conducted, including MR Egger intercept, MR-PRESSO and Cochran Q test. Results Our forward Mendelian randomization study indicated that, of the 91 inflammatory factors examined, seven showed a causal relationship with heart failure. Four of these factors were significantly causally related to the incidence of heart failure: CXCL9 and IFN-γ as promotive factors, and LIFR and UPA as potential protective factors. Three inflammatory factors had a potential causal relationship with heart failure, with DNER as a potential protective factor, and MMP-1 and CD6 as potential promotive factors. Reverse Mendelian randomization suggested that the onset of heart failure might potentially influence the levels of four inflammatory factors, with ARTN and FGF5 decreasing after the onset of heart failure, and SLAM and MMP-10 increasing. Additionally, reliability tests of this Mendelian randomization, including MR-Egger intercept and MR-PRESSO tests, revealed no evidence of pleiotropy, and Cochran's Q test also confirmed the reliability of our results. Conclusion We identified CXCL9, IFN-γ, LIFR, and UPA as potential inflammatory factors associated with heart failure through forward Mendelian randomization. These findings suggest potential targets but require further validation.
Collapse
Affiliation(s)
- Xuanchun Huang
- Cardiology Department, Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Lanshuo Hu
- Institute of Digestive Diseases, Xiyuan Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Jun Li
- Cardiology Department, Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Shiyi Tao
- Cardiology Department, Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Tiantian Xue
- Cardiology Department, Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Ouyang X, Liu Z. Regulatory T cells and macrophages in atherosclerosis: from mechanisms to clinical significance. Front Immunol 2024; 15:1435021. [PMID: 39582868 PMCID: PMC11581946 DOI: 10.3389/fimmu.2024.1435021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Atherosclerosis is a complex pathological process, which causes diseases that threaten the health of an increasing number of people. Studies have found that the original view of lipid accumulation is not comprehensive because the use of lipid-lowering drugs alone cannot effectively treat atherosclerosis. As the study of the pathogenesis of atherosclerosis develops in-depth, the impact of immune-inflammatory response on atherosclerosis has garnered a great deal of attention. Some new advances have been made in the role of regulatory T cells (Tregs) and macrophages with unique immunomodulatory functions in atherosclerosis. Herein, the role of Tregs, macrophages, the mechanisms of Tregs-regulated macrophages, and the effects of potential factors on Tregs and macrophages in atherosclerosis are overviewed. Targeting Tregs and macrophages may provide new research strategies for the treatment of atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Xin Ouyang
- Clinical Medical College, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhongyong Liu
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
11
|
Fiore NT, Hayes JP, Williams SI, Moalem-Taylor G. Interleukin-35 alleviates neuropathic pain and induces an anti-inflammatory shift in spinal microglia in nerve-injured male mice. Brain Behav Immun 2024; 122:287-300. [PMID: 39097202 DOI: 10.1016/j.bbi.2024.07.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Immune cells are critical in promoting neuroinflammation and neuropathic pain and in facilitating pain resolution, depending on their inflammatory and immunoregulatory cytokine response. Interleukin (IL)-35, secreted by regulatory immune cells, is a member of the IL-12 family with a potent immunosuppressive function. In this study, we investigated the effects of IL-35 on pain behaviors, spinal microglia phenotype following peripheral nerve injury, and in vitro microglial cultures in male and female mice. Intrathecal recombinant IL-35 treatment alleviated mechanical pain hypersensitivity prominently in male mice, with only a modest effect in female mice after sciatic nerve chronic constriction injury (CCI). IL-35 treatment resulted in sex-specific microglial changes following CCI, reducing inflammatory microglial markers and upregulating anti-inflammatory markers in male mice. Spatial transcriptomic analysis revealed that IL-35 suppressed microglial complement activation in the superficial dorsal horn in male mice after CCI. Moreover, in vitro studies showed that IL-35 treatment of cultured inflammatory microglia mitigated their hypertrophied morphology, increased their cell motility, and decreased their phagocytic activity, indicating a phenotypic shift towards homeostatic microglia. Further, IL-35 altered microglial cytokines/chemokines in vitro, suppressing the release of IL-9 and monocyte-chemoattractant protein-1 and increasing IL-10 in the supernatant of male microglial cultures. Our findings indicate that treatment with IL-35 modulates spinal microglia and alleviates neuropathic pain in male mice, suggesting IL-35 as a potential sex-specific targeted immunomodulatory treatment for neuropathic pain.
Collapse
Affiliation(s)
- Nathan T Fiore
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia
| | - Jessica P Hayes
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia
| | - Sarah I Williams
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia
| | - Gila Moalem-Taylor
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia.
| |
Collapse
|
12
|
Zeng J, Liu J, Zhao N, Wong IN, Huang R. Caulerpa chemnitzia polysaccharide exerts immunomodulatory activity in macrophages by mediating the succinate/PHD2/HIF-1α/IL-1β pathway. Int J Biol Macromol 2024; 277:134450. [PMID: 39098690 DOI: 10.1016/j.ijbiomac.2024.134450] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Algal polysaccharide is an important food functional factor with diverse bioactive and low toxicity. Previous studies have confirmed Caulerpa chemnitzia polysaccharides (CRVP) have immunomodulatory activity, but the immunomodulatory mechanism of CRVP in macrophages has not been thoroughly explored yet. In our research, we found that CRVP has outstanding immunomodulatory activity in macrophages, which is reflected in promoting cell proliferation, upregulating cytokines (IL-1β, IL-6, and TNF-α) expression, and increasing NO and ROS levels. Additionally, the result of joint analysis of untargeted metabolomics showed metabolism played a major role in the immunomodulatory of CRVP and suggested succinic acid was a key metabolite. Further verification indicated that the accumulation of succinic acid in macrophages after administered with CRVP, induced the down-regulation of prolyl hydroxylase domain 2 (PHD2) and up-regulation of hypoxia-inducible factor-1α (HIF-1α), thereby enhancing IL-1β expression. Together, the immunomodulatory activity of CRVP in macrophages via succinate/PHD2/HIF-1α/IL-1β pathway.
Collapse
Affiliation(s)
- Jinzi Zeng
- Guangdong Provincial Key Laboratory of Food Quality and Safety/College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Liu
- Laboratory of Pathogenic Biology, Guangdong Medical University, Zhanjiang 524023, China
| | - Ning Zhao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen 518104, China
| | - Io Nam Wong
- Faculty of Medicine, Macau University of Science and Technology, Macau 999078, Macau.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety/College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
13
|
Kumar V, Narisawa M, Cheng XW. Overview of multifunctional Tregs in cardiovascular disease: From insights into cellular functions to clinical implications. FASEB J 2024; 38:e23786. [PMID: 38979903 DOI: 10.1096/fj.202400839r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/01/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
Regulatory T cells (Tregs) are crucial in regulating T-cell-mediated immune responses. Numerous studies have shown that dysfunction or decreased numbers of Tregs may be involved in inflammatory cardiovascular diseases (CVDs) such as atherosclerosis, hypertension, myocardial infarction, myocarditis, cardiomyopathy, valvular heart diseases, heart failure, and abdominal aortic aneurysm. Tregs can help to ameliorate CVDs by suppressing excessive inflammation through various mechanisms, including inhibition of T cells and B cells, inhibition of macrophage-induced inflammation, inhibition of dendritic cells and foam cell formation, and induction of anti-inflammatory macrophages. Enhancing or restoring the immunosuppressive activity of Tregs may thus serve as a fundamental immunotherapy to treat hypertension and CVDs. However, the precise molecular mechanisms underlying the Tregs-induced protection against hypertension and CVDs remain to be investigated. This review focuses on recent advances in our understanding of Tregs subsets and function in CVDs. In addition, we discuss promising strategies for using Tregs through various pharmacological approaches to treat hypertension and CVDs.
Collapse
Affiliation(s)
- Vipin Kumar
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P.R. China
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, P.R. China
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
14
|
Feng J, Li K, Xie F, Han L, Wu Y. IL-35 ameliorates lipopolysaccharide-induced endothelial dysfunction by inhibiting endothelial-to-mesenchymal transition. Int Immunopharmacol 2024; 129:111567. [PMID: 38335651 DOI: 10.1016/j.intimp.2024.111567] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome (SIRS) caused mainly by bacterial infection. The morbidity and mortality rates of sepsis are extremely high. About 18 million people worldwide suffer from severe sepsis each year, and about 14,000 people die from it every day. Previous studies have revealed that endothelial dysfunction plays a vital role in the pathological change of sepsis. Furthermore, endothelial-mesenchymal transition (EndMT, EndoMT) is capable of triggering endothelial dysfunction. And yet, it remains obscure whether interleukin-35 (IL-35) can alleviate endothelial dysfunction by attenuating LPS-induced EndMT. Here, through in vivo and in vitro experiments, we revealed that IL-35 has a previously unknown function to attenuate LPS-induced endothelial dysfunction by inhibiting LPS-induced EndMT. Mechanistically, IL-35 acts by regulating the NFκB signaling pathway.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Kai Li
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Feng Xie
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Leilei Han
- 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
|
15
|
Yi P, Yu W, Xiong Y, Dong Y, Huang Q, Lin Y, Du Y, Hua F. IL-35: New Target for Immunotherapy Targeting the Tumor Microenvironment. Mol Cancer Ther 2024; 23:148-158. [PMID: 37988561 DOI: 10.1158/1535-7163.mct-23-0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/15/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023]
Abstract
Interleukin 35(IL-35) is a newly discovered inhibitory cytokine of the IL12 family. More recently, IL-35 was found to be increased in the tumor microenvironment (TME) and peripheral blood of many patients with cancer, indicating that it plays an important role in the TME. Tumors secrete cytokines that recruit myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg) into the TME to promote malignant progression, which is a great challenge for cancer treatment. Radiotherapy causes serious adverse effects, and tumor resistance to immune checkpoint inhibitors is still an unsolved challenge. Thus, new cancer therapy approaches are urgently needed. Numerous studies have shown that IL-35 can recruit immunosuppressive cells to enable tumor immune escape by promoting the conversion of immune cells into a tumor growth-promoting phenotype as well as facilitating tumor angiogenesis. IL-35-neutralizing antibodies were found to boost the chemotherapeutic effect of gemcitabine and considerably reduce the microvascular density of pancreatic cancer in mice. Therefore, targeting IL-35 in the TME provides a promising cancer treatment target. In addition, IL-35 may be used as an independent prognostic factor for some tumors in the near future. This review intends to reveal the interplay of IL-35 with immune cells in the TME, which may provide new options for the treatment of cancer.
Collapse
Affiliation(s)
- Pengcheng Yi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Wenjun Yu
- Fuzhou First People's Hospital of Jiangxi Province, Fuzhou City, Jiangxi Province, P.R. China
| | - Yanhong Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yao Dong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Qiang Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| | - Yunfei Du
- Department of Anesthesiology, Nanchang Central Hospital, Nanchang, Jiangxi, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, P.R. China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang City, Jiangxi Province, P.R. China
| |
Collapse
|
16
|
Chaszczewska-Markowska M, Górna K, Bogunia-Kubik K, Brzecka A, Kosacka M. The Influence of Comorbidities on Chemokine and Cytokine Profile in Obstructive Sleep Apnea Patients: Preliminary Results. J Clin Med 2023; 12:jcm12030801. [PMID: 36769452 PMCID: PMC9918226 DOI: 10.3390/jcm12030801] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION Obstructive sleep apnea (OSA) is frequently associated with a chronic inflammatory state and cardiovascular/metabolic complications. The aim of this study was to evaluate the influence of certain comorbidities on a panel of 45 chemokines and cytokines in OSA patients with special regard to their possible association with cardiovascular diseases. MATERIAL AND METHODS This cross-sectional study was performed on 61 newly diagnosed OSA patients. For the measurement of the plasma concentration of chemokines and cytokines, the magnetic bead-based multiplex assay for the Luminex® platform was used. RESULTS In the patients with concomitant COPD, there were increased levels of pro-inflammatory cytokines (CCL11, CD-40 ligand) and decreased anti-inflammatory cytokine (IL-10), while in diabetes, there were increased levels of pro-inflammatory cytokines (IL-6, TRIAL). Obesity was associated with increased levels of both pro-inflammatory (IL-13) and anti-inflammatory (IL-1RA) cytokines. Hypertension was associated with increased levels of both pro-inflammatory (CCL3) and anti-inflammatory (IL-10) cytokines. Increased daytime pCO2, low mean nocturnal SaO2, and the oxygen desaturation index were associated with increased levels of pro-inflammatory cytokines (CXCL1, PDGF-AB, TNF-α, and IL-15). CONCLUSIONS In OSA patients with concomitant diabetes and COPD, elevated levels of certain pro-inflammatory and decreased levels of certain anti-inflammatory cytokines may favor the persistence of a chronic inflammatory state with further consequences. Nocturnal hypoxemia, frequent episodes of desaturation, and increased daytime pCO2 are factors contributing to the chronic inflammatory state in OSA patients.
Collapse
Affiliation(s)
- Monika Chaszczewska-Markowska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-422 Wroclaw, Poland
| | - Katarzyna Górna
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-422 Wroclaw, Poland
- Correspondence:
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-422 Wroclaw, Poland
| | - Anna Brzecka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, 53-439 Wroclaw, Poland
| | - Monika Kosacka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, 53-439 Wroclaw, Poland
| |
Collapse
|
17
|
Song Z, Zhao Y, Li Z, Cao C, Liu G, Liu Q, Zhang X, Dai D, Zheng Z, Zhao C, Yu H. Study on the Micro Removal Process of Inner Surface of Cobalt Chromium Alloy Cardiovascular Stent Tubes. MICROMACHINES 2022; 13:1374. [PMID: 36143997 PMCID: PMC9501946 DOI: 10.3390/mi13091374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 06/16/2023]
Abstract
Due to the special manufacturing process of cobalt-chromium alloy cardiovascular stent tubes, there are serious surface defects in their inner walls, which affects the therapeutic effect after implantation. At the same time, the traditional processing technology cannot finish the inner wall of a cardiovascular stent tube. In light of the above problems, magnetic abrasive finishing (MAF) equipment for the inner wall of an ultra-fine and ultra-long cardiovascular stent tube is proposed, and MAF technology is used to improve the surface quality of its inner wall. High-performance spherical magnetic abrasive powders are used to finish the inner wall of a cobalt-chromium alloy cardiovascular stent tube with an inner diameter of 1.6 mm and an outer diameter of 1.8 mm. The effects of finishing time, tube rotational speed, feed speed of the magnetic pole, MAPs filling quantity, and MAP abrasive size on the surface roughness and material removal thickness of cobalt-chromium alloy cardiovascular stent tube are investigated. The results show that the surface roughness of the inner wall of the cobalt-chromium alloy cardiovascular stent decreases from 0.485 μm to 0.101 μm, and the material removal thickness of the defect layer is 4.3 μm. MAF technology is used to solve the problem of the poor surface quality of the inner walls of ultra-fine and ultra-long cobalt-chromium alloy cardiovascular stent tubes.
Collapse
|