1
|
Zhang S, Qian L, Li S, Liu Z. Association between soluble suppression of tumorigenicity 2 and risk and severity of coronary artery disease: a case control study. BMC Cardiovasc Disord 2025; 25:334. [PMID: 40295953 PMCID: PMC12038991 DOI: 10.1186/s12872-025-04787-5] [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: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND To investigate the differential expression of soluble suppression of tumorigenicity 2 (sST2) in patients with coronary artery disease (CAD) and healthy controls, and the correlation between sST2 and the severity of coronary artery atherosclerosis. METHODS A total of 911 CAD patients were selected as the CAD group, and 322 healthy people were selected as the control group. We measured serum sST2 level by chemiluminescence immunoassay, and applied the Gensini scoring system to quantify the severity of coronary artery atherosclerosis. We utilized Mann-Whitney U test to assess the difference of sST2 level between the two groups, and adopted Spearman correlation test to evaluate the correlation between sST2 level and Gensini score and inflammatory indexes. RESULTS Compared with the control group, the expression level of sST2 in CAD group was significantly increased [29.20 (20.67, 46.34) vs. 19.69 (15.97, 25.02), P < 0.001]. Logistic regression showed that sST2 expression could increase CAD risk (OR = 1.099, 95%CI: 1.080 ~ 1.119, P < 0.001). Analysis of variance revealed that the sST2 expression level increased gradually in unstable angina pectoris group (UA), non-ST elevation myocardial infarction group (NSTEMI) and ST elevation myocardial infarction group (STEMI) [UA: 23.05 (17.54, 30.75), NSTEMI: 30.71 (21.31, 42.97), STEMI: 51.05 (32.85, 80.04), P < 0.001]. Spearman correlation analysis demonstrated significantly positive associations between sST2 expression level and Gensini score (r = 0.137, P < 0.001), and systemic inflammatory indexes MHR (r = 0.188, P < 0.001), NLR (r = 0.469, P < 0.001), PLR (r = 0.285, P < 0.001) and MLR (r = 0.368, P < 0.001), but negatively correlated with AFR (r=-0.135, P < 0.001). By receiver operating characteristic (ROC) curve analysis, the sST2 expression level had excellent predictive effect in STEMI with the area under the curve (AUC) value of 0.926 (95%CI: 0.903-0.948, P < 0.001) and sensitivity and specificity of 72.3% and 99.7% respectively, superior to NSTEMI with an AUC of 0.760 (95%CI: 0.719-0.802, P < 0.001) and UA with an AUC of 0.616 (95%CI: 0.576-0.656, P < 0.001). CONCLUSIONS sST2 could not only serve as a biomarker for the clinical auxiliary diagnosis of CAD, but also act as a potential indicator for disease progression or risk stratification. Dynamic monitoring of sST2 levels might assist in evaluating treatment efficacy.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, Jiangsu, China
| | - Lu Qian
- Xuzhou Blood Center, Xuzhou, 221002, Jiangsu, China
| | - Shibao Li
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, Jiangsu, China
| | - Zhijian Liu
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, No.99 Huaihai West Road, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
2
|
Pramanda AN, Farabi F, Prameswari HS, Achmad C, Tiksnadi BB. Myocardial-alternation index (MMI) is correlated with soluble suppression of tumorigenecity-2 (sST2) in patients with ischemic cardiomyopathy. Egypt Heart J 2025; 77:39. [PMID: 40261549 PMCID: PMC12014887 DOI: 10.1186/s43044-025-00634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/23/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Ischemic cardiomyopathy is a condition that represents myocardial dysfunction due to obstructive coronary artery disease. In ischemic cardiomyopathy, both structural and electrical remodeling occur. Myocardial biomarker, soluble ST2 (sST2) is able to predict patient's mortality and morbidity, and structural remodeling of the heart is responsible for its expression. ECG dispersion mapping (ECG-DM) as evaluated by myocardial micro-alternation index (MMI) may predict alteration of the myocardial electrophysiology with high sensitivity and specificity. The association between structural and electrical remodeling in ischemic cardiomyopathy is not fully understood. This study aims to evaluate the correlation between MMI and sST2 level in patients with ischemic cardiomyopathy. RESULT Total patients who met for the inclusion criteria were 30 patients. Mean age was 57.97 ± 10.04 years; most patients were male (80%). 27 (90%) patients had class II NYHA functional class. The most common risk factors were smoking (20 (66,7%)) and hypertension (17 (56,7%)). Median MMI was 34.0% (IQR: 23.0-42.3%) and median sST2 was 5.6 ng/mL (IQR: 2.0-11.5 ng/mL). This study found that MMI had a significant correlation with sST2, indicating a link between structural and electrical remodeling in ischemic cardiomyopathy (r = 0.583, p < 0,05). CONCLUSION There was a correlation between MMI and sST2 in patients with ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Andra Naufal Pramanda
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Fatih Farabi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Hawani Sasmaya Prameswari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Chaerul Achmad
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Badai Bhatara Tiksnadi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia.
| |
Collapse
|
3
|
Weng D, Shi W, Hu Y, Su Y, Li A, Wei S, Guo S. Neutralization of IL-33 ameliorates septic myocardial injury through anti-inflammatory, anti-oxidative, and anti-apoptotic by regulating the NF-κB/STAT3/SOCS3 signaling pathway. Biochem Pharmacol 2025; 237:116954. [PMID: 40258576 DOI: 10.1016/j.bcp.2025.116954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/18/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Septic myocardial injury, a severe sepsis complication linked to high morbidity and mortality, remains a major global clinical challenge. Interleukin-33 (IL-33), a damage-associated pro-inflammatory factor, has been implicated in regulating immune responses and inflammation, but its specific role in septic myocardial injury has not been fully elucidated. This study examined IL-33's role in septic myocardial injury using Gene Expression Omnibus (GEO) database datasets, alongside in vitro and in vivo experiments. Our results indicated a significant upregulation of IL-33 in septic myocardial injury, as demonstrated in both clinical and experimental settings. Blocking IL-33 significantly enhanced cardiac function and alleviated cardiomyocyte damage. Mechanistic investigations revealed that neutralizing IL-33 mitigates inflammation, oxidative stress, and apoptosis in cardiomyocytes by regulating the nuclear factor kappa B (NF-κB)/signal transducer and activator of transcription 3 (STAT3)/suppressors of cytokine signaling 3 (SOCS3) signaling pathway. Peritoneal macrophages are recognized as a potential origin of IL-33, and targeting IL-33 derived from these cells further reduced cardiomyocyte injury. The study underscores IL-33's crucial involvement in septic myocardial injury pathogenesis, indicating that IL-33 may serve as a promising therapeutic target.
Collapse
Affiliation(s)
- Danlei Weng
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Wei Shi
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Yue Hu
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Yanqian Su
- Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Andong Li
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Shuxing Wei
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Worker's Stadium South Road, Beijing 100020, China.
| |
Collapse
|
4
|
Song M, Dai H, Zhou Q, Meng X. The immunology of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2025; 16:1542208. [PMID: 40260277 PMCID: PMC12009709 DOI: 10.3389/fendo.2025.1542208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Diabetic cardiomyopathy is a notable microvascular complication of diabetes, characterized primarily by myocardial fibrosis and functional abnormalities. Long-term hyperglycemia induces excessive activation and recruitment of immune cells and triggers the cascade of inflammatory responses, resulting in systemic and local cardiac inflammation. Emerging evidence highlights the significant roles of immunology in modulating the pathology of diabetic cardiomyopathy. As the primary effectors of inflammatory reactions, immune cells are consistently present in cardiac tissue and can be recruited under pathological hyperglycemia circumstances. A disproportionate favor to proinflammatory types of immune cells and the increased proinflammatory cytokine levels mediate fibroblast proliferation, phenotypic transformation, and collagen synthesis and ultimately rise to cardiac fibrosis and hypertrophy. Meanwhile, the severity of cardiac fibrosis is also strongly associated with the diverse phenotypes and phenotypic alterations of the immune cells, including macrophages, dendritic cells, mast cells, neutrophils, and natural killer cells in innate immunity and CD4+ T lymphocytes, CD8+ T lymphocytes, and B lymphocytes in adaptive immunity. In this review, we synthesized the current analysis of the critical role played by the immune system and its components in the progression of diabetic cardiomyopathy. Finally, we highlight preclinical and clinical immune targeting strategies and translational implications.
Collapse
Affiliation(s)
| | | | | | - Xiao Meng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
5
|
Somuncu MU, Güdül NE, Köktürk U, Köksal BG, Tatar FP, Avci A. Relationship between sST2 and NT-proBNP levels and postoperative atrial fibrillation in patients having non-cardiac surgery. Minerva Cardiol Angiol 2025; 73:192-200. [PMID: 39535528 DOI: 10.23736/s2724-5683.24.06649-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND We explored the link between sST2 and NT-proBNP levels and postoperative atrial fibrillation (POAF) incidence in non-cardiac surgery patients in this study. METHODS The research involved 302 participants over 40 years old who underwent medium and/or high-risk non-cardiac surgeries. These patients were divided into two groups: those who developed POAF and those who did not. RESULTS The study cohort consisted of a total of 302 patients, with 14 (4.6%) experiencing POAF. POAF was more common in patients with previous heart failure, a high Left Atrial Volume Index (LAVI), and elevated ASA and RCRI scores (all P<0.05). LAVI, sST2, NT-proBNP, and RCRI scores were found to be independent predictors of POAF in patients undergoing non-cardiac surgeries (all P<0.05). The area under the curve (AUC) for sST2 and NT-proBNP in predicting POAF was 0.707 (95% CI 0.544-0.869; P=0.009) and 0.727 (95% CI 0.598-0.857; P=0.004), respectively. Combined elevation of sST2 and NT-proBNP increased the likelihood of developing POAF by approximately 8.5 times (OR: 8.65, CI 95% 1.06-35.3, P=0.044). CONCLUSIONS sST2 and NT-proBNP are valuable predictors of POAF in patients undergoing non-cardiac surgery. Identifying these predictors can help in recognizing high-risk patient groups for POAF.
Collapse
Affiliation(s)
- Mustafa U Somuncu
- Department of Cardiology, Faculty of Medicine, Aydın Adnan Menderes University, Aydın, Türkiye
| | - Naile E Güdül
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Türkiye
| | - Uğur Köktürk
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Türkiye -
| | - Bengü G Köksal
- Department of Anesthesia and Reanimation, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Türkiye
| | - Fatih P Tatar
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Türkiye
| | - Ahmet Avci
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Türkiye
| |
Collapse
|
6
|
Wang J, Li S, Zhou X, Wu H, Ouyang X, Huang Z, Peng L, Chen Q, Wu Y, Li Z, Peng Z, Yang Y, Lu Y, Tang X, Li Y, Li S. Mucosal-associated invariant T cells correlate with myocardial ischaemia and remodelling in coronary artery disease. Clin Transl Immunology 2025; 14:e70029. [PMID: 40130221 PMCID: PMC11931450 DOI: 10.1002/cti2.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/27/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Objectives Myocardial ischaemia and remodelling are major contributors to the progression and mortality of coronary artery disease (CAD). Previous studies have shown immune cell alterations in CAD patients, but their characteristics and associations with myocardial ischaemia and remodelling remain unclear. Methods We compared immune cell changes among patients without CAD, those with CAD and those with CAD and heart failure (HF). Results We found a progressive reduction in circulating mucosal-associated invariant T (MAIT) cells across the three patient groups. MAIT cells exhibited increased expression of activation markers (CD69 and PD-1) and cytotoxic molecules (such as granzyme B). The features of MAIT cells were correlated positively with worsening clinical indicators of myocardial ischaemia and remodelling, including the Gensini score, cTnI, NT-proBNP, LVEF and E/e'. Additionally, the reduction, activation and cytotoxicity of MAIT cells were associated with indicators of myocardial fibrosis (sST2, Gal-3, PICP and PIIINP), a central pathological mechanism of myocardial remodelling. Finally, we preliminarily explored potential triggers for MAIT cell abnormalities in CAD patients and found that impaired intestinal barrier function and increased circulating bacterial antigens may contribute to these changes. Conclusions During CAD progression, we observed a decrease in circulating MAIT cells. Enhanced activation and cytotoxicity of MAIT cells are associated with myocardial ischaemia and remodelling in CAD patients with heart failure, potentially triggered by gut microbial leakage. Our findings suggest a novel strategy for monitoring and intervention in disease progression.
Collapse
Affiliation(s)
- Jiafu Wang
- Department of Cardiovascular MedicineThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Song Li
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xianling Zhou
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Hongxing Wu
- Department of Cardiovascular MedicineThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xiaolan Ouyang
- Department of Cardiovascular MedicineThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Zhuoshan Huang
- Department of Cardiovascular MedicineThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Long Peng
- Department of Cardiovascular MedicineThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Qian Chen
- School of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
| | - Yuman Wu
- Department of Clinical ImmunologyThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Zhitong Li
- Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhou Medical UniversityGuangzhouChina
- The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Ziyi Peng
- Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhou Medical UniversityGuangzhouChina
- The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Yi Yang
- Department of Endocrinology and Metabolic DiseasesThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Yan Lu
- Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhou Medical UniversityGuangzhouChina
- The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Xixiang Tang
- VIP Medical Service CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Yue Li
- Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhou Medical UniversityGuangzhouChina
- The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Suhua Li
- Department of Cardiovascular MedicineThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
7
|
Lotfinaghsh A, Imam A, Pompian A, Stitziel NO, Javaheri A. Clinical Insights from Proteomics in Heart Failure. Curr Heart Fail Rep 2025; 22:12. [PMID: 40063168 DOI: 10.1007/s11897-025-00698-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 05/13/2025]
Abstract
PURPOSE OF REVIEW The pathophysiology of heart failure (HF), a complex and heterogenous condition, remains to be fully understood. Troponin and b-type natriuretic peptide are the only biomarkers that are utilized in clinical practice for HF clinical management. Recent advances in proteomics present a powerful tool to identify risk markers and ultimately, potential molecular mechanisms underlying HF pathogenesis. Herein, we explore traditional and novel heart biomarkers, highlighting their potential role in the pathogenesis of HF. RECENT FINDINGS Recent proteomic analyses have identified numerous proteins including Galectin-3, sST2, GDF-15, FGF21, Endotrophin, THSB-2, ADAMSTL, SVEP1, and anthracycline that are associated with clinical outcomes in HF. These biomarkers are not presently utilized in HF management but may be useful in the future for prediction of death or HF hospitalization. While traditional biomarkers remain essential, proteomic strategies have revealed additional targets that require further mechanistic exploration. Future research should focus on validating these biomarkers and translating proteomic insights into clinical practice to enhance HF management.
Collapse
Affiliation(s)
- Aynaz Lotfinaghsh
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Adnan Imam
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander Pompian
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nathan O Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ali Javaheri
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- John Cochran VA Hospital, St Louis, MO, USA.
| |
Collapse
|
8
|
Kim JA, Lee JE, Bae K, Ahn SS. Elevated Soluble Suppressor of Tumorigenicity 2 Levels in Gout Patients and Its Association with Cardiovascular Disease Risk Indicators. Yonsei Med J 2025; 66:151-159. [PMID: 39999990 PMCID: PMC11865869 DOI: 10.3349/ymj.2024.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/21/2024] [Accepted: 08/16/2024] [Indexed: 02/27/2025] Open
Abstract
PURPOSE To investigate the association between soluble suppressor of tumorigenicity 2 (sST2) levels and cardiovascular disease predictors in patients with gout. MATERIALS AND METHODS We retrospectively reviewed the medical records of patients with gout who were tested for sST2 but did not receive uric acid-lowering therapy. These patients were classified into elevated and normal sST2 groups using a cut-off of >49.6 ng/mL and >35.4 ng/mL in males and females, respectively. Correlations between clinical and laboratory variables, sST2 levels, and elevated sST2 level predictors were assessed using linear and logistic regression analyses. RESULTS Notably, 27 (11.3%) and 211 (88.7%) of the 238 identified patients had elevated and normal sST2 levels, respectively. Linear regression analysis revealed that male sex (β=-0.190, p=0.002), body mass index (BMI) (β=-0.184, p=0.002), white blood cell count (β=0.231, p<0.001), C-reactive protein (β=0.135, p=0.031), and fasting blood glucose (β=0.210, p<0.001) were independently associated with sST2 levels. In multivariate logistic regression analysis, male sex [odds ratio (OR) 0.112, p=0.001], BMI (OR 0.836, p=0.008), creatinine (OR 5.730, p=0.024), and fasting blood glucose (OR 1.042, p=0.002) predicted elevated sST2 levels. Patients with increased sST2 levels had a significantly higher atherosclerotic cardiovascular disease risk score and a greater proportion of high-risk Framingham Risk Score compared to the normal sST2 group (p=0.002 and p<0.001). CONCLUSION Patients with gout and elevated sST2 levels have a higher risk of future cardiovascular disorders, which may provide insights into risk stratification and the implementation of intervention strategies.
Collapse
Affiliation(s)
- Jiyoung Agatha Kim
- Department of Emergency Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Eun Lee
- Graduate School, Yonsei University College of Medicine, Seoul, Korea
| | - Kunhyung Bae
- Department of Orthopaedic Surgery, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Korea.
| | - Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea.
| |
Collapse
|
9
|
Zhang R, Cai Q, Shao D, Luo Q, Zhang Z. Recurrent atrial fibrillation markers post radiofrequency catheter ablation. Clin Chim Acta 2025; 568:120126. [PMID: 39798686 DOI: 10.1016/j.cca.2025.120126] [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: 11/23/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Atrial fibrillation (AF), the most common type of heart arrhythmia, is recognized as an independent risk factor for stroke. Fortunately, catheter ablation (CA) offers an effective treatment option for AF patients. However, numerous studies have reported suboptimal outcomes, as AF recurrence rates often remain elevated even after CA. Consequently, there exists a need for early identification of patients prone to recurrence, necessitating anti-inflammatory and/or antiarrhythmic treatment post-CA. The discovery and application of markers associated with AF recurrence could significantly aid in this early identification process. In this review, we present an overview of AF recurrence markers from three distinct perspectives (biochemical, imaging, and electrocardiographic markers).
Collapse
Affiliation(s)
- Rangrang Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang 830001, China.
| | - Qingyuan Cai
- Department of Cardiology, the First Hospital of Jilin University, Changchun, Jilin Province 130021, China.
| | - Dongpu Shao
- Department of Cardiology, the First Hospital of Jilin University, Changchun, Jilin Province 130021, China.
| | - Qin Luo
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region, Hypertension Research Laboratory, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, Xinjiang 830001, China.
| | - Zhiguo Zhang
- Department of Cardiology, the First Hospital of Jilin University, Changchun, Jilin Province 130021, China.
| |
Collapse
|
10
|
van Genuchten WJ, Averesch H, van Dieren QM, Bonnet D, Odermarsky M, Beghetti M, Roos-Hesselink JW, Reinhardt Z, Male C, Naumburg E, Boersma E, De Wolf D, Helbing WA. Clinical impact of circulating biomarkers in prediction of adverse cardiac events in patients with congenital heart disease. A systematic review. Int J Cardiol 2025; 421:132723. [PMID: 39532255 DOI: 10.1016/j.ijcard.2024.132723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Patients with congenital heart disease (ConHD) are at increased risk for adverse cardiac events. Predicting long-term outcomes and guidance of patient management might benefit from a range of (new) biomarkers. This is a rapidly evolving field with potentially large consequences for clinical decision making. With a systematic review of available biomarkers in ConHD we identified the clinical role of these markers, knowledge gaps and future research directions. METHODS We systematically reviewed the literature on associations between blood biomarkers and outcome measures (mortality or composite adverse outcomes in patients with ConHD. RESULTS The inclusion criteria were met by 102 articles. Biomarkers assessed in more than 3 studies are discussed in the main text, those studied in 3 or less studies are summarized in the supplement. Thus, we discuss 15 biomarkers from 92 studies. These biomarkers were studied in 32,399 / 10,735 patients for the association with mortality and composite adverse outcomes, respectively. Biomarkers that were studied most and had statistically significant associations with mortality or composite adverse outcomes were (NT-pro)BNP, MELD-XI score, Hs-CRP, creatinine, albumin and sodium. Most of these biomarkers are involved in intracardiac processes associated with inflammation or are markers of renal function. CONCLUSION For (NT-pro)BNP, clinical value for prediction of mortality and composite adverse outcomes in adult and paediatric ConHD has been shown. For MELD-XI, hs-CRP, albumin, creatinine, sodium, RDW, and GDF-15, correlations with mortality and composite adverse outcomes have been demonstrated in patient groups with mixed types of ConHD, but clinical utility needs additional exploration.
Collapse
Affiliation(s)
- W J van Genuchten
- Department of Paediatrics, Division of Paediatric Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - H Averesch
- Department of Paediatrics, Division of Paediatric Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Q M van Dieren
- Department of Paediatrics, Division of Paediatric Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - D Bonnet
- Hôpital Necker Enfants Malades, AP-HP, Université Paris Descartes, Paris, France; Cardiology Expert Group of the connect4children (c4c) network
| | - M Odermarsky
- Department of Paediatric Cardiology Paediatric Heart Center Lund University and Skåne University Hospital Lund, Sweden; Cardiology Expert Group of the connect4children (c4c) network
| | - M Beghetti
- Paediatric Cardiology Unit, Department of the Child and Adolescent, Children's University Hospital Geneva, Geneva, Switzerland; Cardiology Expert Group of the connect4children (c4c) network
| | - J W Roos-Hesselink
- Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Z Reinhardt
- Department of Paediatric Cardiology and Transplantation, Freeman Hospital, Newcastle upon Tyne, United Kingdom; Cardiology Expert Group of the connect4children (c4c) network
| | - C Male
- Department of Paediatrics, Medical University of Vienna, Vienna, Austria; Cardiology Expert Group of the connect4children (c4c) network
| | - E Naumburg
- Department of Clinical Sciences, Paediatrics, Umeå University, Umeå, Sweden; Cardiology Expert Group of the connect4children (c4c) network
| | - E Boersma
- Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - D De Wolf
- Departments of Paediatric Cardiology of Ghent and Brussels University Hospitals, Belgium; Cardiology Expert Group of the connect4children (c4c) network
| | - W A Helbing
- Department of Paediatrics, Division of Paediatric Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Cardiology Expert Group of the connect4children (c4c) network.
| |
Collapse
|
11
|
Miura-Takahashi E, Tsudome R, Suematsu Y, Tachibana T, Kato Y, Kuwano T, Sugihara M, Tashiro K, Shiga Y, Kamimura H, Miura SI. An elevated level of soluble suppression of tumorigenicity 2, but not galectin-3, is associated with the presence of coronary artery disease in hypertensive patients. Hypertens Res 2025; 48:650-661. [PMID: 39394514 DOI: 10.1038/s41440-024-01934-x] [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: 06/30/2024] [Revised: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 10/13/2024]
Abstract
We investigated whether there were associations between coronary artery disease (CAD) and soluble suppression of tumorigenicity (sST2) and galectin-3 levels at the time of coronary artery computed tomography angiography (CCTA) for CAD screening. The subjects consisted of 429 patients who underwent CCTA examination. CAD was diagnosed when there was 50% or more stenosis in the coronary artery. Patient backgrounds were collected and plasma levels of sST2 and galectin-3 were measured. The presence or absence of CAD and factors that contributed to CAD were analyzed for all patients and for those with or without hypertension (HTN). The CAD group had significantly higher sST2 levels than the non-CAD group, whereas there was no significant difference in galectin-3 levels. The number of patients in the non-HTN and HTN groups was 174 and 255, respectively. In the HTN group, the CAD group was significantly older than the non-CAD group and had higher sST2 levels. Multivariate analysis showed that the factors that contributed to CAD in the HTN group were age and sST2 levels. On the other hand, in the non-HTN group, the CAD group was significantly older than the non-CAD group, and had a higher proportion of males and higher sST2 levels, while the contributing factors for the CAD group were age and male gender, but not sST2. In conclusion, a higher level of sST2, but not galectin-3, was a contributing factor for CAD in HTN patients. However, in non-HTN patients, a high level of sST2 was not a contributing factor for CAD.
Collapse
Affiliation(s)
| | - Riku Tsudome
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yasunori Suematsu
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Tetsuro Tachibana
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yuta Kato
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Takashi Kuwano
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Makoto Sugihara
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Kokei Tashiro
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yuhei Shiga
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | | | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan.
| |
Collapse
|
12
|
Ikeda S. Is soluble ST2 an useful biomarker for early diagnosis of coronary atherosclerosis? Hypertens Res 2025; 48:839-841. [PMID: 39537983 DOI: 10.1038/s41440-024-01987-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Shuntaro Ikeda
- Department of Heart Failure Management, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan.
| |
Collapse
|
13
|
He Y, Shen X, Zhai K, Nian S. Advances in understanding the role of interleukins in pulmonary fibrosis (Review). Exp Ther Med 2025; 29:25. [PMID: 39650776 PMCID: PMC11619568 DOI: 10.3892/etm.2024.12775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive, irreversible disease characterized by heterogeneous interstitial lung tissue damage. It originates from persistent or repeated lung epithelial injury and leads to the activation and differentiation of fibroblasts into myofibroblasts. Interleukins (ILs) are a group of lymphokines crucial for immunomodulation that are implicated in the pathogenesis of PF. However, different types of ILs exert disparate effects on PF. In the present review, based on the effect on PF, ILs are classified into three categories: i) Promotors of PF; ii) inhibitors of PF; and iii) those that exert dual effects on PF. Several types of ILs can promote PF by provoking inflammation, initiating proliferation and transdifferentiation of epithelial cells, exacerbating lung injury, while other ILs can inhibit PF through suppressing expression of inflammatory factors, modulating the Th1/Th2 balance and autophagy. The present review summarizes the association of ILs and PF, focusing on the roles and mechanisms of ILs underlying PF.
Collapse
Affiliation(s)
- Yuqing He
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xuebin Shen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Kefeng Zhai
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, P.R. China
| | - Sihui Nian
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
14
|
Huang S, Yu LJ, Sun GF, Zhang ZX. Short-term predictive value of sST2 in patients with STEMI following primary PCI: a prospective observational study. BMC Cardiovasc Disord 2025; 25:21. [PMID: 39819309 PMCID: PMC11737151 DOI: 10.1186/s12872-025-04488-z] [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: 06/18/2023] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
AIM The objective of this study was to investigate the level of soluble suppression of tumorigenicity-2 (sST2) in patients with acute ST-segment elevation myocardial infarction (STEMI) following primary percutaneous coronary intervention (PCI), and to provide a new biomarker for clinical management and prognosis assessment. METHOD This was a prospective study. 148 STEMI patients following primary PCI were enrolled and divided into 2 groups by the median value of sST2 and afterwards followed up for 30 days to access the occurrence of major adverse cardiac events (MACEs), which were defined as cardiovascular death, heart failure and recurrent MI. RESULTS sST2 ranged from 20.57 to 98.96 ng/mL. High sST2 group had higher MACEs rate compared to low sST2 group (28.8% vs. 8.0%, P = 0.001). sST2 was positively correlated with age (r = 0.181, P = 0.027), SYNTAX score (r = 0.257, P = 0.002), high-sensitive C-reactive protein (hs-CRP) (r = 0.225, P = 0.006), B-type natriuretic peptide (BNP) (r = 0.225, P = 0.006) and negatively with left ventricle ejection fraction (LVEF) (r = -0.197, P = 0.016). After adjustment for clinical variables, sST2 level (OR 3.680, P = 0.015) and LVEF (OR 0.880, P < 0.001) remained independent predictors of 30-days MACEs. In receiver operating characteristic curve (ROC) analyses, the area under the curve (AUC) of sST2 for predicting 30-days MACEs was 0.755(P < 0.001). The AUC of sST2 combining hs-CRP and LVEF for prediction was 0.828(95%CI [0.743-0.912], P < 0.001). CONCLUSION sST2 level after primary PCI was an independent risk factor of MACEs in STEMI patients through 30 days follow-up.
Collapse
Affiliation(s)
- Shan Huang
- Department of Cardiology, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, No 2999 Jinshan Road, Huli District, Xiamen, China
| | - Lu-Jiao Yu
- Department of Geriatrics, the First Hospital of China Medical University, No.155 North Nanjing Street, Heping District, Shenyang, China
| | - Guang-Feng Sun
- Department of Emergency, School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, No 2999 Jinshan Road, Xiamen, China
| | - Zi-Xin Zhang
- Department of Cardiology, the First Hospital of China Medical University, No.155 North Nanjing Street, Heping District, Shenyang, China.
| |
Collapse
|
15
|
Yao J, Zhang Y, Wang Z, Chen Y, Shi X. Maintenance of Cardiac Microenvironmental Homeostasis: A Joint Battle of Multiple Cells. J Cell Physiol 2025; 240:e31496. [PMID: 39632594 DOI: 10.1002/jcp.31496] [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: 07/02/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Various cells such as cardiomyocytes, fibroblasts and endothelial cells constitute integral components of cardiac tissue. The health and stability of cardiac ecosystem are ensured by the action of a certain type of cell and the intricate interactions between multiple cell types. The dysfunctional cells exert a profound impact on the development of cardiovascular diseases by involving in the pathological process. In this paper, we introduce the dynamic activity, cell surface markers as well as biological function of the various cells in the heart. Besides, we discuss the multiple signaling pathways involved in the cardiac injury including Hippo/YAP, TGF-β/Smads, PI3K/Akt, and MAPK signaling. The complexity of different cell types poses a great challenge to the disease treatment. By characterizing the roles of various cell types in cardiovascular diseases, we sought to discuss the potential strategies for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Youtao Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ziwen Wang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| |
Collapse
|
16
|
Kotas ME, Gordon ED. Innovation through imitation: IL-33 decoys show promise in pulmonary fibrosis. J Pharmacol Exp Ther 2025; 392:100035. [PMID: 39893003 DOI: 10.1016/j.jpet.2024.100035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 02/04/2025] Open
Affiliation(s)
- Maya E Kotas
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Erin D Gordon
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
17
|
Trivedi MV, Jadhav HR, Gaikwad AB. Novel therapeutic targets for cardiorenal syndrome. Drug Discov Today 2025; 30:104285. [PMID: 39761847 DOI: 10.1016/j.drudis.2024.104285] [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: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/31/2024] [Indexed: 01/14/2025]
Abstract
Cardiorenal syndrome (CRS) is an interdependent dysfunction of the heart and kidneys, where failure in one organ precipitates failure in the other. The pathophysiology involves sustained renin-angiotensin-aldosterone-system (RAAS) activation, mitochondrial dysfunction, inflammation, fibrosis, oxidative stress and tissue remodeling, culminating in organ dysfunction. Existing therapies targeting the RAAS, diuretics and other agents have limitations, including diuretic resistance and compensatory sodium reabsorption. Therefore, there is a pressing need for novel druggable targets involved in CRS pathogenesis. This review addresses the challenges of existing treatments and emphasizes the importance of discovering new therapeutic targets. It highlights emerging targets such as Klotho, sex-determining region Y box 9 (SOX9), receptor-interacting protein kinase 3 (RIPK3), β-amino-isobutyric acid (BAIBA), thrombospondin-1 (TSP-1), among others, with their potential roles in CRS.
Collapse
Affiliation(s)
- Mansi Vinodkumar Trivedi
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Hemant R Jadhav
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
18
|
Dragoi IT, Rezus C, Burlui AM, Bratoiu I, Rezus E. Multimodal Screening for Pulmonary Arterial Hypertension in Systemic Scleroderma: Current Methods and Future Directions. MEDICINA (KAUNAS, LITHUANIA) 2024; 61:19. [PMID: 39859001 PMCID: PMC11766816 DOI: 10.3390/medicina61010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025]
Abstract
Systemic sclerosis (SSc) is an immuno-inflammatory rheumatic disease that can affect both the skin and internal organs through fibrosis. Pulmonary arterial hypertension (PAH) is one of the most severe secondary complications. Structural changes in the vascular bed lead to increased pressures in the pulmonary circulation, severely impacting the right heart and significantly affecting mortality. The gold standard for diagnosing PAH is right heart catheterization (RHC), an invasive method for measuring cardiac pressure. Due to the high risk of complications, procedural difficulties, and significant costs, non-invasive screening for SSc-PAH has garnered significant interest. Echocardiography is likely the most important screening tool, providing structural and functional information about the right heart through measurements that have proven their utility over time. In addition to imagistic investigations, serum biomarkers aid in identifying patients at risk for PAH and can provide prognostic information. Currently, well-known serum biomarkers (NT-proBNP, uric acid) are used in screening; however, in recent years, researchers have highlighted new biomarkers that can enhance diagnostic accuracy for SSc patients. Pulmonary involvement can also be assessed through pulmonary function tests, which, using established thresholds, can provide additional information and help select patients requiring RHC. In conclusion, given the invasiveness of RHC, non-invasive screening methods are particularly important for SSc patients.
Collapse
Affiliation(s)
- Ioan Teodor Dragoi
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.T.D.); (I.B.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Alexandra Maria Burlui
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.T.D.); (I.B.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.T.D.); (I.B.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.T.D.); (I.B.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| |
Collapse
|
19
|
Yu J, Li Y, Hu J, Wang Y. Interleukin-33 induces angiogenesis after myocardial infarction via AKT/eNOS signaling pathway. Int Immunopharmacol 2024; 143:113433. [PMID: 39486188 DOI: 10.1016/j.intimp.2024.113433] [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: 08/08/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
Myocardial infarction (MI) is one of the leading causes of mortality and morbidity worldwide. MI-damaged vascular structures are difficult to completely restore due to the heart's low regenerative capacity. Given interleukin-33 (IL-33) as a potent endothelial activator promoting angiogenesis, this study investigated the role of IL-33 in angiogenesis and cardiac repair after MI. A mouse model of MI was established. IL-33 improved cardiac function and induced an increase in vascular density after MI. Besides, IL-33 promoted human endothelial cells proliferation, migration, and differentiation under both normoxic and hypoxic conditions, consistently with increased angiogenesis in vivo. Mechanistic studies demonstrated that IL-33 could promote angiogenesis by activating eNOS and AKT, and stimulating NO production in vivo and in vitro. Given that injection of exogenous IL-33 induced an inflammatory response, we employed a multifunctional biomimetic nanoparticle drug delivery system to deliver IL-33, thereby enhancing its targeting to the heart for fibrotic therapy and reducing inflammation. In conclusion, our results indicate that IL-33 promotes endothelial angiogenesis after MI through AKT/eNOS/NO signaling pathway. PM&EM/IL-33 nanoparticles may hold promising therapeutic potential for protecting cardiac ischemic injury and mitigating inflammation.
Collapse
Affiliation(s)
- Jiaqi Yu
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| | - Yuyu Li
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Jiaxin Hu
- Cardiovascular Disease Center, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, Hubei, China; Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi 445000, China
| | - Yuan Wang
- Beijing Anzhen Hospital, Capital Medical University, Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| |
Collapse
|
20
|
Abubakar M, Irfan U, Abdelkhalek A, Javed I, Khokhar MI, Shakil F, Raza S, Salim SS, Altaf MM, Habib R, Ahmed S, Ahmed F. Comprehensive Quality Analysis of Conventional and Novel Biomarkers in Diagnosing and Predicting Prognosis of Coronary Artery Disease, Acute Coronary Syndrome, and Heart Failure, a Comprehensive Literature Review. J Cardiovasc Transl Res 2024; 17:1258-1285. [PMID: 38995611 DOI: 10.1007/s12265-024-10540-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
Coronary artery disease (CAD), acute coronary syndrome (ACS), and heart failure (HF) are major global health issues with high morbidity and mortality rates. Biomarkers like cardiac troponins (cTn) and natriuretic peptides (NPs) are crucial tools in cardiology, but numerous new biomarkers have emerged, proving increasingly valuable in CAD/ACS. These biomarkers are classified based on their mechanisms, such as fibrosis, metabolism, inflammation, and congestion. The integration of established and emerging biomarkers into clinical practice is an ongoing process, and recognizing their strengths and limitations is crucial for their accurate interpretation, incorporation into clinical settings, and improved management of CVD patients. We explored established biomarkers like cTn, NPs, and CRP, alongside newer biomarkers such as Apo-A1, IL-17E, IgA, Gal-3, sST2, GDF-15, MPO, H-FABP, Lp-PLA2, and ncRNAs; provided evidence of their utility in CAD/ACS diagnosis and prognosis; and empowered clinicians to confidently integrate these biomarkers into clinical practice based on solid evidence.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan.
| | - Umema Irfan
- Department of Internal Medicine, Deccan College of Medical Sciences, Hyderabad, India
| | - Ahmad Abdelkhalek
- Department of Internal Medicine, Zhejiang University, Zhejiang, China
| | - Izzah Javed
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | | | - Fraz Shakil
- Department of Emergency Medicine, Mayo Hospital, Lahore, Pakistan
| | - Saud Raza
- Department of Anesthesia, Social Security Teaching Hospital, Lahore, Punjab, Pakistan
| | - Siffat Saima Salim
- Department of Surgery, Holy Family Red Crescent Medical College Hospital, Dhaka, Bangladesh
| | - Muhammad Mahran Altaf
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | - Rizwan Habib
- Department of Internal Medicine and Emergency, Indus Hospital, Lahore, Pakistan
| | - Simra Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| | - Farea Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| |
Collapse
|
21
|
Ozdemir E, Stavileci B, Ozdemir B, Aksoy FA, Colakoglu Gevher CZ, Dogan A, Ziyrek M. Could soluble ST2 levels be used to detect nondipper hypertensive subgroup in newly diagnosed hypertension patients. Blood Press Monit 2024; 29:284-289. [PMID: 38958500 DOI: 10.1097/mbp.0000000000000714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
AIM ST2 receptor is a member of toll-like/interleukin-1 receptor family. After the activation of IL-33/ST2 signaling pathway clinically detectable amount of soluble form of ST2 (sST2) is released into the circulation. Previous studies showed that sST2 levels were significantly higher in hypertension patients than in controls. In this prospective study, we aimed to analyze this relation and test the predictive accuracy of the sST2 level in diagnosis of nondipping hypertension in newly diagnosed hypertension patients. METHODS Three hundred thirty-seven patients (150 normal, 187 hypertension) who presented with symptoms of hypertension were included in the study. All patients underwent 24-h ambulatory blood pressure monitoring and sST2 measurement. RESULTS Of 187 hypertension patients, 92 of them had nondipping and 95 of them had dipping pattern. sST2 level was significantly higher in nondipping group compared to dipping group and control group (40.79 ± 7.77 vs. 32.47 ± 6.68; P < 0.0001 and 40.79 ± 7.77 vs. 20.09 ± 7.09; P < 0.0001 respectively). Binary logistic regression analysis revealed that; only sST2 level was an independent risk factor for hypertension [ P < 0.0001, β : 1.258, odds ratio (OR) (95% confidence interval (CI)): 1.158-1.366]. and also nondipping hypertension [ P < 0.0001, β : 1.208, OR (95% CI): 1.108-1.317]. CONCLUSION Based on the present study it could be concluded that sST2 level is significantly associated with the newly diagnosed hypertension and nondipping hypertension. Hence it could reliably be used to diagnose hypertension and nondipping hypertension with high sensitivity and specificity.
Collapse
Affiliation(s)
- Emrah Ozdemir
- Department of Cardiology, Biruni University Faculty of Medicine
| | - Berna Stavileci
- Department of Cardiology, Biruni University Faculty of Medicine
| | - Bahar Ozdemir
- Department of Internal Medicine, Dr. Sadi Konuk Training and Research Hospital
| | | | | | - Ali Dogan
- Department of Cardiology, Yeni Yuzyil University Faculty of Medicine
| | - Murat Ziyrek
- Department of Cardiology, Bagcilar Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
22
|
Kosyakovsky LB, de Boer RA, Ho JE. Screening for Heart Failure: Biomarkers to Detect Heightened Risk in the General Population. Curr Heart Fail Rep 2024; 21:591-603. [PMID: 39287754 DOI: 10.1007/s11897-024-00686-6] [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] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE OF REVIEW Heart failure (HF) represents a growing global burden of morbidity and mortality. Identifying individuals at risk for HF development is increasingly important, particularly given the advent of disease-modifying therapies for HF as well as its major risk factors such as obesity actalnd diabetes. We aim to review the key circulating biomarkers associated with future HF which may contribute to HF risk prediction. RECENT FINDINGS While current guidelines recommend the use of natriuretic peptides and cardiac troponins in HF risk stratification, there are a diverse array of other emerging protein, metabolic, transcriptomic, and genomic biomarkers of future HF development. These biomarkers not only lend insight into the underlying pathophysiology of HF, which spans inflammation to cardiac fibrosis, but also offer an opportunity to further refine HF risk in addition to established biomarkers. As evolving techniques in molecular biology enable an increased understanding of the complex biologic contributions to HF pathophysiology, there is an important opportunity to construct integrated clinical and multi-omic models to best capture HF risk. Moving forward, future studies should seek to understand the contributions of sex differences, underlying comorbidity burden, and HF subtypes to an individual's HF risk. Further studies are necessary to fully define the clinical utility of biomarker screening approaches to refine HF risk assessment, as well as to link risk assessment directly to preventive strategies for HF.
Collapse
Affiliation(s)
- Leah B Kosyakovsky
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA
| | - Rudolf A de Boer
- Department of Cardiology, Cardiovascular Institute, Thorax Center, Erasmus MC, Rotterdam, the Netherlands
| | - Jennifer E Ho
- Division of Cardiology, E/CLS 945, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215-5491, USA.
| |
Collapse
|
23
|
Liu W, Luo D, Liu X, Zhang Y, Wang Z. Predictive value of low serum interleukin-33 levels in acute ischemic stroke outcomes. Front Neurol 2024; 15:1503443. [PMID: 39650246 PMCID: PMC11621050 DOI: 10.3389/fneur.2024.1503443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 10/28/2024] [Indexed: 12/11/2024] Open
Abstract
Background Human interleukin-33 (IL-33), a member of the IL-1 family, has been identified as a therapeutic target due to its role as a proinflammatory mediator in various diseases. This study aims to evaluate the prognostic value of serum IL-33 levels in patients admitted with their first-ever acute ischemic stroke. Methods This single-center, prospective, observational study included 216 patients with acute ischemic stroke. Serum IL-33 levels were measured at hospital admission to assess their predictive value for functional outcomes and mortality within 3 months. IL-33 levels were dichotomized at the median into two groups: the reduced group (IL-33 ≤ median) and the normal group (IL-33 > median). Results The median age of the 216 patients was 66 years (interquartile range [IQR], 56-75), with 132 (61.6%) being women. IL-33 serum levels were inversely correlated with stroke severity, as measured by the National Institutes of Health Stroke Scale (NIHSS) score and lesion size. Patients in the reduced IL-33 group had a higher rate of unfavorable outcomes (55.6% vs. 18.5%; absolute difference, 29.2% [95% confidence interval (CI), 24.5% to 34.4%]; odds ratio (OR), 3.19 [95% CI, 1.72 to 5.91]) and mortality (24.1% vs. 3.7%; absolute difference, 15.8% [95% CI, 13.1% to 18.3%]; OR, 4.12 [95% CI, 1.38 to 12.31]) compared to the normal group. Furthermore, IL-33 levels enhanced the prognostic accuracy of the NIHSS for predicting functional outcomes (combined area under the curve [AUC], 0.84; 95% CI, 0.79-0.84; P < 0.001) and mortality (combined AUC, 0.88; 95% CI, 0.83-0.94; P < 0.001). Conclusion This study demonstrates that lower IL-33 levels are associated with increased stroke severity and poorer prognosis. These findings suggest that IL-33 may serve as a valuable biomarker for predicting poor outcomes following acute ischemic stroke.
Collapse
Affiliation(s)
- Wei Liu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Dongliang Luo
- Department of Neurosurgery, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Xingyu Liu
- Department of Neurosurgery, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Yuqing Zhang
- Department of Neurosurgery, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
DeVallance E, Bowdridge E, Garner K, Griffith J, Seman M, Batchelor T, Velayutham M, Goldsmith WT, Hussain S, Kelley EE, Nurkiewicz TR. The alarmin, interleukin-33, increases vascular tone via extracellular signal regulated kinase-mediated Ca 2+ sensitization and endothelial nitric oxide synthase inhibition. J Physiol 2024; 602:6087-6107. [PMID: 39540837 DOI: 10.1113/jp286990] [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: 05/28/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Alarmins are classified by their release from damaged or ruptured cells. Many alarmins have been found to increase vascular tone and oppose endothelium-dependent dilatation (EDD). Interleukin (IL)-33 plays a prominent role in lung injury and can be released during vascular injury and in chronic studies found to be cardioprotective. Our recent work has implicated IL-33 in acute vascular dysfunction following inhalation of engineered nanomaterials (ENM). However, the mechanisms linking IL-33 to vascular tone have not been interrogated. We therefore aimed to determine whether IL-33 directly influenced microvascular tone and endothelial function. Isolated feed arteries and in vivo arterioles from male and female Sprague-Dawley rats were used to determine direct vascular actions of IL-33. Mesenteric feed arteries and arterioles demonstrated reduced intraluminal diameters when treated with increasing concentrations of recombinant IL-33. IL-33 activated extracellular signal regulated kinase (ERK)1/2 of rat aortic smooth muscle cells but not phosphorylation of myosin light chain kinase. This suggested IL-33 may sensitize arterioles to Ca2+-mediated responses. Indeed, IL-33 augmented the myogenic- and phenylephrine-induced vasoconstriction. Additionally, incubation of arterioles with 1 ng IL-33 attenuated ACh-mediated EDD. Mechanistically, in human aortic endothelial cells, we demonstrate that IL-33-mediated ERK1/2 activation leads to inhibitory phosphorylation of serine 602 on endothelial nitric oxide synthase. Finally, we demonstrate that IL-33-ERK1/2 contributes to vascular tone following two known inducers of IL-33; ENM inhalation and the rupture endothelial cells. The present study provides novel evidence that IL-33 increases vascular tone via canonical ERK1/2 activation in microvascular smooth muscle and endothelium. Altogether, it is suggested IL-33 plays a critical role in microvascular homeostasis following barrier cell injury. KEY POINTS: Interleukin (IL)-33 causes a concentration-dependent reduction in feed artery diameter. IL-33 acts on vascular smooth muscle cells to augment Ca2+-mediated processes. IL-33 causes inhibitory phosphorylation of endothelial nitric oxide synthase and opposes endothelium-dependent dilatation. Engineered nanomaterial-induced lung injury and endothelial cell rupture in part act through IL-33 to mediate increased vascular tone.
Collapse
MESH Headings
- Animals
- Interleukin-33/metabolism
- Interleukin-33/pharmacology
- Rats, Sprague-Dawley
- Male
- Nitric Oxide Synthase Type III/metabolism
- Female
- Rats
- Vasodilation/drug effects
- Calcium/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Alarmins/metabolism
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/physiology
- Arterioles/physiology
- Arterioles/drug effects
- Arterioles/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiology
Collapse
Affiliation(s)
- Evan DeVallance
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth Bowdridge
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista Garner
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Julie Griffith
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Madison Seman
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Thomas Batchelor
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Murugesan Velayutham
- Department of Biochemistry and Molecular Medicine, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - W Travis Goldsmith
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Salik Hussain
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Eric E Kelley
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R Nurkiewicz
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
25
|
Jozwiak M, Lim SY, Si X, Monnet X. Biomarkers in cardiogenic shock: old pals, new friends. Ann Intensive Care 2024; 14:157. [PMID: 39414666 PMCID: PMC11485002 DOI: 10.1186/s13613-024-01388-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/29/2024] [Indexed: 10/18/2024] Open
Abstract
In cardiogenic shock, biomarkers should ideally help make the diagnosis, choose the right therapeutic options and monitor the patient in addition to clinical and echocardiographic indices. Among "old" biomarkers that have been used for decades, lactate detects, quantifies, and follows anaerobic metabolism, despite its lack of specificity. Renal and liver biomarkers are indispensable for detecting the effect of shock on organ function and are highly predictive of poor outcomes. Direct biomarkers of cardiac damage such as cardiac troponins, B-type natriuretic and N-terminal pro-B-type natriuretic peptides have a good prognostic value, but they lack specificity to detect a cardiogenic cause of shock, as many factors influence their plasma concentrations in critically ill patients. Among the biomarkers that have been more recently described, dipeptidyl peptidase-3 is one of the most interesting. In addition to its prognostic value, it could represent a therapeutic target in cardiogenic shock in the future as a specific antibody inhibits its activity. Adrenomedullin is a small peptide hormone secreted by various tissues, including vascular smooth muscle cells and endothelium, particularly under pathological conditions. It has a vasodilator effect and has prognostic value during cardiogenic shock. An antibody inhibits its activity and so adrenomedullin could represent a therapeutic target in cardiogenic shock. An increasing number of inflammatory biomarkers are also of proven prognostic value in cardiogenic shock, reflecting the inflammatory reaction associated with the syndrome. Some of them are combined to form prognostic proteomic scores. Alongside clinical variables, biomarkers can be used to establish biological "signatures" characteristic of the pathophysiological pathways involved in cardiogenic shock. This helps describe patient subphenotypes, which could in the future be used in clinical trials to define patient populations responding specifically to a treatment.
Collapse
Affiliation(s)
- Mathieu Jozwiak
- Service de Médecine Intensive Réanimation, CHU de Nice, Hôpital L'Archet 1, 151 Route Saint Antoine de Ginestière, 06200, Nice, France.
- UR2CA, Unité de Recherche Clinique Côte d'Azur, Université Côte d'Azur, 06200, Nice, France.
| | - Sung Yoon Lim
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU 4 CORREVE, Inserm UMR S_999, FHU SEPSIS, CARMAS, Université Paris-Saclay, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Xiang Si
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU 4 CORREVE, Inserm UMR S_999, FHU SEPSIS, CARMAS, Université Paris-Saclay, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France
- Department of Critical Care Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xavier Monnet
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU 4 CORREVE, Inserm UMR S_999, FHU SEPSIS, CARMAS, Université Paris-Saclay, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France
| |
Collapse
|
26
|
Ali S, Khlidj Y, Kumar M, Kumar S, Duhan S, Farooq F, Keisham B, Ponna PK, Modi K. Gender disparities in outcomes of cardiogenic shock secondary to Takotsubo cardiomyopathy. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 46:100453. [PMID: 39319103 PMCID: PMC11419888 DOI: 10.1016/j.ahjo.2024.100453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024]
Abstract
Background Takotsubo cardiomyopathy (TTC) has a preponderance for females, particularly postmenopausal. However, recent data from multicenter registries identified a worse prognosis in male patients, particularly with cardiogenic shock. We aim to investigate gender disparities in outcomes of TTC-associated cardiogenic shock (TTC-CS). Methods The National Readmission Database (2016-2020) was utilized to identify TTC-CS hospitalizations. Cohorts were stratified by gender. A Propensity Score Matching (PSM) model, which utilized complete Mahalanobis Distance Matching within the Propensity Score Caliper following multivariate regression, successfully matched males and females. Pearson's χ2 test was applied to the propensity-matched cohorts to compare outcomes. Results Among 12,803 TTC-CS hospitalizations, the majority (74.1 %) were females (N: 9490), and 25.9 % were males (N: 3313). On propensity-matched cohorts (2609), males were found to have higher in-hospital mortality (31 % vs. 26 %, p < 0.001), higher incidence of sudden cardiac arrest (14 % vs. 10.8 %, p < 0.001), endotracheal intubation (52.1 % vs. 48.8 %, p: 0.001), acute liver injury (18 % vs. 15.9 %, p: 0.004), acute stroke (7.2 % vs. 5.8 %, p: 0.004), cardiac arrhythmias (55.1 % vs. 49.3 %, p < 0.001) and acute kidney injury (63.1 % vs. 49 %, p < 0.001); while female patients were found to have higher utilization of mechanical circulatory support (MCS) modalities (16.1 % vs 13.2 %, p < 0.001).Males had a higher adjusted cost of hospitalization ($54,537 vs. $42,805, p < 0.001) with a higher median length of hospital stay (10 vs. 9 days, p < 0.001). The two groups had no significant difference in 30, 90, and 180-day readmission rates (p > 0.05). From 2016 to 2020; mortality has not changed significantly for TTC-CS, while the use of percutaneous coronary angiogram (PCA) and MCS has down-trended (p-trend < 0.05). Conclusion For TTC-CS hospitalization, males have higher in-hospital mortality and complication rates, along with higher LOS and cost of hospitalization. Despite advances in the management of CS, there was no significant difference in mortality from 2016 to 2020.
Collapse
Affiliation(s)
- Shafaqat Ali
- Department of Internal Medicine, Louisiana State University, Shreveport, LA, USA
| | - Yehya Khlidj
- Department of Medicine, University of Algiers 1, Algiers, Algeria
| | - Manoj Kumar
- Department of Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, IL, USA
| | - Sanjay Kumar
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens, NY, USA
| | - Sanchit Duhan
- Department of Cardiology, Carle Foundation Hospital, Urbana, IL, USA
| | - Faryal Farooq
- Department of Medicine, Allama Iqbal Medical College Lahore, Pakistan
| | - Bijeta Keisham
- Department of Medicine, Shandong Second Medical University, Weifang, China
| | - Pramod Kumar Ponna
- Department of Internal Medicine, Louisiana State University, Shreveport, LA, USA
| | - Kalgi Modi
- Department of Cardiology, Louisiana State University, Shreveport, LA, USA
| |
Collapse
|
27
|
Yadigaroğlu M, Güzel M, Erdem E, Görgün S, Aksu EA, Ocak M, Yadigaroğlu NÖ, Demir MT, Yücel M. IL-33/sST2 signaling pathway in pulmonary thromboembolism: A clinical observational study. Cytokine 2024; 182:156707. [PMID: 39084069 DOI: 10.1016/j.cyto.2024.156707] [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: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/20/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Pulmonary thromboembolism (PTE) is a cardiovascular emergency that can result in mortality. In the interleukin-33 (IL-33) /soluble suppression of tumorigenicity 2 (sST2) signaling pathway, increased sST2 is a cardiovascular risk factor. This study aimed to investigate the effectiveness of biomarkers in the IL-33/sST2 signaling pathway in determining PTE diagnosis, clinical severity, and mortality. METHOD This study was conducted as a single-center, prospective, observational study. Patients admitted to the emergency department and diagnosed with PTE constituted the patient group (n = 112), and healthy volunteers with similar sociodemographic characteristics constituted the control group (n = 62). Biomarkers in the IL-33/sST2 signaling pathway were evaluated for diagnosis, clinical severity, and prognosis. RESULTS IL-33 was lower in the patient group than in the control group (275.89 versus 403.35 pg/mL), while sST2 levels were higher in the patient group than in the control group (53.16 versus 11.78 ng/mL) (p < 0.001 and p = 0.001; respectively). The AUC of IL-33 to diagnose PTE was 0.656 (95 % CI: 0.580-0.726). The optimal IL-33 cut-off point to diagnose PTE was ≤304.11 pg/mL (56.2 % sensitivity, 79 % specificity). The AUC of sST2 to diagnose PTE was 0.818 (95 % CI: 0.752-0.872). The optimal sST2 cut-off point to diagnose PTE was >14.48 ng/mL (83 % sensitivity, 71 % specificity). IL-33 levels were lower in patients with mortality (169.85 versus 332.04 pg/mL) compared to patients without mortality, whereas sST2 levels were higher in patients with mortality (118.32 versus 28.07 ng/mL) compared to patients without mortality (p > 0.001 for both). The AUC of IL-33 to predict the mortality of PTE was 0.801 (95 % CI: 0.715-0.870). The optimal IL-33 cut-off point to predict the mortality of PTE was ≤212.05 pg/mL (75 % sensitivity, 79.5 % specificity). The AUC of sST2 to predict the mortality of PTE was 0.824 (95 % CI: 0.740-0.889). The optimal sST2 cut-off point to predict the mortality of PTE was >81 ng/mL (95.8 % sensitivity, 78.4 % specificity). CONCLUSION In the IL-33/ST2 signaling pathway, decreased IL-33 and increased sST2 are valuable biomarkers for diagnosis and prediction of mortality in patients with PTE.
Collapse
Affiliation(s)
- Metin Yadigaroğlu
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| | - Murat Güzel
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| | - Elif Erdem
- Şırnak State Hospital, Department of Emergency Medicine, Şırnak, Turkey.
| | - Selim Görgün
- Samsun Education and Research Hospital, Department of Microbiology and Clinical Microbiology, Samsun, Turkey.
| | - Esra Arslan Aksu
- Samsun University, Faculty of Medicine, Department of Chest Diseases, Samsun, Turkey.
| | - Metin Ocak
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| | | | - Mehmet Tevfik Demir
- Samsun Education and Research Hospital, Department of Emergency Medicine, Samsun, Turkey.
| | - Murat Yücel
- Samsun University, Faculty of Medicine, Department of Emergency Medicine, Samsun, Turkey.
| |
Collapse
|
28
|
Vorlat A, van Eijk J, Wiersma S, Smid L, Depooter S, Paelinck B, Guerti K, Peeters B, Sturkenboom N, Van Craenenbroeck E, Heidbuchel H, Van De Heyning C. Clinical determinants and biomarkers associated with cardiac fibrosis after heart transplantation as assessed by magnetic resonance: Size matters. IJC HEART & VASCULATURE 2024; 54:101479. [PMID: 39221115 PMCID: PMC11365389 DOI: 10.1016/j.ijcha.2024.101479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Background Cardiac fibrosis is increasingly recognized as a marker of worse outcomes in long-term follow-up after heart transplantation (HTX). We investigated the clinical determinants and biomarkers of focal and interstitial cardiac fibrosis as assessed with cardiac magnetic resonance (CMR). Methods Consecutive HTX recipients underwent CMR with late gadolinium enhancement for focal myocardial fibrosis and T1 mapping for interstitial fibrosis. We calculated the correlations of these findings with clinical parameters, history, biomarkers of fibrosis (B-type natriuretic peptide (BNP), growth differentiation factor-15, galectin-3 and soluble ligand ST2) and echocardiography. Results Forty-eight HTX patients were included: median age 63 ± 13 years, 11 ± 6 years after heart transplantation. Only donor weight (p 0.044) and the rate of a > 30 % mismatch between donor and recipient weight (p 0.02) were significantly different in patients with vs. without late LGE. Extracellular volume (ECV) was correlated with the weight mismatch between donor and recipient (r = 0.32, p 0.04), resulting in a higher ECV for oversized donors. BNP was the only biomarker of the four studied that was correlated with interstitial fibrosis as assessed by ECV (r = 0.35, p 0.04). T1 relaxation time was correlated with treated acute cellular rejection grade ≥ 2 (ISHLT grading) (r = 0.34, p 0.02). Conclusion Both focal and interstitial fibrosis, as determined by CMR, after heart transplantation are correlated with donor and recipient weight mismatch. BNP was the only biomarker clinically relevant to interstitial cardiac fibrosis.
Collapse
Affiliation(s)
- Anne Vorlat
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Belgium
| | | | | | - Leroy Smid
- Medicine, University of Antwerp, Belgium
| | | | - Bernard Paelinck
- Department Cardiac Surgery, Antwerp University Hospital, University of Antwerp, Belgium
| | - Khadija Guerti
- Department of Clinical Chemistry, Antwerp University Hospital, University of Antwerp, Belgium
| | - Bart Peeters
- Department of Clinical Chemistry, Antwerp University Hospital, University of Antwerp, Belgium
| | - Nicole Sturkenboom
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Belgium
| | | | - Hein Heidbuchel
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Belgium
| | | |
Collapse
|
29
|
Seong D, Mataraso S, Espinosa C, Berson E, Reincke SM, Xue L, Kashiwagi C, Kim Y, Shu CH, Chung P, Ghanem M, Xie F, Wong RJ, Angst MS, Gaudilliere B, Shaw GM, Stevenson DK, Aghaeepour N. Generating pregnant patient biological profiles by deconvoluting clinical records with electronic health record foundation models. Brief Bioinform 2024; 25:bbae574. [PMID: 39545787 PMCID: PMC11565587 DOI: 10.1093/bib/bbae574] [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: 07/31/2024] [Revised: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Translational biology posits a strong bi-directional link between clinical phenotypes and a patient's biological profile. By leveraging this bi-directional link, we can efficiently deconvolute pre-existing clinical information into biological profiles. However, traditional computational tools are limited in their ability to resolve this link because of the relatively small sizes of paired clinical-biological datasets for training and the high dimensionality/sparsity of tabular clinical data. Here, we use state-of-the-art foundation models (FMs) for electronic health record (EHR) data to generate proteomics profiles of pregnant patients, thereby deconvoluting pre-existing clinical information into biological profiles without the cost and effort of running large-scale traditional omics studies. We show that FM-derived representations of a patient's EHR data coupled with a fully connected neural network prediction head can generate 206 blood protein expression levels. Interestingly, these proteins were enriched for developmental pathways, while proteins not able to be generated from EHR data were enriched for metabolic pathways. Finally, we show a proteomic signature of gestational diabetes that includes proteins with established and novel links to gestational diabetes. These results showcase the power of FM-derived EHR representations in efficiently generating biological states of pregnant patients. This capability can revolutionize disease understanding and therapeutic development, offering a cost-effective, time-efficient, and less invasive alternative to traditional methods of generating proteomics.
Collapse
Affiliation(s)
- David Seong
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Medical Scientist Training Program, Stanford University School of Medicine, 1265 Welch Road, Stanford CA, 94305, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Camilo Espinosa
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Eloise Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - S Momsen Reincke
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Lei Xue
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Chloe Kashiwagi
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Yeasul Kim
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Chi-Hung Shu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Philip Chung
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Marc Ghanem
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Feng Xie
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
| | - Nima Aghaeepour
- Immunology Program, Stanford University School of Medicine, 240 Pasteur Drive, Palo Alto CA, 94304, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford CA, 94305, United States
- Department of Biomedical Data Science, Stanford University, 1265 Welch Road, Stanford CA, 94305, United States
| |
Collapse
|
30
|
Zhang C, Li G, Zhang F, Zhang Y, Hong S, Gao S, Liu Y, Du J, Li Y. IL-33 Facilitates Fibro-Adipogenic Progenitors to Establish the Pro-Regenerative Niche after Muscle Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405299. [PMID: 39037903 PMCID: PMC11425282 DOI: 10.1002/advs.202405299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/03/2024] [Indexed: 07/24/2024]
Abstract
During the process of muscle regeneration post-injury in adults, muscle stem cells (MuSCs) function is facilitated by neighboring cells within the pro-regenerative niche. However, the precise mechanism triggering the initiation of signaling in the pro-regenerative niche remains unknown. Using single-cell RNA sequencing, 14 different muscle cells are comprehensively mapped during the initial stage following injury. Among these, macrophages and fibro-adipogenic progenitor cells (FAPs) exhibit the most pronounced intercellular communication with other cells. In the FAP subclusters, the study identifies an activated FAP phenotype that secretes chemokines, such as CXCL1, CXCL5, CCL2, and CCL7, to recruit macrophages after injury. Il1rl1, encoding the protein of the interleukin-33 (IL-33) receptor, is identified as a highly expressed signature surface marker of the FAP phenotype. Following muscle injury, autocrine IL-33, an alarmin, has been observed to activate quiescent FAPs toward this inflammatory phenotype through the IL1RL1-MAPK/NF-κB signaling pathway. Il1rl1 deficiency results in decreased chemokine expression and recruitment of macrophages, accompanied by impaired muscle regeneration. These findings elucidate a novel mechanism involving the IL-33/IL1RL1 signaling pathway in promoting the activation of FAPs and facilitating muscle regeneration, which can aid the development of therapeutic strategies for muscle-related disorders and injuries.
Collapse
Affiliation(s)
- Congcong Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Guoqi Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Fan Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Yanhong Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Shiyao Hong
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Yan Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
- Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, 100029, China
| |
Collapse
|
31
|
Meekers E, Dupont M. Role of Imaging and Biomarkers in Identifying, Monitoring, and Promoting Myocardial Recovery. Methodist Debakey Cardiovasc J 2024; 20:42-53. [PMID: 39193116 PMCID: PMC11348843 DOI: 10.14797/mdcvj.1381] [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: 03/24/2024] [Accepted: 06/22/2024] [Indexed: 08/29/2024] Open
Abstract
Reverse remodeling, the overarching concept behind myocardial recovery, describes the process in which the maladaptive cardiac structural and functional alterations are reversed by removing the underlying etiology or by therapy. This review addresses different imaging modalities and biomarkers as possible predictors for reverse remodeling in patients with chronic heart failure. Although echocardiography remains the imaging modality of choice in daily practice, the presence and amount of fibrosis on cardiac magnetic resonance is a better predictor and inversely correlated with the likelihood for reverse remodeling. A decrease in NT-proBNP levels and serum soluble ST3 during follow-up is associated with better clinical and structural outcomes. The role of troponins and galectine-3 is less clear. There is a promising role for microRNAs in the future, although more research is necessary. Accurate predictors of reverse remodeling could help identify patients with an increased likelihood for reverse remodeling and, in turn, improve patient-tailored medicine.
Collapse
Affiliation(s)
- Evelyne Meekers
- Ziekenhuis Oost-Limburg A.V., Genk, Belgium
- Hasselt University, Diepenbeek, Belgium
| | | |
Collapse
|
32
|
Feng Y, He LQ. Soluble ST2: A Novel Biomarker for Diagnosis and Prognosis of Cardiovascular Disease. Curr Med Sci 2024; 44:669-679. [PMID: 39096477 DOI: 10.1007/s11596-024-2907-x] [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: 12/14/2023] [Accepted: 05/30/2024] [Indexed: 08/05/2024]
Abstract
The increasing incidence of cardiovascular disease (CVD) is a significant global health concern, affecting millions of individuals each year. Accurate diagnosis of acute CVD poses a formidable challenge, as misdiagnosis can significantly decrease patient survival rates. Traditional biomarkers have played a vital role in the diagnosis and prognosis of CVDs, but they can be influenced by various factors, such as age, sex, and renal function. Soluble ST2 (sST2) is a novel biomarker that is closely associated with different CVDs. Its low reference change value makes it suitable for continuous measurement, unaffected by age, kidney function, and other confounding factors, facilitating risk stratification of CVDs. Furthermore, the combination of sST2 with other biomarkers can enhance diagnostic accuracy and prognostic value. This review aims to provide a comprehensive overview of sST2, focusing on its diagnostic and prognostic value as a myocardial marker for different types of CVDs and discussing the current limitations of sST2.
Collapse
Affiliation(s)
- Yin Feng
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Qun He
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
33
|
Bisen S, Verma SK, Mukhopadhyay CS, Singh NK. A neutrophil elastase-generated mature form of IL-33 is a potent regulator of endothelial cell activation and proliferative retinopathy. Exp Mol Med 2024; 56:1703-1716. [PMID: 39085349 PMCID: PMC11372157 DOI: 10.1038/s12276-024-01279-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 08/02/2024] Open
Abstract
Human interleukin-33 (IL-33) is a 270 amino acid protein that belongs to the IL-1 cytokine family and plays an important role in various inflammatory disorders. Neutrophil proteases (Cathepsin G and Elastase) and mast cell proteases (tryptase and chymase) regulate the activity of IL-33 by processing full-length IL-33 into its mature form. There is little evidence on the role of these mature forms of IL-33 in retinal endothelial cell signaling and pathological retinal angiogenesis. Here, we cloned, expressed, and purified the various mature forms of human IL-33 and then evaluated the effects of IL-3395-270, IL-3399-270, IL-33109-270, and IL-33112-270 on angiogenesis in human retinal microvascular endothelial cells (HRMVECs). We observed that IL-3395-270, IL-3399-270, IL-33109-270, and IL-33112-270 significantly induced HRMVEC migration, tube formation and sprouting angiogenesis. However, only IL-3399-270 could induce HRMVEC proliferation. We used a murine model of oxygen-induced retinopathy (OIR) to assess the role of these mature forms of IL-33 in pathological retinal neovascularization. Our 3'-mRNA sequencing and signaling studies indicated that IL-3399-270 and IL-33109-270 were more potent at inducing endothelial cell activation and angiogenesis than the other mature forms. We found that genetic deletion of IL-33 significantly reduced OIR-induced retinal neovascularization in the mouse retina and that intraperitoneal administration of mature forms of IL-33, mainly IL-3399-270 and IL-33109-270, significantly restored ischemia-induced angiogenic sprouting and tuft formation in the hypoxic retinas of IL-33-/- mice. Thus, our study results suggest that blockade or inhibition of IL-33 cleavage by neutrophil proteases could help mitigate pathological angiogenesis in proliferative retinopathies.
Collapse
Affiliation(s)
- Shivantika Bisen
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, 48202, USA
| | - Shailendra Kumar Verma
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, 48202, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, San Diego, CA, 92037, USA
| | - Chandra Sekhar Mukhopadhyay
- Department of Bioinformatics, School of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, 141004, India
| | - Nikhlesh K Singh
- Integrative Biosciences Center, Wayne State University, Detroit, MI, 48202, USA.
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
34
|
Hassan GF, Cohen LS, Alexander-Brett J. IL-33: Friend or foe in transplantation? J Heart Lung Transplant 2024; 43:1235-1240. [PMID: 38452960 PMCID: PMC11246814 DOI: 10.1016/j.healun.2024.02.1459] [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: 12/20/2023] [Revised: 02/17/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024] Open
Abstract
Several reports have highlighted the dichotomous nature of the Interleukin-33 (IL-33) system in cardiac and lung disease, where this cytokine can exert both protective effects and drive pro-inflammatory responses in a context-specific manner. This State-of-the-Art review focuses on preclinical mechanistic studies of the IL-33 system in development of allograft rejection in heart and lung transplantation. We address the scope of potential cellular sources of IL-33 and pathways for cellular release that may impact the study of this cytokine system in transplant models. We then highlight soluble IL-33 receptor as a biomarker in cardiac allograft rejection and detail preclinical models that collectively demonstrate a role for this cytokine in driving type-2 immune programs to protect cardiac allografts. We contrast this with investigation of IL-33 in lung transplantation, which has yielded mixed and somewhat conflicting results when comparing human studies with preclinical models, which have implicated the IL-33 system in both allograft tolerance and acceleration of chronic rejection. We summarize and interpret these results in aggregate and provide future directions for study of IL-33 in heart and lung transplantation.
Collapse
Affiliation(s)
- Ghandi F Hassan
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Lucy S Cohen
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, Saint Louis, Missouri
| | - Jen Alexander-Brett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, Saint Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri.
| |
Collapse
|
35
|
Hammer A, Marka F, Baumer U, Hofer F, Kazem N, Koller L, Steinacher E, Zimpfer D, Andreas M, Steinlechner B, Demyanets S, Niessner A, Sulzgruber P, Stojkovic S. Soluble ST2 is associated with postoperative atrial fibrillation after cardiac surgery in postmenopausal women. Clin Chim Acta 2024; 561:119815. [PMID: 38879062 DOI: 10.1016/j.cca.2024.119815] [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: 05/03/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Postoperative atrial fibrillation (POAF) represents the most common complication following cardiac surgery. Approximately one-third of patients experiencing POAF transition to atrial fibrillation within a year, challenging the notion of POAF as merely a transient event. Soluble ST2 (sST2) is an established biomarker regarding fibrosis and myocardial stretch, however, its role in predicting the onset of POAF remains unclear. METHODS Preoperative sST2 levels have been assessed in 496 individuals with no prior history of AF who underwent elective cardiac surgery, including valve, coronary artery bypass graft surgery, or a combined procedure. RESULTS The average age was 70 years, and 29.4 % were female. Overall, 42.3 % developed POAF. sST2 levels were found to be significantly higher in patients with POAF. Interestingly, sST2 was only predictive of POAF in females with an adjusted OR of 1.894 (95 %CI:1.103-3.253; p = 0.021) and not males (OR:1.091; 95 %CI:0.849-1.402; p = 0.495). Furthermore, within a linear regression model it was observed that for every 1 ng/mL increase in sST2 levels, the average POAF duration extended by 39.5 min (95 %CI:15.8-63.4 min; p = 0.001). CONCLUSION sST2 predicts the onset of POAF in women but not men undergoing cardiac surgery. Furthermore, sST2 levels were associated with the subsequent burden of POAF. Thus, assessment of sST2 in addition to clinical risk factors could improve risk stratification for development of POAF following elective cardiac surgery.
Collapse
Affiliation(s)
- Andreas Hammer
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Frieda Marka
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Ulrike Baumer
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria
| | - Felix Hofer
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria
| | - Niema Kazem
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria
| | - Lorenz Koller
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria
| | - Eva Steinacher
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria
| | - Daniel Zimpfer
- Division of Cardiac Surgery, Dept. of Surgery, Medical University of Vienna, Austria
| | - Martin Andreas
- Division of Cardiac Surgery, Dept. of Surgery, Medical University of Vienna, Austria
| | - Barbara Steinlechner
- Division of Cardiothoracic Anesthesiology and Intensive Care, Dept. of Anesthesiology, Intensive Care and Pain Medicine, Medical University of Vienna, Austria
| | - Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Department of Laboratory Medicine, Clinic Hietzing, Vienna, Austria
| | - Alexander Niessner
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria
| | - Patrick Sulzgruber
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria.
| | - Stefan Stojkovic
- Division of Cardiology, Dept. of Internal Medicine II. Medical University of Vienna, Austria
| |
Collapse
|
36
|
Hai S, Li X, Xie E, Wu W, Gao Q, Yu B, Hu J, Xu F, Zheng X, Zhang BH, Wu D, Yan W, Ning Q, Wang X. Intestinal IL-33 promotes microbiota-derived trimethylamine N -oxide synthesis and drives metabolic dysfunction-associated steatotic liver disease progression by exerting dual regulation on HIF-1α. Hepatology 2024:01515467-990000000-00950. [PMID: 38985971 DOI: 10.1097/hep.0000000000000985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND AIMS Gut microbiota plays a prominent role in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). IL-33 is highly expressed at mucosal barrier sites and regulates intestinal homeostasis. Herein, we aimed to investigate the role and mechanism of intestinal IL-33 in MASLD. APPROACH AND RESULTS In both humans and mice with MASLD, hepatic expression of IL-33 and its receptor suppression of tumorigenicity 2 (ST2) showed no significant change compared to controls, while serum soluble ST2 levels in humans, as well as intestinal IL-33 and ST2 expression in mice were significantly increased in MASLD. Deletion of global or intestinal IL-33 in mice alleviated metabolic disorders, inflammation, and fibrosis associated with MASLD by reducing intestinal barrier permeability and rectifying gut microbiota dysbiosis. Transplantation of gut microbiota from IL-33 deficiency mice prevented MASLD progression in wild-type mice. Moreover, IL-33 deficiency resulted in a decrease in the abundance of trimethylamine N -oxide-producing bacteria. Inhibition of trimethylamine N -oxide synthesis by 3,3-dimethyl-1-butanol mitigated hepatic oxidative stress in mice with MASLD. Nuclear IL-33 bound to hypoxia-inducible factor-1α and suppressed its activation, directly damaging the integrity of the intestinal barrier. Extracellular IL-33 destroyed the balance of intestinal Th1/Th17 and facilitated Th1 differentiation through the ST2- Hif1a - Tbx21 axis. Knockout of ST2 resulted in a diminished MASLD phenotype resembling that observed in IL-33 deficiency mice. CONCLUSIONS Intestinal IL-33 enhanced gut microbiota-derived trimethylamine N -oxide synthesis and aggravated MASLD progression through dual regulation on hypoxia-inducible factor-1α. Targeting IL-33 and its associated microbiota may provide a potential therapeutic strategy for managing MASLD.
Collapse
Affiliation(s)
- Suping Hai
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xitang Li
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Erliang Xie
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhui Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Gao
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Binghui Yu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Junjian Hu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Feiyang Xu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xizhe Zheng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Bin-Hao Zhang
- Department of Surgery, Hepatic Surgery Center, Institute of Hepato-Pancreato-Biliary Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Weiming Yan
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Shao C, Cao Y, Wang Z, Wang X, Li C, Hao X, Wang L, Du Z, Yang F, Jiang C, Wang H, Hao Y, Han J, Hou X. Soluble ST2 predicts continuous renal replacement therapy in patients receiving venoarterial extracorporeal membrane oxygenation. Perfusion 2024; 39:927-934. [PMID: 37051884 DOI: 10.1177/02676591231169410] [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] [Indexed: 04/14/2023]
Abstract
OBJECTIVE This study aimed to evaluate the relationship between plasma soluble ST2 (sST2) levels 24 h after extracorporeal membrane oxygenation (ECMO) initiation and continuous renal replacement therapy (CRRT) in patients receiving venoarterial ECMO (V-A ECMO) support. METHODS AND RESULTS Data of patients who received ECMO support for postcardiotomy cardiogenic shock between January 2017 and July 2019 were retrospectively collected from Beijing Anzhen Hospital, Capital Medical University. Ultimately, 116 patients were included in the present study for analysis. The concentration of sST2 was determined by enzyme-linked immunosorbent assay (ELISA). The log10 sST2 levels were higher in patients undergoing CRRT than those who did not (6.06 vs. 6.22, p = 0.019). Patients undergoing CRRT had a lower survival rate than those who did not (32.8% vs. 67.3%, p < 0.001). In the univariate logistic regression analysis, sST2, HCO3-, lactate, and creatinine levels 24 h after ECMO initiation were related to CRRT (p < 0.05). In the multivariate logistic regression analysis, HCO3- and sST2 were identified as independent risk factors for CRRT use in patients undergoing ECMO (p < 0.05). The area under receiver operator characteristic curve (AUC) for sST2 and HCO3- together was 0.72 (95% confidence interval (CI), 0.79-0.91), which was better than those of sST2 or HCO3- alone (0.63 vs. 0.67). CONCLUSIONS sST2 and HCO3-levels at 24 h after ECMO initiation were associated with CRRT and could predict CRRT use in postcardiotomy cardiogenic shock patients undergoing ECMO.
Collapse
Affiliation(s)
- Chengcheng Shao
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Yu Cao
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Sohu Inc, Beijing, China
| | - Zengtao Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaomeng Wang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Chenglong Li
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Xing Hao
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Liangshan Wang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Zhongtao Du
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Feng Yang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Chunjing Jiang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Hong Wang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| | - Yu Hao
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Sohu Inc, Beijing, China
| | - Junyan Han
- Biomedical Innovation Center, Beijing Shijitan Hospital, Capital Medical University, Sohu Inc, Beijing, China
| | - Xiaotong Hou
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, No.10 Tieyi Road, Beijing 100038, China
| |
Collapse
|
38
|
Ruan J, Tian Q, Li S, Zhou X, Sun Q, Wang Y, Xiao Y, Li M, Chang K, Yi X. The IL-33-ST2 axis plays a vital role in endometriosis via promoting epithelial-mesenchymal transition by phosphorylating β-catenin. Cell Commun Signal 2024; 22:318. [PMID: 38858740 PMCID: PMC11163813 DOI: 10.1186/s12964-024-01683-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
OBJECTIVES Interleukin 33 (IL-33) is a crucial inflammatory factor that functions as an alarm signal in endometriosis (EMs). Epithelial-mesenchymal transition (EMT), a process related to inflammatory signals, intracellular reactive oxygen species (ROS) production, and lipid peroxidation, have been proposed as potential mechanisms that contribute to the development and progression of EMs. IL-33 is highly upregulated in the ectopic milieu. Moreover, ectopic endometrial cells constitutively express interleukin-33 receptor ST2 (IL-33R). However, the role of IL-33/ST2 in the EMT of EMs remains largely unknown. In this study, we aimed to mechanistically determine the role of IL-33/ST2 in EMs-associated fibrosis. MATERIALS AND METHODS We established a non-lethal oxidative stress model to explore the conditions that trigger IL-33 induction. We performed α-smooth muscle actin (α-SMA) protein detection, cell counting kit-8 (CCK-8) assays, and scratch assays to analyze the impact of IL-33 on primary endometrial stromal cells (ESCs) proliferation and invasion. Clinical samples from patients with or without EMs were subjected to immunohistochemical (IHC) and and immunofluorescence(IF) staining to assess the clinical relevance of IL-33 receptor ST2 and EMT-related proteins. Furthermore, we used the ectopic human endometrial epithelial cell line 12Z and normal human epithelial cell line EEC to evaluate the effects of IL-33 on Wnt/β-catenin signaling. The effect of IL-33 on EMT-associated fibrosis was validated in vivo by intraperitoneal injections of IL-33 and antiST2. RESULTS We observed that ectopic milieu, characterized by ROS, TGF-β1, and high level of estrogen, triggers the secretion of IL-33 from ectopic ESCs. Ectopic endometrial lesions exhibited higher level of fibrotic characteristics and ST2 expression than that in the normal endometrium. Exogenous recombinant human (rhIL-33) enhanced ESC migration and survival. Similarly, 12Z cells displayed a higher degree of EMT characteristics with elevated expression of CCN4 and Fra-1, downstream target genes of the WNT/β-catenin pathway, than that observed in EECs. Conversely, blocking IL-33 with neutralizing antibodies, knocking down ST2 or β-catenin with siRNA, and β-catenin dephosphorylation abolished its effects on EMT promotion. In vivo validation demonstrated that IL-33 significantly promotes EMs-related fibrosis through the activation of Wnt/β-catenin signaling. CONCLUSION Our data strongly support the vital role of the IL-33/ST2 pathway in EMs-associated fibrosis and emphasize the importance of the EMT in the pathophysiology of fibrosis. Targeting the IL-33/ST2/Wnt/β-catenin axis may hold promise as a feasible therapeutic approach for controlling fibrosis in EMs.
Collapse
Affiliation(s)
- Jingyao Ruan
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Qi Tian
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Siting Li
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Xiaoyu Zhou
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Qianzhi Sun
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yuning Wang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yinping Xiao
- Department of Pathology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Kaikai Chang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| | - Xiaofang Yi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| |
Collapse
|
39
|
Patil M, Singh S, Dubey PK, Tousif S, Umbarkar P, Zhang Q, Lal H, Sewell-Loftin MK, Umeshappa CS, Ghebre YT, Pogwizd S, Zhang J, Krishnamurthy P. Fibroblast-Specific Depletion of Human Antigen R Alleviates Myocardial Fibrosis Induced by Cardiac Stress. JACC Basic Transl Sci 2024; 9:754-770. [PMID: 39070272 PMCID: PMC11282885 DOI: 10.1016/j.jacbts.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 07/30/2024]
Abstract
Cardiac fibrosis can be mitigated by limiting fibroblast-to-myofibroblast differentiation and proliferation. Human antigen R (HuR) modulates messenger RNA stability and expression of multiple genes. However, the direct role of cardiac myofibroblast HuR is unknown. Myofibroblast-specific deletion of HuR limited cardiac fibrosis and preserved cardiac functions in pressure overload injury. Knockdown of HuR in transforming growth factor-β1-treated cardiac fibroblasts suppressed myofibroblast differentiation and proliferation. HuR deletion abrogated the expression and messenger RNA stability of cyclins D1 and A2, suggesting a potential mechanism by which HuR promotes myofibroblast proliferation. Overall, these data suggest that inhibition of HuR could be a potential therapeutic approach to limit cardiac fibrosis.
Collapse
Affiliation(s)
- Mallikarjun Patil
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sarojini Singh
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Praveen Kumar Dubey
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sultan Tousif
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qinkun Zhang
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hind Lal
- Division of Cardiovascular Disease, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Yohannes T. Ghebre
- Department of Radiation Oncology, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Steven Pogwizd
- Comprehensive Cardiovascular Center, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Heersink School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
40
|
Chen Z, Balachandran YL, Chong WP, Chan KWY. Roles of Cytokines in Alzheimer's Disease. Int J Mol Sci 2024; 25:5803. [PMID: 38891990 PMCID: PMC11171747 DOI: 10.3390/ijms25115803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
The neuroimmune system is a collection of immune cells, cytokines, and the glymphatic system that plays a pivotal role in the pathogenesis and progression of Alzheimer's disease (AD). Of particular focus are cytokines, a group of immune signaling molecules that facilitate communication among immune cells and contribute to inflammation in AD. Extensive research has shown that the dysregulated secretion of certain cytokines (IL-1β, IL-17, IL-12, IL-23, IL-6, and TNF-α) promotes neuroinflammation and exacerbates neuronal damage in AD. However, anti-inflammatory cytokines (IL-2, IL-3, IL-33, and IL-35) are also secreted during AD onset and progression, thereby preventing neuroinflammation. This review summarizes the involvement of pro- and anti-inflammatory cytokines in AD pathology and discusses their therapeutic potential.
Collapse
Affiliation(s)
- Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Yekkuni L. Balachandran
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Wai Po Chong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Kannie W. Y. Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Shenzhen Research Institute, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
41
|
Hudson JA, Ferrand RA, Gitau SN, Mureithi MW, Maffia P, Alam SR, Shah ASV. HIV-Associated Cardiovascular Disease Pathogenesis: An Emerging Understanding Through Imaging and Immunology. Circ Res 2024; 134:1546-1565. [PMID: 38781300 DOI: 10.1161/circresaha.124.323890] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Cardiac abnormalities were identified early in the epidemic of AIDS, predating the isolation and characterization of the etiologic agent, HIV. Several decades later, the causation and pathogenesis of cardiovascular disease (CVD) linked to HIV infection continue to be the focus of intense speculation. Before the widespread use of antiretroviral therapy, HIV-associated CVD was primarily characterized by HIV-associated cardiomyopathy linked to profound immunodeficiency. With increasing antiretroviral therapy use, viral load suppression, and establishment of immune competency, the effects of HIV on the cardiovascular system are more subtle. Yet, people living with HIV still face an increased incidence of cardiovascular pathology. Advances in cardiac imaging modalities and immunology have deepened our understanding of the pathogenesis of HIV-associated CVD. This review provides an overview of the pathogenesis of HIV-associated CVD integrating data from imaging and immunologic studies with particular relevance to the HIV population originating from high-endemic regions, such as sub-Saharan Africa. The review highlights key evidence gaps in the field and suggests future directions for research to better understand the complex HIV-CVD interactions.
Collapse
Affiliation(s)
- Jonathan A Hudson
- Kings College London BHF Centre, School of Cardiovascular and Metabolic Medicine & Sciences, United Kingdom (J.A.H.)
| | - Rashida A Ferrand
- Department of Clinical Research (R.A.F.), London School of Hygiene and Tropical Medicine, United Kingdom
- Biomedical Research and Training Institute, Harare, Zimbabwe (R.A.F.)
| | - Samuel N Gitau
- Department of Radiology, Aga Khan University Nairobi, Kenya (S.N.G.)
| | - Marianne Wanjiru Mureithi
- Department of Medical Microbiology and Immunology, Faculty of Health Sciences (M.W.M.), University of Nairobi, Kenya
| | - Pasquale Maffia
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (P.M.)
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Italy (P.M.)
- Africa-Europe Cluster of Research Excellence in Non-Communicable Diseases and Multimorbidity, African Research Universities Alliance and The Guild of European Research-Intensive Universities, Glasgow, United Kingdom (P.M.)
| | - Shirjel R Alam
- Department of Cardiology, North Bristol NHS Trust, United Kingdom (S.R.A.)
| | - Anoop S V Shah
- Department of Non-Communicable Disease Epidemiology (A.S.V.S.), London School of Hygiene and Tropical Medicine, United Kingdom
- Department of Cardiology, Imperial College NHS Trust, London, United Kingdom (A.S.V.S.)
| |
Collapse
|
42
|
Bai Y, Zhou R, Xie X, Zhu A, Nan Y, Wu T, Hu X, Cao Z, Ju D, Fan J. A Novel Bifunctional Fusion Protein (Anti-IL-17A-sST2) Protects against Acute Liver Failure, Modulating the TLR4/MyD88 Pathway and NLRP3 Inflammasome Activation. Biomedicines 2024; 12:1118. [PMID: 38791080 PMCID: PMC11117730 DOI: 10.3390/biomedicines12051118] [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: 03/24/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Acute liver failure (ALF) is a serious inflammatory disorder with high mortality rates, which poses a significant threat to human health. The IL-33/ST2 signal is a crucial regulator in inflammation responses associated with lipopolysaccharide (LPS)-induced macrophages. The IL-17A signaling pathway promotes the release of chemokines and inflammatory cytokines, recruiting neutrophils and T cells under LPS stimulation, thus facilitating inflammatory responses. Here, the potential therapeutic benefits of neutralizing the IL-17A signal and modulating the IL-33/ST2 signal in ALF were investigated. A novel dual-functional fusion protein, anti-IL-17A-sST2, was constructed, which displayed high purity and biological activities. The administration of anti-IL-17A-sST2 resulted in significant anti-inflammatory benefits in ALF mice, amelioration of hepatocyte necrosis and interstitial congestion, and reduction in TNF-α and IL-6. Furthermore, anti-IL-17A-sST2 injection downregulated the expression of TLR4 and NLRP3 as well as important molecules such as MyD88, caspase-1, and IL-1β. The results suggest that anti-IL-17A-sST2 reduced the secretion of inflammatory factors, attenuated the inflammatory response, and protected hepatic function by regulating the TLR4/MyD88 pathway and inhibiting the NLRP3 inflammasome, providing a new therapeutic approach for ALF.
Collapse
Affiliation(s)
- Yu Bai
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Rongrui Zhou
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xinlei Xie
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - An Zhu
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yanyang Nan
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tao Wu
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiaozhi Hu
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhonglian Cao
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dianwen Ju
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Fudan Zhangjiang Institute, Shanghai 201203, China
| | - Jiajun Fan
- Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Fudan Zhangjiang Institute, Shanghai 201203, China
- Shanghai Hailu Biological Technology Co., Ltd., Shanghai 201200, China
| |
Collapse
|
43
|
Shi T, Ge J, Li S, Zhang Y. Soluble suppression of tumorigenicity 2 associated with major adverse cardiac events in children with myocarditis. Front Cardiovasc Med 2024; 11:1404432. [PMID: 38807947 PMCID: PMC11130408 DOI: 10.3389/fcvm.2024.1404432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
Objective Soluble suppression of tumorigenicity 2 (sST2) is associated with the prognosis of some cardiac diseases, but studies on sST2 and the prognosis of patients with myocarditis are rare. This study investigated the relationship between major adverse cardiovascular events (MACEs) and sST2 during hospitalization in pediatric patients with myocarditis. Methods This was a single-center retrospective cohort study. A total of 252 patients aged ≤14 years diagnosed with myocarditis were enrolled. Events during the hospitalization were defined as MACEs (all-cause death > new heart failure > ventricular arrhythmia). Results A total of 25 people had MACEs during their hospital stay. The mortality during hospitalization was 6/23 (26%) in patients with heart failure and 3/10 (30%) in patients with ventricular arrhythmias. After including these risk factors in a multivariate logistic regression analysis, NT-proBNP (OR 4.323; 95% CI, 2.433-7.679; p < 0.001) and sST2 (OR 1.020; 95% CI, 1.003-1.037; p = 0.022) remained statistically significant and were independent risk factors for MACEs during hospitalization in pediatric myocarditis patients. Conclusions Elevated levels of NT-proBNP and sST2 were independently associated with major adverse cardiovascular events during hospitalization in children with myocarditis, and both showed good predictive efficacy.
Collapse
Affiliation(s)
- Tongtong Shi
- Department of Cardiology, The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ge
- Department of Clinical Nutrition, The Affiliated Huai'an Hospital of Xuzhou Medical University and Huai'an Second People’s Hospital, Huai'an, Jiangsu, China
| | - Shan Li
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and Huai'an Second People’s Hospital, Huai'an, Jiangsu, China
| | - Yali Zhang
- Department of Clinical Nutrition, The Affiliated Huai'an Hospital of Xuzhou Medical University and Huai'an Second People’s Hospital, Huai'an, Jiangsu, China
| |
Collapse
|
44
|
Wei BM, Fox LP, Kaffenberger BH, Korman AM, Micheletti RG, Mostaghimi A, Noe MH, Rosenbach M, Shinkai K, Kwah JH, Phillips EJ, Bolognia JL, Damsky W, Nelson CA. Drug-induced hypersensitivity syndrome/drug reaction with eosinophilia and systemic symptoms. Part I. Epidemiology, pathogenesis, clinicopathological features, and prognosis. J Am Acad Dermatol 2024; 90:885-908. [PMID: 37516359 DOI: 10.1016/j.jaad.2023.02.072] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/11/2023] [Accepted: 02/26/2023] [Indexed: 07/31/2023]
Abstract
Drug-induced hypersensitivity syndrome (DiHS), also known as drug reaction with eosinophilia and systemic symptoms (DRESS), is a severe cutaneous adverse reaction (SCAR) characterized by an exanthem, fever, and hematologic and visceral organ involvement. Anticonvulsants, antibiotics, and allopurinol are the most common triggers. The pathogenesis involves a complex interplay between drugs, viruses, and the immune system primarily mediated by T-cells. DiHS/DRESS typically presents with a morbilliform eruption 2-6 weeks after drug exposure, and is associated with significant morbidity, mortality, and risk of relapse. Long-term sequelae primarily relate to organ dysfunction and autoimmune diseases. Part I of this continuing medical education activity on DiHS/DRESS provides an update on epidemiology, novel insights into pathogenesis, and a description of clinicopathological features and prognosis.
Collapse
Affiliation(s)
- Brian M Wei
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Lindy P Fox
- Department of Dermatology, University of California, San Francisco, California
| | | | - Abraham M Korman
- Department of Dermatology, The Ohio State University, Columbus, Ohio
| | - Robert G Micheletti
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Arash Mostaghimi
- Department of Dermatology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Megan H Noe
- Department of Dermatology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Misha Rosenbach
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kanade Shinkai
- Department of Dermatology, University of California, San Francisco, California
| | - Jason H Kwah
- Department of Medicine, Section of Rheumatology, Allergy and Immunology, Yale School of Medicine, New Haven, Connecticut
| | - Elizabeth J Phillips
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jean L Bolognia
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut; Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Caroline A Nelson
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
45
|
Boulet J, Sridhar VS, Bouabdallaoui N, Tardif JC, White M. Inflammation in heart failure: pathophysiology and therapeutic strategies. Inflamm Res 2024; 73:709-723. [PMID: 38546848 PMCID: PMC11058911 DOI: 10.1007/s00011-023-01845-6] [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: 09/07/2023] [Revised: 11/09/2023] [Accepted: 12/19/2023] [Indexed: 04/30/2024] Open
Abstract
A role for inflammation in the development and progression of heart failure (HF) has been proposed for decades. Multiple studies have demonstrated the potential involvement of several groups of cytokines and chemokines in acute and chronic HF, though targeting these pathways in early therapeutic trials have produced mixed results. These studies served to highlight the complexity and nuances of how pro-inflammatory pathways contribute to the pathogenesis of HF. More recent investigations have highlighted how inflammation may play distinct roles based on HF syndrome phenotypes, findings that may guide the development of novel therapies. In this review, we propose a contemporary update on the role of inflammation mediated by the innate and adaptive immune systems with HF, highlighting differences that exist across the ejection fraction spectrum. This will specifically be looked at through the lens of established and novel biomarkers of inflammation. Subsequently, we review how improvements in inflammatory pathways may mediate clinical benefits of existing guideline-directed medical therapies for HF, as well as future therapies in the pipeline targeting HF and inflammation.
Collapse
Affiliation(s)
- Jacinthe Boulet
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Vikas S Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
| | - Nadia Bouabdallaoui
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Jean-Claude Tardif
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Michel White
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada.
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, QC, H1C 1C8, Montreal, Canada.
| |
Collapse
|
46
|
Rafaqat S, Radoman Vujacic I, Behnoush AH, Sharif S, Klisic A. Role of Cardiac Biomarkers in Hepatic Disorders: A Literature Review. Metab Syndr Relat Disord 2024; 22:251-262. [PMID: 38377607 DOI: 10.1089/met.2023.0282] [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] [Indexed: 02/22/2024] Open
Abstract
Various studies have reported the association between cardiac markers and hepatic disorders. The main objective of this review article was to elucidate the significance of important cardiac indicators such as ischemia-modified albumin, cardiac troponin, cardiac natriuretic peptides, creatine kinase, creatine kinase-MB, lactate dehydrogenase, heart-type fatty acid-binding protein, osteopontin, soluble suppression of tumorigenicity 2, C-reactive protein, and lipoprotein(a) in the development of hepatic disorders. In addition, it highlighted recent notable discoveries and accomplishments in this field and identified areas requiring further investigation, ongoing discussions, and potential avenues for future research. Early identification and control of these cardiac markers might be helpful to control the prevalence of hepatic disorders associated with cardiovascular diseases.
Collapse
Affiliation(s)
- Saira Rafaqat
- Department of Zoology (Molecular Physiology), Lahore College for Women University, Lahore, Pakistan
| | - Irena Radoman Vujacic
- Department of Internal Medicine, Clinical Center of Montenegro, University of Montenegro-Faculty of Medicine, Podgorica, Montenegro
| | | | - Saima Sharif
- Department of Zoology (Molecular Physiology), Lahore College for Women University, Lahore, Pakistan
| | - Aleksandra Klisic
- University of Montenegro-Faculty of Medicine, Podgorica, Montenegro
- Center for Laboratory Diagnostics, Primary Health Care Center, Podgorica, Montenegro
| |
Collapse
|
47
|
Lazar-Poloczek E, Romuk E, Jacheć W, Wróbel-Nowicka K, Świętek A, Wojciechowska C. Association of NT-proBNP and sST2 with Left Ventricular Ejection Fraction and Oxidative Stress in Patients with Stable Dilated Cardiomyopathy. Biomedicines 2024; 12:707. [PMID: 38672063 PMCID: PMC11048168 DOI: 10.3390/biomedicines12040707] [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: 12/27/2023] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
The aim of this study was to analyze the relationship between levels of sST2, NT-proBNP and oxidative stress markers in patients with reduced ejection fraction (HFrEF) due to non-ischemic cardiomyopathy. A total of 88 patients with HFrEF were divided into four groups based on left ventricular ejection fraction (≤25% and >25%) and NYHA functional class (group 1-LVEF > 25% and NYHA class I or II; group 2-LVEF > 25% and NYHA class III or IV; group III-LVEF ≤ 25% and NYHA class I or II; group IV-LVEF ≤ 25% and NYHA class III or IV). In 39 (44.32%) patients LVEF was reduced below 25%, and 22 of them (56.41%) were in NYHA functional class III/IV. Of the 49 (55.68%) patients with LVEF ≥ 25%, only 18.37% were in NYHA functional class III/IV (p < 0.001). Patients with LVEF ≥ 25% had lower levels of NT-proBNP, total oxidant status (TOS), total antioxidant capacity (TAC), and oxidative stress index (OSI). The levels of NT-proBNP but not sST-2 correlated positively with NYHA functional class (p < 0.001) and negatively with LVEF (p < 0.001). The levels of sST-2 were associated with increased TAC (p = 0.009) and uric acid (p = 0.040). These findings indicate that only NT-proBNP was related to the severity of heart failure, whereas sST2 correlated with total antioxidant capacity. Therefore, in stable patients with HFrEF due to dilated cardiomyopathy, sST2 may be an additional biomarker reflecting the redox status, but not the severity of heart failure.
Collapse
Affiliation(s)
- Elżbieta Lazar-Poloczek
- 2nd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10, M.C-Skłodowska St., 41-800 Zabrze, Poland; (E.L.-P.); (W.J.)
| | - Ewa Romuk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19, Jordan St., 41-808 Zabrze, Poland;
| | - Wojciech Jacheć
- 2nd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10, M.C-Skłodowska St., 41-800 Zabrze, Poland; (E.L.-P.); (W.J.)
| | - Karolina Wróbel-Nowicka
- Medical Laboratory in Specialistic Hospital in Zabrze, 10, M.C-Skłodowska St., 41-800 Zabrze, Poland;
| | - Agata Świętek
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19, Jordan St., 41-808 Zabrze, Poland;
- Silesia LabMed Research and Implementation Center, Medical University of Silesia in Katowice, 19, Jordan St., 41-808 Zabrze, Poland
| | - Celina Wojciechowska
- 2nd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10, M.C-Skłodowska St., 41-800 Zabrze, Poland; (E.L.-P.); (W.J.)
| |
Collapse
|
48
|
Lai J, Huang C, Li B, Han Y. Soluble ST2 as a possible biomarker for inflammation in patients with acute heart failure. J Cardiovasc Med (Hagerstown) 2024; 25:186-192. [PMID: 38305120 DOI: 10.2459/jcm.0000000000001587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
AIM The aim of this study was to explore the relationship between peripheral circulating serum soluble suppression of tumorigenicity-2 (sST2) levels and inflammatory biomarkers in patients with acute heart failure (AHF). METHODS One hundred and eleven consecutive AHF patients with NYHA class II-IV were enrolled, and peripheral blood was collected within 24 h of admission for the detection of NT-ProBNP, sST2, hypersensitive troponin I, cytokines, precalcitoninogen, C-reactive protein, in addition to routine standard of care blood tests. RESULTS The median sST2 of 111 patients was 47.50 ng/ml (24.25-86.15 IQR), of whom 43 patients (38.7%) had sST2 35 ng/ml or less; linear correlation analysis showed that serum sST2 correlated with NT-ProBNP ( r2 = 0.32), NEU% ( r2 = 0.41), NLR ( r2 = 0.36), CRP ( r2 = 0.50), IL-18 ( r2 = 0.43) ( P < 0.001), and correlated with Hs-cTnI ( r2 = 0.19), NUE ( r2 = 0.25), LYM ( r2 = -0.23), IL-2RA ( r2 = 0.29) ( P < 0.05). Multiple linear regression analysis depicted that CRP (β = 0.318), IL-18 (β = 0.368), NEU% (β = 0.346), NLR (β = -0.304), and NT-ProBNP (β = 0.324) significantly correlated with sST2 values, respectively ( P < 0.05). ST2 levels have a linear association with length of hospitalization. CONCLUSION Peripheral blood inflammatory markers (CRP, IL-18, NEU%, NLR) in patients with AHF had a close relationship with sST2 levels, and the mechanism of action of sST2 may be related to the inflammatory response.
Collapse
Affiliation(s)
- Jiacheng Lai
- Department of Emergency Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei
- Emergency and Trauma Center, The International Medical Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou
| | - Chongjian Huang
- Department of Emergency Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei
- Department of Emergency Medicine, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Bin Li
- Department of Emergency Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei
| | - Yongsheng Han
- Department of Emergency Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei
| |
Collapse
|
49
|
Isailă OM, Moroianu LA, Hostiuc S. Current Trends in Biohumoral Screening for the Risk of Sudden Cardiac Death: A Systematic Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:418. [PMID: 38541144 PMCID: PMC10972295 DOI: 10.3390/medicina60030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 07/23/2024]
Abstract
Background and Objectives: Sudden cardiac death (SCD) represents a challenge to health systems globally and is met with increased frequency in the population. Over time, multiple screening methods have been proposed, including the analysis of various plasma biomarkers. This article aims to analyze for illustrative purposes the specialized literature in terms of current biomarkers and testing trends, in the case of cardiovascular diseases and implicitly sudden cardiac death. Materials and Methods: In this regard, we searched the PubMed database from 2010 to the present time using the keywords "sudden cardiac death" and "biomarkers". The inclusion criteria were clinical trials that analyzed the effectiveness of screening methods in terms of biomarkers used in stratifying the risk of cardiac distress and/or sudden cardiac death. We excluded reviews, meta-analyses, and studies looking at the effectiveness of treatments. Results: An extended approach was found, through studies that brought to the forefront both classical markers analyzed by new, more performant methods, markers for other pathologies that also determined cardiovascular impact, non-specific molecules with effects on the cardiovascular system, and state-of-the-art markers, such as microRNA. Some molecules were analyzed simultaneously in certain groups of patients. Conclusion: The observed current trend revealed the tendency to define the clinical-biological particularities of the person to be screened.
Collapse
Affiliation(s)
- Oana-Maria Isailă
- Department of Legal Medicine and Bioethics, Faculty of Dentistry, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Lavinia-Alexandra Moroianu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 47 Domneasca Street, 800008 Galati, Romania;
| | - Sorin Hostiuc
- Department of Legal Medicine and Bioethics, Faculty of Dentistry, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
50
|
Pimpalwar N, Celik S, Karbalaei Sadegh M, Czuba T, Gidlöf O, Smith JG. Analysis of genetic variant associated with heart failure mortality implicates thymic stromal lymphopoietin as mediator of strain-induced myocardial fibroblast-mast cell crosstalk and fibrosis. FASEB J 2024; 38:e23510. [PMID: 38407489 DOI: 10.1096/fj.202302000rr] [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: 09/30/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024]
Abstract
Heart failure (HF) is a leading cause of death and disability globally. Heritable factors and the extent and pattern of myocardial fibrosis are important determinants of outcomes in patients with HF. In a genome-wide association study of mortality in HF, we recently identified a genetic polymorphism on chromosome 5q22 associated with HF mortality. Here, we sought to study the mechanisms by which this variant may influence myocardial disease processes. We find that the risk allele is located in an enhancer motif upstream of the TSLP gene (encoding thymic stromal lymphopoietin), conferring increased binding of the transcription factor nescient helix-loop helix 1 (NHLH1) and increased TSLP expression in human heart. Further, we find that increased strain of primary human myocardial fibroblasts results in increased TSLP expression and that the TSLP receptor is expressed in myocardial mast cells in human single nuclei RNA sequence data. Finally, we show that TSLP overexpression induces increased transforming growth factor β expression in myocardial mast cells and tissue fibrosis. Collectively, our findings based on follow-up of a human genetic finding implicate a novel pathway in myocardial tissue homeostasis and remodeling.
Collapse
Affiliation(s)
- Neha Pimpalwar
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Selvi Celik
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Mardjaneh Karbalaei Sadegh
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Tomasz Czuba
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Olof Gidlöf
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Heart Failure and Valvular Disease, Skåne University Hospital, Lund, Sweden
| |
Collapse
|