1
|
Lim WW, Leung JH, Xie C, Cheng AWT, Su L, Lum LN, Toh A, Kong SC, Takano AM, Hausenloy DJ, Chua YC. Circulating Interleukins as Biomarkers in Non-Small Cell Lung Cancer Patients: A Pilot Study Compared to Normal Individuals. Diseases 2024; 12:221. [PMID: 39329890 PMCID: PMC11430979 DOI: 10.3390/diseases12090221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Identifying biomarkers in non-small cell lung cancer (NSCLC) can improve diagnosis and patient stratification. We evaluated plasmas and sera for interleukins (IL)-11, IL-6, IL-8, IL-17A, and IL-33 as biomarkers in primary NSCLC patients undergoing surgical treatment against normal volunteers. Exhaled-breath condensates (EBCs), a potential source without invasive procedures, were explored in normal individuals. Due to separate recruitment criteria and intrinsic cohort differences, the NSCLC and control cohorts were not well matched for age (median age: 65 vs. 40 years; p < 0.0001) and smoking status (p = 0.0058). Interleukins were first assessed through conventional ELISA. IL-11 was elevated in NSCLC plasma compared to controls (49.71 ± 16.90 vs. 27.67 ± 14.06 pg/mL, respectively, p < 0.0001) but undetectable in sera and EBCs by conventional ELISA. Therefore, high-sensitivity PCR-based IL-11 ELISA was repeated, albeit with concentration discrepancies. IL11 gene and protein upregulation by RT-qPCR and immunohistochemistry, respectively, were validated in NSCLC tumors. The lack of detection sensitivity across IL-6, IL-8, IL-17A, and IL-33 suggests the need for further, precise assays. Surprisingly, biomarker concentrations can be dissimilar across paired plasmas and sera. Our results identified a need to optimize detection limits for biomarker detection and caution against over-reliance on just one form of blood sample for biomarker assessment.
Collapse
Affiliation(s)
- Wei-Wen Lim
- National Heart Research Institute of Singapore, National Heart Center Singapore, Singapore 169609, Singapore
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore, Singapore 169857, Singapore
| | - Jason H Leung
- Department of Cardiothoracic Surgery, National Heart Center Singapore, Singapore 169609, Singapore
| | - Chen Xie
- National Heart Research Institute of Singapore, National Heart Center Singapore, Singapore 169609, Singapore
| | - Angelina W T Cheng
- National Heart Research Institute of Singapore, National Heart Center Singapore, Singapore 169609, Singapore
| | - Liping Su
- National Heart Research Institute of Singapore, National Heart Center Singapore, Singapore 169609, Singapore
| | - Luh-Nah Lum
- Clinical and Translational Research Office, National Heart Center Singapore, Singapore 169609, Singapore
| | - Aishah Toh
- Clinical and Translational Research Office, National Heart Center Singapore, Singapore 169609, Singapore
| | - Siew-Ching Kong
- Clinical and Translational Research Office, National Heart Center Singapore, Singapore 169609, Singapore
| | - Angela M Takano
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Derek J Hausenloy
- National Heart Research Institute of Singapore, National Heart Center Singapore, Singapore 169609, Singapore
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore, Singapore 169857, Singapore
- Yong Loo Lin School of Medicine, National University Singapore, Singapore 117597, Singapore
- The Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK
| | - Yang C Chua
- Department of Cardiothoracic Surgery, National Heart Center Singapore, Singapore 169609, Singapore
| |
Collapse
|
2
|
Ganizada BH, Veltrop RJA, Akbulut AC, Koenen RR, Accord R, Lorusso R, Maessen JG, Reesink K, Bidar E, Schurgers LJ. Unveiling cellular and molecular aspects of ascending thoracic aortic aneurysms and dissections. Basic Res Cardiol 2024; 119:371-395. [PMID: 38700707 PMCID: PMC11143007 DOI: 10.1007/s00395-024-01053-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/03/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024]
Abstract
Ascending thoracic aortic aneurysm (ATAA) remains a significant medical concern, with its asymptomatic nature posing diagnostic and monitoring challenges, thereby increasing the risk of aortic wall dissection and rupture. Current management of aortic repair relies on an aortic diameter threshold. However, this approach underestimates the complexity of aortic wall disease due to important knowledge gaps in understanding its underlying pathologic mechanisms.Since traditional risk factors cannot explain the initiation and progression of ATAA leading to dissection, local vascular factors such as extracellular matrix (ECM) and vascular smooth muscle cells (VSMCs) might harbor targets for early diagnosis and intervention. Derived from diverse embryonic lineages, VSMCs exhibit varied responses to genetic abnormalities that regulate their contractility. The transition of VSMCs into different phenotypes is an adaptive response to stress stimuli such as hemodynamic changes resulting from cardiovascular disease, aging, lifestyle, and genetic predisposition. Upon longer exposure to stress stimuli, VSMC phenotypic switching can instigate pathologic remodeling that contributes to the pathogenesis of ATAA.This review aims to illuminate the current understanding of cellular and molecular characteristics associated with ATAA and dissection, emphasizing the need for a more nuanced comprehension of the impaired ECM-VSMC network.
Collapse
MESH Headings
- Humans
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Aortic Dissection/metabolism
- Animals
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Vascular Remodeling
- Extracellular Matrix/pathology
- Extracellular Matrix/metabolism
- Phenotype
Collapse
Affiliation(s)
- Berta H Ganizada
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Rogier J A Veltrop
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Asim C Akbulut
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Ryan Accord
- Department of Cardiothoracic Surgery, Center for Congenital Heart Disease, University Medical Center Groningen, Groningen, The Netherlands
| | - Roberto Lorusso
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Jos G Maessen
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Koen Reesink
- Department of Biomedical Engineering, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Elham Bidar
- Department of Cardiothoracic Surgery, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
- CARIM, Cardiovascular Research Institute Maastricht, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
3
|
Guo X, Zhu X, Zhou S, Dong X. Serum levels of IL-9 and IL-11 serve as predictors for the occurrence of early neurologic deterioration in patients with cerebral infarction. Clin Chim Acta 2024; 558:119683. [PMID: 38643817 DOI: 10.1016/j.cca.2024.119683] [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: 02/22/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND AND AIM Early neurological deterioration (END) is a common complication of cerebral infarction and a significant contributor to poor prognosis. Our study aimed to investigate the predictive value of interleukin-9 (IL-9) and interleukin-11 (IL-11) in relation to the occurrence of END in patients with cerebral infarction. MATERIALS AND METHODS 102 patients with cerebral infarction and 64 healthy controls were collected. Patients were categorized into two groups based on the development of END following admission: the END group (n = 44) and the non-END group (n = 58). Enzyme-linked immunosorbent assay was used to determine the serum levels of IL-9, IL-11, and BDNF. RESULTS Serum IL-9 was higher and IL-11 lower in the END group than those in the non-END group (P < 0.01). IL-9 correlated positively with NIHSS score (r = 0.627) and infarction volume (r = 0.686), while IL-11 correlated negatively (r = -0.613, -0.679, respectively). Logistic regression identified age, NIHSS score, and IL-9 as risk factors (P < 0.01), and IL-11 as protective (P < 0.01). Combined IL-9 and IL-11 had an ROC curve area of 0.849. BDNF correlated negatively with IL-9 (r = -0.703) and positively with IL-11 (r = 0.711). CONCLUSION Serum IL-9 and IL-11 levels can predict the occurrence of END in patient with cerebral infarction and are correlated with serum BDNF levels.
Collapse
Affiliation(s)
- Xianglin Guo
- Department of Emergency, People's Hospital of Yangzhong City, Yangzhong, Jiangsu 212200, China
| | - Xiaoyan Zhu
- Department of Neurology, People's Hospital of Yangzhong City, Yangzhong, Jiangsu 212200, China; School of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, Jiangsu 225000, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225000, China
| | - Shuai Zhou
- Department of Neurology, People's Hospital of Yangzhong City, Yangzhong, Jiangsu 212200, China; School of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, Jiangsu 225000, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225000, China
| | - Xiaohong Dong
- Department of Neurology, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China.
| |
Collapse
|
4
|
Matniyaz Y, Luo YX, Jiang Y, Zhang KY, Wang WZ, Pan T, Wang DJ, Xue YX. Short- and Long-term survival prediction in patients with acute type A aortic dissection undergoing open surgery. J Cardiothorac Surg 2024; 19:171. [PMID: 38566106 PMCID: PMC10988835 DOI: 10.1186/s13019-024-02687-x] [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: 08/07/2023] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Acute Type A aortic dissection (ATAAD) is a life-threatening cardiovascular disease associated with high mortality rates, where surgical intervention remains the primary life-saving treatment. However, the mortality rate for ATAAD operations continues to be alarmingly high. To address this critical issue, our study aimed to assess the correlation between preoperative laboratory examination, clinical imaging data, and postoperative mortality in ATAAD patients. Additionally, we sought to establish a reliable prediction model for evaluating the risk of postoperative death. METHODS In this study, a total of 384 patients with acute type A aortic dissection (ATAAD) who were admitted to the emergency department for surgical treatment were included. Based on preoperative laboratory examination and clinical imaging data of ATAAD patients, logistic analysis was used to obtain independent risk factors for postoperative in-hospital death. The survival prediction model was based on cox regression analysis and displayed as a nomogram. RESULTS Logistic analysis identified several independent risk factors for postoperative in-hospital death, including Marfan syndrome, previous cardiac surgery history, previous renal dialysis history, direct bilirubin, serum phosphorus, D-dimer, white blood cell, multiple aortic ruptures and age. A survival prediction model based on cox regression analysis was established and presented as a nomogram. The model exhibited good discrimination and significantly improved the prediction of death risk in ATAAD patients. CONCLUSIONS In this study, we developed a novel survival prediction model for acute type A aortic dissection based on preoperative clinical features. The model demonstrated good discriminatory power and improved accuracy in predicting the risk of death in ATAAD patients undergoing open surgery.
Collapse
Affiliation(s)
- Yusanjan Matniyaz
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
- Department of Cardiac Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Yuan-Xi Luo
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Yi Jiang
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Ke-Yin Zhang
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Wen-Zhe Wang
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China
| | - Tuo Pan
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
| | - Dong-Jin Wang
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
- Department of Cardiac Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
- Department of Cardiac Surgery, Affiliated Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
- Department of Cardiac Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
| | - Yun-Xing Xue
- Department of Cardiac Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Number 321 Zhongshan Road, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
5
|
Braga YLL, do Carmo Neto JR, Franco PIR, Helmo FR, Dos Reis MA, de Oliveira FA, Celes MRN, da Silva MV, Machado JR. The Influence of IL-11 on Cardiac Fibrosis in Experimental Models: A Systematic Review. J Cardiovasc Dev Dis 2024; 11:65. [PMID: 38392279 PMCID: PMC10888948 DOI: 10.3390/jcdd11020065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Fibrosis is one of the main factors that impair the function of many organs. In the heart, fibrosis leads to contractile dysfunction and arrhythmias, which are important in the development of heart failure. Interleukin (IL)-11 is regulated in various heart diseases and has recently been reported to be an important cytokine in fibrosis in this organ. However, this topic has been little explored, and many questions persist. Thus, this systematic review aimed to report on possible IL-11 therapies evaluated in rodent model-induced cardiac fibrosis. Inclusion criteria were experimental in vivo studies that used different rodent models for cardiac fibrosis associated with IL-11 interventions, without year and language restrictions. The search in PubMed, Web of Science, and Embase databases was performed in October 2022. The risk of bias assessment of the studies was based on the guidelines of the SYRCLE tool, and data from the selected articles were also presented in a table as a narrative description. This review was based on eight studies in which five different interventions were used: recombinant human IL-11 (rhIL-11), anti-IL11 (X203), recombinant mouse IL-11 (rmIL-11), lentivirus (LV)-IL-11 + lutein, and anti-IL11RA (X209). Based on the included studies, the results were variable, with IL-11 overexpression inducing cardiac fibrosis, while inhibition protected against this process, preserving the function of this organ. Therefore, IL-11 stands out as a promising therapeutic target for cardiac fibrosis. However, further studies are needed to understand the mechanisms triggered by each treatment, as well as its safety and immunogenicity.
Collapse
Affiliation(s)
- Yarlla Loyane Lira Braga
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - José Rodrigues do Carmo Neto
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - Pablo Igor Ribeiro Franco
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | | | | | - Flávia Aparecida de Oliveira
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - Mara Rúbia Nunes Celes
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
| | - Marcos Vinícius da Silva
- Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Juliana Reis Machado
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania 74605-450, GO, Brazil
- General Pathology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
- Department of General Pathology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| |
Collapse
|
6
|
Li J, Yu C, Yu K, Chen Z, Xing D, Zha B, Xie W, Ouyang H. SPINT2 is involved in the proliferation, migration and phenotypic switching of aortic smooth muscle cells: Implications for the pathogenesis of thoracic aortic dissection. Exp Ther Med 2023; 26:546. [PMID: 37928510 PMCID: PMC10623238 DOI: 10.3892/etm.2023.12245] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/25/2023] [Indexed: 11/07/2023] Open
Abstract
Thoracic aortic dissection (TAD) is a severe and extremely dangerous cardiovascular disease. Proliferation, migration and phenotypic switching of vascular smooth muscle cells (SMCs) are major pathogenetic mechanisms involved in the development of TAD. The present study was designed to investigate the expression and potential function of serine peptidase inhibitor Kunitz type 2 (SPINT2) in TAD. The gene expression profile data for ascending aorta from patients with TAD were downloaded from the GEO database with the accession number GSE52093. Bioinformatics analysis using GEO2R indicated that the differentially expressed SPINT2 was prominently decreased in TAD. The expression levels of SPINT2 mRNA and protein in aortic dissection specimens and normal aorta tissues were measured using reverse transcription-quantitative PCR and western blotting. SPINT2 expression was downregulated in clinical samples from aortic dissection specimens of patients with TAD compared with the corresponding expression noted in tissues derived from patients without TAD. In vitro, platelet-derived growth factor BB (PDGF-BB) was applied to induce the isolated primary mouse aortic SMC phenotypic modulation (a significant upregulation in the expression levels of synthetic markers), and the SMCs were infected with the adenoviral vector, Ad-SPINT2, to construct SPINT2-overexpressed cell lines. SMC viability was detected by an MTT assay and SMC proliferation was detected via the presence of Ki-67-positive cells (immunofluorescence staining). To explore the effects of SPINT2 on SMC migration, a wound healing assay was conducted. ELISA and western blotting assays were used to measure the content and expression levels of MMP-2 and MMP-9. The expression levels of vimentin, collagen I, α-SMA and SM22α were measured using western blotting. The PDGF-BB-induced proliferation and migration of SMCs were recovered by SPINT2 overexpression. The increase in the expression levels of SPINT2 reduced the expression levels of active matrix metalloproteinases (MMPs), MMP-2 and MMP-9. Overexpression of SPINT2 suppressed SMC switching from a contractile to a synthetic type, as evidenced by decreased vimentin and collagen I expression levels along with increased α-smooth muscle actin and smooth muscle protein 22-α expression levels. Furthermore, activation of ERK was inhibited in SPINT2-overexpressing SMCs. A specific ERK agonist, 12-O-tetradecanoylphorbol-13-acetate, reversed the SPINT2-mediated inhibition of SMC migration and the phenotypic switching. Collectively, the data indicated that SPINT2 was implicated in the proliferation, migration and phenotypic switching of aortic SMCs, suggesting that it may be involved in TAD progression.
Collapse
Affiliation(s)
- Jun Li
- Department of Vascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Changjun Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Kangmin Yu
- Department of Vascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Zhiyong Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Dan Xing
- Department of Medical Record Management, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Binshan Zha
- Department of Vascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Wentao Xie
- Department of Vascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Huan Ouyang
- Department of Vascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
7
|
Ye W, Wang Q, Zhao L, Wang C, Zhang D, Zhou M, Chen F, Wang W, Zhu Z, Guo W, Liu Y, Zou H, Xue Y. Blockade of IL-11 Trans-Signaling or JAK2/STAT3 Signaling Ameliorates the Profibrotic Effect of IL-11. Immunol Invest 2023; 52:703-716. [PMID: 37401665 DOI: 10.1080/08820139.2023.2222746] [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] [Indexed: 07/05/2023]
Abstract
OBJECTIVES Systemic sclerosis (SSc) is a rare rheumatic disease characterized by vascular damage, dysregulated immune response, and fibrosis. Interleukin-11 (IL-11) is upregulated in SSc. This study aimed to investigate the pathological and therapeutic role of the IL-11 trans-signaling pathway in SSc. METHODS Plasma IL-11 level was evaluated in 32 patients with SSc and 15 healthy controls, while the expression levels of ADAM10, ADAM17, IL-11, IL-11 Rα, or IL-11 co-stained with CD3 or CD163 in the skin of SSc patients and healthy controls were analyzed. Fibroblasts were treated with IL-11 and ionomycin to evaluate the profibrotic effect of IL-11 trans-signaling pathway. TJ301 (sgp130Fc) and WP1066 (a JAK2/STAT3 inhibitor) intervention groups were set up to investigate the antifibrotic effect of targeting IL-11. RESULTS Levels of plasma IL-11 were extremely low in most SSc patients and healthy controls. In contrast, levels of IL-11, IL-11 Rα, and ADAM10, but not ADAM17, were significantly elevated in the skin of SSc patients. Moreover, the numbers of IL-11+ CD3+ cells and IL-11+ CD163+ cells were increased in the skin of SSc patients. Besides, IL-11 and ADAM10 were also elevated in the skin and pulmonary of bleomycin-induced SSc mouse. Fibroblasts co-stimulated with IL-11 and ionomycin showed increased expression of COL3 and phosphorylation of STAT3, which could be inhibited by TJ301 or WP1066. TJ301 also ameliorated skin and lung fibrosis in BLM-induced SSc mouse. CONCLUSIONS IL-11 induces fibrosis in SSc by regulating the trans-signaling pathway. Blockage of sgp130Fc or inhibition of the JAK2/STAT3 pathway could ameliorate the profibrotic effect of IL-11.
Collapse
Affiliation(s)
- Wenjing Ye
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qian Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Zhao
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Changcheng Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dandan Zhang
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mengyu Zhou
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fangfang Chen
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiguo Wang
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zaihua Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenyu Guo
- Clinical Development, I-Mab Biopharma, Hangzhou, China
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Xue
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Song J, Peng H, Lai M, Kang H, Chen X, Cheng Y, Su X. Relationship between inflammatory-related cytokines with aortic dissection. Int Immunopharmacol 2023; 122:110618. [PMID: 37480750 DOI: 10.1016/j.intimp.2023.110618] [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: 04/17/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/24/2023]
Abstract
Aortic dissection, characterized by severe intramural hematoma formation and acute endometrial rupture, is caused by excessive bleeding within the aortic wall or a severe tear within the intimal layer of the aorta, which subsequently promotes the separation or dissection in the layers of the aortic wall. Epidemiological surveys showed that aortic dissection was most observed among those patients from 55 to 80 years of age, with a prevalence of approximately 40 cases per 100,000 individuals per year, posing serious risks to future health and leading to high mortality. Other risk factors of aortic dissection progression contained dyslipidemia, hypertension, and genetic disorders, such as Marfan syndrome. Currently, emerging evidence indicates the pathological progression of aortic dissection is significantly complicated, which is correlated with the aberrant infiltration of pro-inflammatory cells into the aortic wall, subsequently facilitating the apoptosis of vascular smooth muscle cells (VSMCs) and inducing the aberrant expression of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interferon (IF). Other pro-inflammatory-related cytokines, including the colony-stimulating factor (CSF), chemotactic factor, and growth factor (GF), played an essential function in facilitating aortic dissection. Multiple studies focused on the important relationship between pro-inflammatory cytokines and aortic dissection, which could deepen the understanding of aortic dissection and further guide the therapeutic strategies in clinical practice. The present review elucidated pro-inflammatory cytokines' functions in modulating the risk of aortic dissection are summarized. Moreover, the emerging evidence that aimed to elucidate the potential mechanisms wherebyvarious pro-inflammatory cytokines affected the pathological development of aortic dissection was also listed.
Collapse
Affiliation(s)
- Jingjin Song
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Hua Peng
- Department of Cardiac Macrovascular Surgery, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Min Lai
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Huiyuan Kang
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiaofang Chen
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ye Cheng
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Xin Su
- Department of Cardiology, The Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
9
|
Wu J, Ma W, Qiu Z, Zhou Z. Roles and mechanism of IL-11 in vascular diseases. Front Cardiovasc Med 2023; 10:1171697. [PMID: 37304948 PMCID: PMC10250654 DOI: 10.3389/fcvm.2023.1171697] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/28/2023] [Indexed: 06/13/2023] Open
Abstract
Vascular diseases are the leading cause of morbidity and mortality worldwide. Therefore, effective treatment strategies that can reduce the risk of vascular diseases are urgently needed. The relationship between Interleukin-11 (IL-11) and development of vascular diseases has gained increasing attention. IL-11, a target for therapeutic research, was initially thought to participate in stimulating platelet production. Additional research concluded that IL-11 is effective in treating several vascular diseases. However, the function and mechanism of IL-11 in these diseases remain unknown. This review summarizes IL-11 expression, function, and signal transduction mechanism. This study also focuses on the role of IL-11 in coronary artery disease, hypertension, pulmonary hypertension, cerebrovascular disease, aortic disease, and other vascular diseases and its potential as a therapeutic target. Consequently, this study provides new insight into the clinical diagnosis and treatment of vascular diseases.
Collapse
Affiliation(s)
- Jiacheng Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenrui Ma
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Wuhan, China
| | - Zhihua Qiu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Luo J, Shi H, Ran H, Zhang C, Wu Q, Shao Y. Identification of key biomarkers and immune infiltration in the thoracic acute aortic dissection by bioinformatics analysis. BMC Cardiovasc Disord 2023; 23:75. [PMID: 36755239 PMCID: PMC9909862 DOI: 10.1186/s12872-023-03110-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Thoracic acute aortic dissection (TAAD), one of the most fatal cardiovascular diseases, leads to sudden death, however, its mechanism remains unclear. METHODS Three Gene Expression Omnibus datasets were employed to detect differentially expressed genes (DEGs). A similar function and co-expression network was identified by weighted gene co-expression network analysis. The least absolute shrinkage and selection operator, random forest, and support vector machines-recursive feature elimination were utilized to filter diagnostic TAAD markers, and then screened markers were validated by quantitative real-time PCR and another independent dataset. CIBERSORT was deployed to analyze and evaluate immune cell infiltration in TAAD tissues. RESULTS Twenty-five DEGs were identified and narrowed down to three after screening. Finally, two genes, SLC11A1 and FGL2, were verified by another dataset and qRT-PCR. Function analysis revealed that SLC11A1 and FGL2 play significant roles in immune-inflammatory responses. CONCLUSION SLC11A1 and FGL2 are differently expressed in aortic dissection and may be involved in immune-inflammatory responses.
Collapse
Affiliation(s)
- Jun Luo
- grid.452206.70000 0004 1758 417XDepartment of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Haoming Shi
- grid.203458.80000 0000 8653 0555Chongqing Medical University, Chongqing, China
| | - Haoyu Ran
- grid.203458.80000 0000 8653 0555Chongqing Medical University, Chongqing, China
| | - Cheng Zhang
- grid.452206.70000 0004 1758 417XDepartment of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Qingchen Wu
- grid.452206.70000 0004 1758 417XDepartment of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China
| | - Yue Shao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
11
|
Cong X, Tian B, Zhu X, Zhang X, Gu W, Zhao H, Hao S, Ning Z. Interleukin-11 Is Elevated in Patients with Atrial Fibrillation, Correlates with Serum Fibrosis Markers, and Represents a Therapeutic Target for Atrial Fibrosis. Cerebrovasc Dis 2023; 52:575-586. [PMID: 36599329 DOI: 10.1159/000527740] [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: 08/04/2022] [Accepted: 10/11/2022] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Inflammatory cytokines are closely associated with developing cardiac fibrosis. This research aimed to explore the significant role of IL-11 in atrial fibrosis progression and potential therapeutic targets. METHODS 207 AF patients and 160 healthy subjects were included in the case-control study. Blood samples were analyzed for the level of IL-11 by enzyme-linked immunosorbent assay (ELISA). Angiotensin II (Ang II)-treated fibrosis mouse models were generated, and expression of IL-11 mRNA and protein was detected by RT-qPCR and Western blot. IL-11 antagonist was used to evaluating atrial fibrosis-related markers. RESULTS The persistent atrial fibrillation patients (n = 76) had significantly larger left atrial size, higher serum levels of hypertrophic protein BNP, proinflammatory cytokine high-sensitivity C-reactive protein (hs-CRP), and interleukin-6 (IL-6) compared to paroxysmal atrial fibrillation patients (n = 131), and healthy subjects (all p < 0.05). Pearson correlation analysis revealed significant positive correlation between serum IL-11 and cardiac fibrosis markers BNP (r = 0.394, p < 0.001), CTX-I (r = 0.418, p < 0.001), PICP (r = 0.306, p < 0.001), PIIINP (r = 0.335, p < 0.001), and TGF-β1 (r = 0.273, p < 0.001). In the fibrosis mouse model, Ang II infusion significantly upregulated IL-11 mRNA and protein expression in the left atrium of mice (p < 0.05), as well as staining intensity of Masson trichrome, the intensity of α-SMA, and it increased mRNA expression of collagen I and III in atrial tissue. IL-11 antagonist treatment significantly attenuated Masson trichrome, number of α-SMA-positive myofibroblasts in atrial tissue. Also, it significantly reduced the p-ERK1/2 in atrial tissue of mice infused with Ang II (p < 0.05). CONCLUSIONS IL-11 is upregulated in the serum of AF patients, and IL-11 inhibitor significantly inhibited Ang II-induced atrial fibrosis, a key pathological feature of AF. Therefore, IL-11 could be a potential therapeutic target for AF.
Collapse
Affiliation(s)
- Xinpeng Cong
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Bei Tian
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Xi Zhu
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Xiaogang Zhang
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Wei Gu
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Hanjun Zhao
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Shuwen Hao
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| | - Zhongping Ning
- Department of Cardiology, Zhoupu Hospital Affiliated to Shanghai Health Medical College, Shanghai, China
| |
Collapse
|
12
|
Cai Y, Zhou Y, Li Z, Xia P, ChenFu X, Shi A, Zhang J, Yu P. Non-coding RNAs in necroptosis, pyroptosis, and ferroptosis in cardiovascular diseases. Front Cardiovasc Med 2022; 9:909716. [PMID: 35990979 PMCID: PMC9386081 DOI: 10.3389/fcvm.2022.909716] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
Accumulating evidence has proved that non-coding RNAs (ncRNAs) play a critical role in the genetic programming and gene regulation of cardiovascular diseases (CVDs). Cardiovascular disease morbidity and mortality are rising and have become a primary public health issue that requires immediate resolution through effective intervention. Numerous studies have revealed that new types of cell death, such as pyroptosis, necroptosis, and ferroptosis, play critical cellular roles in CVD progression. It is worth noting that ncRNAs are critical novel regulators of cardiovascular risk factors and cell functions by mediating pyroptosis, necroptosis, and ferroptosis. Thus, ncRNAs can be regarded as promising therapeutic targets for treating and diagnosing cardiovascular diseases. Recently, there has been a surge of interest in the mediation of ncRNAs on three types of cell death in regulating tissue homeostasis and pathophysiological conditions in CVDs. Although our understanding of ncRNAs remains in its infancy, the studies reviewed here may provide important new insights into how ncRNAs interact with CVDs. This review summarizes what is known about the functions of ncRNAs in modulating cell death-associated CVDs and their role in CVDs, as well as their current limitations and future prospects.
Collapse
Affiliation(s)
- Yuxi Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiwen Zhou
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Panpan Xia
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of National Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Xinxi ChenFu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of National Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Ao Shi
- School of Medicine, University of Nicosia, Nicosia, Cyprus
- School of Medicine, St. George University of London, London, United Kingdom
| | - Jing Zhang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jing Zhang
| | - Peng Yu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- *Correspondence: Peng Yu
| |
Collapse
|
13
|
Leung JH, Ng B, Lim WW. Interleukin-11: A Potential Biomarker and Molecular Therapeutic Target in Non-Small Cell Lung Cancer. Cells 2022; 11:cells11142257. [PMID: 35883698 PMCID: PMC9318853 DOI: 10.3390/cells11142257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 85% of lung cancer and is a fast progressive disease when left untreated. Identification of potential biomarkers in NSCLC is an ongoing area of research that aims to detect, diagnose, and prognosticate patients early to optimize treatment. We review the role of interleukin-11 (IL11), a stromal-cell derived pleiotropic cytokine with profibrotic and cellular remodeling properties, as a potential biomarker in NSCLC. This review identifies the need for biomarkers in NSCLC, the potential sources of IL11, and summarizes the available information leveraging upon published literature, publicly available datasets, and online tools. We identify accumulating evidence suggesting IL11 to be a potential biomarker in NSCLC patients. Further in-depth studies into the pathophysiological effects of IL11 on stromal-tumor interaction in NSCLC are warranted and current available literature highlights the potential value of IL11 detection as a diagnostic and prognostic biomarker in NSCLC.
Collapse
Affiliation(s)
- Jason Hongting Leung
- Department of Cardiothoracic Surgery, National Heart Center Singapore, Singapore 169609, Singapore
- Correspondence:
| | - Benjamin Ng
- National Heart Research Institute Singapore, National Heart Center Singapore, Singapore 169609, Singapore; (B.N.); (W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169609, Singapore
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Center Singapore, Singapore 169609, Singapore; (B.N.); (W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169609, Singapore
| |
Collapse
|
14
|
Yin ZQ, Han H, Yan X, Zheng QJ. Research progress on the pathogenesis of aortic dissection. Curr Probl Cardiol 2022:101249. [PMID: 35568084 DOI: 10.1016/j.cpcardiol.2022.101249] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 12/20/2022]
Abstract
Aortic dissection (AD) is a critical cardiovascular disease due to the separation of media and adventitia caused by the rupture of vascular wall intima. The disease has a high mortality rate of about 1% to 3% for each additional hour, since the adventitia of the aorta can rupture and bleed to death at any time. Although great progress has been made in clinical treatment of aortic dissection, and the mortality rate has been significantly reduced, the pathogenesis is still not very clear. At present, related studies have confirmed that inflammation of aortic wall promotes the occurrence and development of AD. Although the mechanism of aortic dissection is more complicated, some studies have shown that the infiltration of monocytes/macrophages into the aortic wall is the main pathogenic mechanism of the disease. This review introduces the latest research results on the mechanism of macrophage infiltration and plasticity in aortic dissection.
Collapse
Affiliation(s)
- Zhi-Qiang Yin
- Department of Cardiovascular Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University); The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Xianchun Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shanxi, China
| | - Qi-Jun Zheng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University); The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
15
|
Feng Y, Ye D, Wang Z, Pan H, Lu X, Wang M, Xu Y, Yu J, Zhang J, Zhao M, Xu S, Pan W, Yin Z, Ye J, Wan J. The Role of Interleukin-6 Family Members in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:818890. [PMID: 35402550 PMCID: PMC8983865 DOI: 10.3389/fcvm.2022.818890] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is one of the main causes of human mortality. Cytokines play crucial roles in the development of cardiovascular disease. Interleukin (IL)-6 family members are a series of cytokines, including IL-6, IL-11, IL-30, IL-31, OSM, LIF, CNTF, CT-1, CT-2, and CLC, that regulate multiple biological effects. Experimental and clinical evidence shows that IL-6 family members are closely related to cardiovascular diseases such as atherosclerosis, hypertension, aortic dissection, cardiac fibrosis, and cardiomyopathy. This review mainly discusses the role of IL-6 family members in cardiovascular disease for the sake of identifying possible intervention targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Junping Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- Jing Ye
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Jun Wan
| |
Collapse
|
16
|
Corden B, Lim WW, Song W, Chen X, Ko NSJ, Su L, Tee NGZ, Adami E, Schafer S, Cook SA. Therapeutic Targeting of Interleukin-11 Signalling Reduces Pressure Overload-Induced Cardiac Fibrosis in Mice. J Cardiovasc Transl Res 2021; 14:222-228. [PMID: 32592090 DOI: 10.1007/s12265-020-10054-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/15/2020] [Indexed: 10/24/2022]
Abstract
There are currently no specific treatments for cardiac fibrosis. We tested the efficacy of a neutralising anti-IL11 antibody (X203) to reduce cardiac fibrosis in two preclinical models: transverse aortic constriction (TAC) and chronic angiotensin II infusion (AngII). In the first model, male C57BL/6J mice were subjected to TAC for 2 weeks. In the second model, mice received continuous angiotensin II for 4 weeks via subcutaneous pump. In both models, mice received either 20 mg/kg of X203 or isotype-control antibody twice-weekly, starting 24 h after surgery. Cardiac fibrosis and extracellular matrix gene expression were assessed by RT-qPCR, Western blot, histology and collagen (hydroxyproline) assays. In both models, X203 significantly reduced pro-fibrotic gene expression and myocardial fibrosis (TAC: 51% reduction in total collagen, P < 0.001, 39% in perivascular fibrosis, P < 0.001; AngII: 17% reduction in total collagen, P = 0.04, 83% in perivascular fibrosis, P < 0.001). Pharmacological targeting of IL11 reduces cardiac fibrosis in preclinical models. Figa Graphical Abstract.
Collapse
Affiliation(s)
- Ben Corden
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Weihua Song
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Xie Chen
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Nicole S J Ko
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Liping Su
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Nicole G Z Tee
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Eleonora Adami
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Sebastian Schafer
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Stuart A Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
17
|
Interleukin-11 is important for vascular smooth muscle phenotypic switching and aortic inflammation, fibrosis and remodeling in mouse models. Sci Rep 2020; 10:17853. [PMID: 33082445 PMCID: PMC7576123 DOI: 10.1038/s41598-020-74944-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
Transforming growth factor beta-1 (TGFβ1) is a major driver of vascular smooth muscle cell (VSMC) phenotypic switching, an important pathobiology in arterial disease.
We performed RNA-sequencing of TGFβ1-stimulated human aortic or arterial VSMCs which revealed large and consistent upregulation of Interleukin 11 (IL11). IL11 has an unknown function in VSMCs, which highly express the IL11 receptor alpha, suggestive of an autocrine loop. In vitro, IL11 activated ERK signaling, but inhibited STAT3 activity, and caused VSMC phenotypic switching to a similar extent as TGFβ1 or angiotensin II (ANGII) stimulation. Genetic or therapeutic inhibition of IL11 signaling reduced TGFβ1- or ANGII-induced VSMC phenotypic switching, placing IL11 activity downstream of these factors. Aortas of mice with Myh11-driven IL11 expression were remodeled and had reduced contractile but increased matrix and inflammatory genes expression. In two models of arterial pressure loading, IL11 was upregulated in the aorta and neutralizing IL11 antibodies reduced remodeling along with matrix and pro-inflammatory gene expression. These data show that IL11 plays an important role in VSMC phenotype switching, vascular inflammation and aortic pathobiology.
Collapse
|
18
|
Malecki C, Hambly BD, Jeremy RW, Robertson EN. The Role of Inflammation and Myeloperoxidase-Related Oxidative Stress in the Pathogenesis of Genetically Triggered Thoracic Aortic Aneurysms. Int J Mol Sci 2020; 21:ijms21207678. [PMID: 33081376 PMCID: PMC7590002 DOI: 10.3390/ijms21207678] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Genetically triggered thoracic aortic aneurysms (TAAs) are usually considered to exhibit minimal levels of inflammation. However, emerging data demonstrate that specific features of an inflammatory response can be observed in TAA, and that the extent of the inflammatory response can be correlated with the severity, in both mouse models and in human studies. Myeloperoxidase (MPO) is a key mediator of the inflammatory response, via production of specific oxidative species, e.g., the hypohalous acids. Specific tissue modifications, mediated by hypohalous acids, have been documented in multiple cardiovascular pathologies, including atherosclerosis associated with coronary artery disease, abdominal aortic, and cerebral aneurysms. Similarly, data are now emerging that show the capacity of MPO-derived oxidative species to regulate mechanisms important in TAA pathogenesis, including alterations in extracellular matrix homeostasis, activation of matrix metalloproteinases, induction of endothelial dysfunction and vascular smooth muscle cell phenotypic switching, and activation of ERK1/2 signaling. The weight of evidence supports a role for inflammation in exacerbating the severity of TAA progression, expanding our understanding of the pathogenesis of TAA, identifying potential biomarkers for early detection of TAA, monitoring severity and progression, and for defining potential novel therapeutic targets.
Collapse
Affiliation(s)
- Cassandra Malecki
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
- Correspondence:
| | - Brett D. Hambly
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
| | - Richmond W. Jeremy
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Elizabeth N. Robertson
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (B.D.H.); (R.W.J.); (E.N.R.)
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| |
Collapse
|
19
|
Zeng T, Gan J, Liu Y, Shi L, Lu Z, Xue Y, Xiong R, Liu L, Yang Z, Lin Y, Yuan J. ADAMTS-5 Decreases in Aortas and Plasma From Aortic Dissection Patients and Alleviates Angiotensin II-Induced Smooth Muscle-Cell Apoptosis. Front Cardiovasc Med 2020; 7:136. [PMID: 32923459 PMCID: PMC7456925 DOI: 10.3389/fcvm.2020.00136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/30/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Acute aortic dissection (AAD) is associated with degeneration of the aortic media and accompanied by vascular extracellular matrix (ECM) remodeling. Recently, a disintegrin and metalloproteinase with thrombospondin type 1 motifs-5 (ADAMTS-5) has been reported to be involved in ECM remodeling and vascular diseases. The aim of this study was to examine ADAMTS-5 levels in AAD patients and investigate the underlying mechanisms. Methods: Aortic tissue samples were collected from normal donors and AAD patients, and the expression of ADAMTS-5 was analyzed in all aortic tissues. In addition, plasma levels of ADAMTS-5, matrix metalloproteinase (MMP)-2 and MMP-9, and tumor necrosis factor-α (TNF-α) were measured in repeated samples from AAD patients and compared to the non-AAD (NAD) group. In addition, we investigated the effects of ADAMTS-5 in smooth muscle cell (SMC) apoptosis. Results: The results showed that ADAMTS-5 expression was significantly reduced in the aortas of AAD patients and that SMCs were the main source of ADAMTS-5. In addition, the plasma ADAMTS-5 level was lower, but plasma MMP-2, MMP-9, and TNF-α levels were increased in the AAD patients. Multivariate linear regression analyses showed that a decreased ADAMTS-5 level in patients was independently associated with an increased risk of AAD. Furthermore, recombinant human ADAMTS-5 significantly ameliorated angiotensin (Ang II)-evoked SMC apoptosis. Conclusions: ADAMTS-5 shows promise as a novel potential biomarker for AAD, and regulation of SMC is a possible mechanism for the effects of ADAMTS-5.
Collapse
Affiliation(s)
- Tao Zeng
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jianting Gan
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yu Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zhengde Lu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yan Xue
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Rixin Xiong
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 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
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jun Yuan
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
20
|
Predictors for the development of preoperative oxygenation impairment in acute aortic dissection in hypertensive patients. BMC Cardiovasc Disord 2020; 20:365. [PMID: 32778051 PMCID: PMC7416810 DOI: 10.1186/s12872-020-01652-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/04/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Acute aortic dissection (AAD) is an acute life-threatening cardiovascular disease, which is frequently complicated with oxygenation impairment (OI). We aim to investigate predictors of the development of OI in the patients with AAD. METHODS We retrospectively collected clinical data of AAD in hypertensive patients from July 2012 to March 2020. The patients included in this study were divided into OI (+) group (oxygenation index≤200) and OI (-) group (oxygenation index> 200). Both groups were compared according to demographic and clinical characteristics, and laboratory findings. Characteristics of hypertension in the patients with AAD were described. Predictors for the development of OI were assessed. And cutoff values were determined by receiver operating characteristics (ROC) curve. RESULTS A total of 208 patients were included in this study and the incidence of OI was 32.2%. In OI (+) group, patients had significantly higher peak body temperature (37.85 ± 0.60 vs 37.64 ± 0.44 °C, P = .005), higher levels of CRP (42.70 ± 28.27 vs 13.90 ± 18.70 mg/L, P = .000) and procalcitonin (1.07 ± 3.92 vs 0.31 ± 0.77μg/L, P = .027), and lower levels of albumin (34.21 ± 5.65 vs 37.73 ± 4.70 g/L, P = .000). Spearman's rank correlation test showed that the minimum oxygenation index was positively correlated with albumin, and was negatively correlated with the peak body temperature, serum CRP, procalcitonin, BNP and troponin. The stepwise multiple linear regression analysis showed that the peak body temperature, serum CRP and albumin were independently associated with development of OI. An optimal cutoff value for CRP for predicting OI was ≥9.20 mg/L, with a sensitivity of 91.0% and a specificity of 61.0%. CONCLUSIONS The peak body temperature, serum CRP and albumin were independent predictors of OI development in the patients with AAD. The serum CRP on admission≥9.20 mg/L might be a valuable and reliable indicator in predicting the development of OI.
Collapse
|
21
|
Corden B, Adami E, Sweeney M, Schafer S, Cook SA. IL-11 in cardiac and renal fibrosis: Late to the party but a central player. Br J Pharmacol 2020; 177:1695-1708. [PMID: 32022251 PMCID: PMC7070163 DOI: 10.1111/bph.15013] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is a pathophysiological hallmark of cardiorenal disease. In the heart, fibrosis leads to contractile dysfunction and arrhythmias; in the kidney, it is the final common pathway for many diseases and predicts end-stage renal failure. Despite this, there are currently no specific anti-fibrotic treatments available for cardiac or renal disease. Recently and unexpectedly, IL-11 was found to be of major importance for cardiorenal fibroblast activation and fibrosis. In mouse models, IL-11 overexpression caused fibrosis of the heart and kidney while genetic deletion of Il11ra1 protected against fibrosis and preserved organ function. Neutralizing antibodies against IL-11 or IL-11RA have been developed that have anti-fibrotic activity in human fibroblasts and protect against fibrosis in murine models of disease. While IL-11 biology has been little studied and, we suggest, largely misunderstood, its autocrine activity in myofibroblasts appears non-redundant for fibrosis, which offers new opportunities to better understand and potentially target cardiorenal fibrosis.
Collapse
Affiliation(s)
- Benjamin Corden
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingapore
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Eleonora Adami
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingapore
| | - Mark Sweeney
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Sebastian Schafer
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingapore
| | - Stuart A. Cook
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingapore
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|
22
|
Wang X, Zhang H, Cao L, He Y, Ma A, Guo W. The Role of Macrophages in Aortic Dissection. Front Physiol 2020; 11:54. [PMID: 32116765 PMCID: PMC7013038 DOI: 10.3389/fphys.2020.00054] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/21/2020] [Indexed: 12/21/2022] Open
Abstract
Aortic dissection (AD) is a fatal disease that accounts for a large proportion of aortic-related deaths and has an incidence of about 3–4 per 100,000 individuals every year. Recent studies have found that inflammation plays an important role in the development of AD, and that macrophages are the hub of inflammation in the aortic wall. Aortic samples from AD patients reveal a large amount of macrophage infiltration. The sites of macrophage infiltration and activity vary throughout the different stages of AD, with involvement even in the tissue repair phase of AD. Angiotensin II has been shown to be an important factor in the stimulation of macrophage activity. Stimulated macrophages can secrete metalloproteinases, inflammatory factors and other substances to cause matrix destruction, smooth muscle cell apoptosis, neovascularization and more, all of which destroy the aortic wall structure. At the same time, there are a number of factors that regulate macrophages to reduce the formation of AD and induce the repair of torn aortic tissues. The aim of this review is to take a close look at the roles of macrophages throughout the course of AD disease.
Collapse
Affiliation(s)
- Xinhao Wang
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongpeng Zhang
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Long Cao
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,Department of General Surgery, PLA No. 983 Hospital, Tianjin, China
| | - Yuan He
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Airong Ma
- Department of Obstetrics, Zibo Central Hospital, Zibo, China
| | - Wei Guo
- Department of Vascular and Endovascular Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
Interleukin-18 Expression Increases in the Aorta and Plasma of Patients with Acute Aortic Dissection. Mediators Inflamm 2019; 2019:8691294. [PMID: 31427887 PMCID: PMC6679882 DOI: 10.1155/2019/8691294] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
Background Interleukin- (IL-) 18 is a proinflammatory cytokine related to cardiovascular diseases, including hypertension and atherosclerosis. This study is aimed at determining whether IL-18 is related to aortic dissection (AD) and identifying the underlying mechanisms. Methods IL-18 expression in human aorta samples from AD (n = 8) and non-AD (NAD, n = 7) patients was measured. In addition, the IL-18, IL-6, interferon- (IFN-) γ, and IL-18-binding protein (IL-18BP) concentrations in plasma samples collected from the NAD and AD patients were detected. The effects of IL-18 on macrophage differentiation and smooth muscle cell (SMC) apoptosis were investigated in vitro. Results IL-18 expression was significantly increased in the aorta samples from the AD patients compared with those from the NAD patients, especially in the torn section. Aortic IL-18 was mainly derived from macrophages and also partly derived from CD4+ T lymphocytes and vascular SMCs. Plasma IL-18, IFN-γ, and IL-6 levels were significantly higher in the AD group than in the NAD group, and the IL-18 levels were positively correlated with the IFN-γ and IL-6 levels. In addition, plasma IL-18BP and free IL-18 levels were also elevated in the AD group. Linear regression analysis showed that the IL-18 level was independently associated with the presence of AD. In addition, anti-mouse IL-18-neutralizing monoclonal antibodies (anti-IL-18 nAb) inhibited angiotensin II-induced M1 macrophage differentiation and SMC apoptosis in vitro. Conclusion IL-18 may participate in AD by regulating macrophage differentiation and macrophage-induced SMC apoptosis.
Collapse
|
24
|
Biomarkers of Inflammation in Left Ventricular Diastolic Dysfunction. DISEASE MARKERS 2019; 2019:7583690. [PMID: 31275453 PMCID: PMC6589287 DOI: 10.1155/2019/7583690] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/21/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023]
Abstract
Left ventricular diastolic dysfunction (LVDD) is an important precursor to many different cardiovascular diseases. Diastolic abnormalities have been studied extensively in the past decade, and it has been confirmed that one of the mechanisms leading to heart failure is a chronic, low-grade inflammatory reaction. The triggers are classical cardiovascular risk factors, grouped under the name of metabolic syndrome (MetS), or other systemic diseases that have an inflammatory substrate such as chronic obstructive pulmonary disease. The triggers could induce myocardial apoptosis and reduce ventricular wall compliance through the release of cytokines by multiple pathways such as (1) immune reaction, (2) prolonged cell hypoxemia, or (3) excessive activation of neuroendocrine and autonomic nerve function disorder. The systemic proinflammatory state causes coronary microvascular endothelial inflammation which reduces nitric oxide bioavailability, cyclic guanosine monophosphate content, and protein kinase G (PKG) activity in adjacent cardiomyocytes favoring hypertrophy development and increases resting tension. So far, it has been found that inflammatory cytokines associated with the heart failure mechanism include TNF-α, IL-6, IL-8, IL-10, IL-1α, IL-1β, IL-2, TGF-β, and IFN-γ. Some of them could be used as diagnosis biomarkers. The present review aims at discussing the inflammatory mechanisms behind diastolic dysfunction and their triggering conditions, cytokines, and possible future inflammatory biomarkers useful for diagnosis.
Collapse
|
25
|
Abstract
Interleukin (IL)-11 belongs to the IL-6 family of cytokines, discovered over 30 years ago. While early studies focused on the ability of IL-11 to stimulate megakaryocytopoiesis, the importance of this cytokine to inflammatory disease and cancers is only just beginning to be uncovered. This review outlines recent advances in our understanding of IL-11 biology, and highlights the development of novel therapeutics with the potential for clinical targeting of signaling by this cytokine in multiple diseases.
Collapse
Affiliation(s)
- Paul M Nguyen
- a The Walter and Eliza Hall Institute of Medical Research , Victoria , Australia
- b The Department of Medical Biology, The University of Melbourne , Victoria , Australia
| | - Suad M Abdirahman
- a The Walter and Eliza Hall Institute of Medical Research , Victoria , Australia
- b The Department of Medical Biology, The University of Melbourne , Victoria , Australia
| | - Tracy L Putoczki
- a The Walter and Eliza Hall Institute of Medical Research , Victoria , Australia
- b The Department of Medical Biology, The University of Melbourne , Victoria , Australia
| |
Collapse
|
26
|
Ye J, Wang Z, Ye D, Wang Y, Wang M, Ji Q, Huang Y, Liu L, Shi Y, Shi L, Zeng T, Xu Y, Liu J, Jiang H, Lin Y, Wan J. Increased Interleukin-11 Levels Are Correlated with Cardiac Events in Patients with Chronic Heart Failure. Mediators Inflamm 2019; 2019:1575410. [PMID: 30728748 PMCID: PMC6341241 DOI: 10.1155/2019/1575410] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 11/01/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Interleukin-11 (IL-11) is an important inflammatory cytokine and has been demonstrated to participate in cardiovascular diseases. However, there have been no studies about the role of IL-11 in heart failure (HF). The present study is aimed at investigating whether IL-11 levels are associated with the cardiac prognosis in patients with HF. METHODS The plasma concentrations of IL-11 were measured in 240 patients with chronic HF (CHF) and 80 control subjects without signs of significant heart disease. In addition, we prospectively followed these CHF patients to endpoints of cardiac events. RESULTS Compared with the control group, the plasma IL-11 concentrations were significantly increased in the CHF patients and gradually increased in the New York Heart Association (NYHA) functional class II group, the NYHA functional class III group, and the NYHA functional class IV group. The receiver operating characteristic (ROC) curve revealed that the predictive role of IL-11 in HF is not as good as N-terminal B-type natriuretic peptide (BNP), although IL-11 has a certain value in predicting cardiac events. In addition, the CHF patients were divided into 3 groups according to the plasma IL-11 concentration category (low, T1; middle, T2; and high, T3). The multivariate Cox hazard analysis showed that the high plasma IL-11 concentrations were independently associated with the presence of cardiac events after adjustment for confounding factors. Furthermore, the CHF patients were divided into two groups based on the median plasma IL-11 concentrations. The Kaplan-Meier analysis revealed that the patients with high IL-11 concentrations had a higher risk of cardiac events compared with those with low IL-11 concentrations. CONCLUSIONS Higher plasma IL-11 levels significantly increase the presence of cardiac events and suggest a poor outcome; although the diagnostic value of IL-11 in CHF is not as good as BNP, there is a certain value in predicting cardiac events in CHF.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Huang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Tao Zeng
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Huimin Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yingzhong Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| |
Collapse
|
27
|
Ye J, Wang Y, Wang Z, Ji Q, Huang Y, Zeng T, Hu H, Ye D, Wan J, Lin Y. Circulating Th1, Th2, Th9, Th17, Th22, and Treg Levels in Aortic Dissection Patients. Mediators Inflamm 2018; 2018:5697149. [PMID: 30258282 PMCID: PMC6146596 DOI: 10.1155/2018/5697149] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 06/21/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Previous studies demonstrated that the subsets of CD4+ T helper (Th) cells are closely related to vascular diseases, including atherosclerosis and hypertension. This study is aimed at investigating the circulating Th1, Th2, Th9, Th17, Th22, and Treg levels in aortic dissection (AD) patients. METHODS Blood samples from AD (n = 56) and non-AD (NAD, n = 24) patients were collected, and the circulating levels of Th1, Th2, Th9, Th17, Th22, and Treg cells and their transcription factors and functional cytokines were measured by flow cytometric analysis, quantitative polymerase chain reaction, and enzyme-linked immunosorbent assays, respectively. In addition, the human aortic vascular smooth muscle cells (HASMCs) were treated with saline, angiotensin II (Ang II), or plasma from AD patients. RESULTS Compared with the levels in the NAD group, the Th1, Th9, Th17, Th22, and their transcription factor levels were increased and the Th2, Treg, and their transcription factor levels exhibited a decreasing trend in AD patients. In addition, higher IFN-γ, IL-9, IL-17, and IL-22 levels and lower IL-4 and IL-35 levels were observed in AD patients. Simple linear regression analysis and binary logistic regression analysis suggested that Th1/IFN-γ, IL-9, Th17/IL-17, and Th22/IL-22 positively regulated the occurrence of AD, while Th2/IL-4 and Treg/IL-35 negatively regulated the occurrence of AD. Plasma from AD patients further increased Bax mRNA levels but decreased Bcl2 and α-SMA mRNA levels in Ang II-treated HASMCs. CONCLUSIONS Changes in Th1, Th2, Th9, Th17, Th22, and Treg activity are associated with the onset of AD. Different subsets of CD4+ T cells play different roles in the presence of AD.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Emergency & Critical Care Center, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yuan Wang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Zhen Wang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Emergency & Critical Care Center, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Emergency & Critical Care Center, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ying Huang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Department of Ultrasound, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Tao Zeng
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Haiying Hu
- Department of Cardiology, Handan First Hospital, Handan 056002, China
| | - Di Ye
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Jun Wan
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| |
Collapse
|
28
|
Zeng T, Shi L, Ji Q, Shi Y, Huang Y, Liu Y, Gan J, Yuan J, Lu Z, Xue Y, Hu H, Liu L, Lin Y. Cytokines in aortic dissection. Clin Chim Acta 2018; 486:177-182. [PMID: 30086263 DOI: 10.1016/j.cca.2018.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/03/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023]
Abstract
Aortic dissection (AD) is one of the most dangerous forms of vascular disease, characterized by endometrial rupture and intramural hematoma formation. Generally, the pathological process is complicated and closely related to the infiltration of inflammatory cells into the aortic wall and apoptosis of vascular smooth muscle cells. Currently, multiple cytokines, including interleukins, interferon, the tumor necrosis factor superfamily, colony stimulating factor, chemotactic factor, growth factor and so on, have all been demonstrated to play a critical role in AD. Additionally, studies of the link between cytokines and AD could deepen our understanding of the disease and may guide future treatment therapies; therefore, this review focuses on the role of cytokines in AD.
Collapse
Affiliation(s)
- Tao Zeng
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Shi
- 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; Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing 100029, China
| | - Ying Shi
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Huang
- Department of Ultrasound, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Yu Liu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jianting Gan
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jun Yuan
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zhengde Lu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yan Xue
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Haiying Hu
- Department of Cardiology, Handan First Hospital, Handan 056002, China
| | - Ling Liu
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| | - Yingzhong Lin
- Department of Cardiology, the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| |
Collapse
|