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
|
Li X, Tian Y, Cao H, Cheng J. Serum sST2: key biomarkers in COVID-19 patients with implications for coronary artery disease. BMC Infect Dis 2025; 25:471. [PMID: 40197291 PMCID: PMC11974224 DOI: 10.1186/s12879-025-10849-y] [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: 06/04/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND As the coronavirus disease-2019 (COVID-19) pandemic persists, post-COVID-19 syndrome (PS), characterized by symptoms like chest pain, fatigue, and palpitations, is becoming a significant medical and social issue. COVID-19 patients with existing coronary artery disease (CAD) may face higher risks of complications. It is crucial to assess if PS patients also have CAD, though data is limited. METHODS We studied 75 COVID-19 patients and 68 non-COVID-19 patients admitted to our hospital between 2022/12/20 to 2023/01/20. Demographic, laboratory, and clinical data were collected upon admission. The Gensini score (GS) was used to assess coronary atherosclerosis severity. Patients were categorized by GS and clinical traits to identify potential independent risks linked to CAD and COVID-19 severity. RESULTS COVID-19 patients with existing CAD had higher levels of serum soluble growth stimulation expression of gene 2 protein (sST2), myeloperoxidase, ALT, AST, PT, B-type natriuretic peptide (BNP), and hypersensitive troponin-I (hs-cTnI), along with longer hospital stays, more ICU admissions, and increased heart failure and ACS morbidity compared to those without CAD. Univariate and multivariate analysis identified sST2 as an independent risk factor for COVID-19 patients with coexisting CAD (odds ratio 1.122). sST2 levels were positively correlated with coronary angiography GS (r = 0.474, p < 0.001) in COVID-19 patients and were significantly higher in cases with GS ≥ 32, regardless of COVID-19 status (p < 0.001) and specifically in COVID-19 patients (p = 0.006). ROC analysis showed sST2 predicted ICU admission, hospital stay duration, and morbidity of HF and ACS similarly to GS. CONCLUSIONS Admission serum sST2 levels should be considered in COVID-19 patients with CAD-like symptoms for treatment planning and could serve as a prognostic biomarker for COVID-19 with co-existing CAD in clinical practice.
Collapse
Affiliation(s)
- Xueqin Li
- Department of Laboratory Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yaxin Tian
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
- Department of Health Statistics, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Hongyan Cao
- Department of Health Statistics, Shanxi Provincial Key Laboratory of Major Diseases Risk Assessment, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Jinfang Cheng
- Department of Cardiovascular Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyaun, 030032, China.
| |
Collapse
|
3
|
Akenroye A, Boyce JA, Kita H. Targeting alarmins in asthma: From bench to clinic. J Allergy Clin Immunol 2025; 155:1133-1148. [PMID: 39855362 DOI: 10.1016/j.jaci.2025.01.017] [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: 09/20/2024] [Revised: 12/24/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Over the past 2 decades, mechanistic studies of allergic and type 2 (T2)-mediated airway inflammation have led to multiple approved therapies for the treatment of moderate-to-severe asthma. The approval and availability of these monoclonal antibodies targeting IgE, a T2 cytokine (IL-5) and/or cytokine receptors (IL-5Rα, IL-4Rα) has been central to the progresses made in the management of moderate-to-severe asthma over this period. However, there are persistent gaps in clinician's ability to provide precise care, given that many patients with T2-high asthma do not respond to IgE- or T2 cytokine-targeting therapies and that patients with T2-low asthma have few therapeutic options. The new frontier of precision medicine in asthma, as well as in other allergic diseases, includes the targeting of epithelium-derived cytokines known as alarmins, including thymic stromal lymphopoietin, IL-25, IL-33, and their receptors. The effects of these alarmins, which can act upstream of immune cells, involve both the innate and adaptive systems and hold potential for the treatment of both T2-high and -low disease. Tezepelumab, an anti-thymic stromal lymphopoietin antibody, has already been approved for the treatment of severe asthma. In this review, we discuss our current understanding of alarmin biology with a primary focus on allergic airway diseases. We link the mechanistic corollaries to the clinical implications and advances in drug development targeting alarmins, with a particular focus on currently approved treatments, those under study, and future potential targets in alarmin signaling pathways.
Collapse
Affiliation(s)
- Ayobami Akenroye
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| | - Joshua A Boyce
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, the Department of Medicine, and the Department of Immunology, Mayo Clinic Arizona, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn
| |
Collapse
|
4
|
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
|
5
|
Bellos I, Marinaki S, Lagiou P, Benetou V. Association of soluble suppression of tumorigenicity 2 with mortality and adverse outcomes in chronic kidney disease: a systematic review and meta-analysis. Clin Exp Nephrol 2024; 28:988-1003. [PMID: 38678167 PMCID: PMC11493800 DOI: 10.1007/s10157-024-02506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Early risk stratification is necessary to prevent chronic kidney disease progression and complications. This systematic review aims to evaluate the association of soluble suppression of tumorigenicity 2 (sST2), a member of the interleukin-1 receptor family, with all-cause mortality, cardiovascular disease and renal function deterioration among chronic kidney disease patients. METHODS PubMed, Scopus, Web of Science, CENTRAL and Google Scholar were systematically searched from inception to December 20, 2023. Cohort studies examining the prognostic role of sST2 levels in pre-dialysis and dialysis patients were included. In case of 3 or more studies per outcome, conventional and dose-response meta-analyses were conducted. RESULTS Overall, 21 studies were included comprising 15,100 patients. In pre-dialysis patients, the qualitative synthesis of studies suggested that high sST2 is associated with significantly increased all-cause mortality, while evidence regarding cardiovascular events or kidney disease progression was conflicting. In the dialysis population, high sST2 was linked to an elevated risk of all-cause (Hazard ratio-HR: 3.00, 95% confidence intervals-CI: 1.95-4.61) and cardiovascular (HR: 2.38, 95% CI: 1.69-3.34) mortality. Dose-response meta-analysis suggested a log-linear association of sST2 with both all-cause (χ2: 34.65, p value < 0.001) and cardiovascular (χ2: 29.14, p value < 0.001) mortality, whereas findings regarding cardiovascular events were limited with mixed results. CONCLUSIONS High sST2 values are associated with an increased risk of all-cause mortality in pre-dialysis and dialysis patients, as well as with an elevated risk of cardiovascular mortality in the dialysis population. Further studies are needed to elucidate its potential association with cardiovascular events and kidney disease progression.
Collapse
Affiliation(s)
- Ioannis Bellos
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece.
- Department of Nephrology and Renal Transplantation, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Smaragdi Marinaki
- Department of Nephrology and Renal Transplantation, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece
| | - Vassiliki Benetou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece
| |
Collapse
|
6
|
Mathias K, Machado RS, Tiscoski ADB, Dos Santos D, Lippert FW, Costa MA, Gonçalves CL, Generoso JS, Prophiro JS, Giustina AD, Petronilho F. IL-33 in Ischemic Stroke: Brain vs. Periphery. Inflammation 2024:10.1007/s10753-024-02148-6. [PMID: 39294293 DOI: 10.1007/s10753-024-02148-6] [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: 05/14/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/20/2024]
Abstract
Cerebrovascular disease is the second-leading cause of death and disability worldwide, with stroke being the most common cause. In ischemic stroke, several processes combine to produce immunosuppression, leaving the post-stroke body susceptible to infection, which in turn affects neuroinflammation. Interleukin-33 (IL-33), a member of the interleukin-1 family (IL-1), functions as a modulator of immune responses and inflammation, playing a crucial role in the establishment of immunologic responses. IL-33 has been shown to have a protective effect on brain injury and represents a potential target by modulating inflammatory cytokines and stimulating immune regulatory cells. With an emphasis on preclinical and clinical studies, this review covers the impact of IL-33 on immune system mechanisms following ischemic stroke.
Collapse
Affiliation(s)
- Khiany Mathias
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
- Health Sciences Unit, Program in Health Sciences, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Richard Simon Machado
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
- Health Sciences Unit, Program in Health Sciences, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Anita Dal Bó Tiscoski
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - David Dos Santos
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Fabricio Weinheimer Lippert
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Maiara Aguiar Costa
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Cinara Ludvig Gonçalves
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Jaqueline Silva Generoso
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Josiane Somariva Prophiro
- Health Sciences Unit, Program in Health Sciences, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Amanda Della Giustina
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, ON, Canada
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Health Sciences Unit, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil.
| |
Collapse
|
7
|
Gizatulina TP, Mamarina AV, Martyanova LU, Belonogov DV, Kolunin GV, Petelina TI, Shirokov NE, Gorbatenko EA. The soluble ST2 level predicts risk of atrial fibrillation recurrences in long-term period after radiofrequency ablation. BMC Cardiovasc Disord 2024; 24:460. [PMID: 39198735 PMCID: PMC11350941 DOI: 10.1186/s12872-024-04119-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: 03/29/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The hypothesis of the study was the assumption that the serum levels of soluble ST2 (sST2) and growth differentiation factor (GDF-15) can be predictors of atrial fibrillation (AF) recurrence in long-term period after primary radiofrequency catheter ablation (RFA). METHODS Of the 165 patients included in the prospective follow-up, the final analysis included 131 patients whose follow-up duration reached 18 months after the end of the blanking period (3 months after RFA). The median age of patients was 59.0 (50.0; 64.0) years, and 80 (61%) were men. Paroxysmal AF was present in 103 (79%) and persistent AF in 28 (21%) patients. All patients underwent transthoracic and transesophageal echocardiography, and electroanatomic mapping was used to assess the area of low-voltage zones (LVZ). sST2 and GDF-15 levels were determined by ELISA using GDF-15/MIC-1 analytical kits (BioVender, Czech Republic) and Presage ST2 (Critical Diagnostics, USA) before RFA. After RFA, patients had regular follow-up visits at 3-6-9-12-18 months with 12-lead ECG or Holter ECG monitoring and with clinical evaluation. The primary endpoint was the occurrence of the first symptomatic AF recurrence (AFr) lasting > 30 s, recorded on an ECG or during daily ECG monitoring, after a blanking period. RESULTS At the 18-month follow-up, 47 patients (35.9%) had AFr. The groups with and without AFr didn`t differ in the LVZ area. The medians of NT-proBNP, GDF-15 and sST2 also didn`t differ significantly between the groups, but in patients with AFr, the proportion of those with sST2 ≥ 36 ng/ml (the border of the lower and middle terziles) was higher (p = 0.03). According to the one-factor Cox regression analysis, AFr were associated with four factors: AF history ≥ 1 year, early AFr (during the blanking period), left atrial appendage flow velocity (LAAFV) < 54 cm/sec and sST2 ≥ 36 ng/ml. In the multivariate Cox analysis two independent predictors of AFr were obtained: sST2 ≥ 36 ng/ml (HR = 3.8; 95% CI 1.5-9.8, p = 0.006) and LAAFV < 54 сm/sec (HR = 1.96; 95% CI 1.01-3.82, p = 0.048). CONCLUSIONS Serum sST2 level with a cut-off value of 36 ng/ml or more can be used as a predictor of AF recurrence in the long-term period after primary RFA.
Collapse
Affiliation(s)
- Tatiana P Gizatulina
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation.
| | - Aleksandra V Mamarina
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation
| | - Leysan U Martyanova
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation
| | - Denis V Belonogov
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation
| | - Grigoriy V Kolunin
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation
| | - Tatiana I Petelina
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation
| | - Nikita E Shirokov
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation
| | - Elena A Gorbatenko
- Tyumen Cardiology Research Center, Tomsk National Research Medical Center of the Russian Academy of Sciences, 111 Melnikaite Street, Tyumen, 625026, Russian Federation
| |
Collapse
|
8
|
Clausen H, Friberg E, Lannering K, Koivu A, Sairanen M, Mellander M, Liuba P. Newborn Screening for High-Risk Congenital Heart Disease by Dried Blood Spot Biomarker Analysis. JAMA Netw Open 2024; 7:e2418097. [PMID: 38913376 PMCID: PMC11197454 DOI: 10.1001/jamanetworkopen.2024.18097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/20/2024] [Indexed: 06/25/2024] Open
Abstract
Importance Congenital heart disease (CHD) is the most common human organ malformation, affecting approximately 1 of 125 newborns globally. Objectives Assessing the performance of 2 diagnostic tests using minimal amounts of dried blood spots (DBS) to identify high-risk CHD compared with controls in a Swedish cohort of neonates. Design, Setting, and Participants This diagnostic study took place in Sweden between 2019 and 2023 and enrolled full-term babies born between 2005 and 2023. All cases were identified through centralized pediatric cardiothoracic surgical services in Lund and Gothenburg, Sweden. Controls were followed up for 1 year to ensure no late presentations of high-risk CHD occurred. Cases were verified through surgical records and echocardiography. Exposure High-risk CHD, defined as cases requiring cardiac surgical management during infancy due to evolving signs of heart failure or types in which the postnatal circulation depends on patency of the arterial duct. Using 3-μL DBS samples, automated quantitative tests for NT-proBNP and interleukin 1 receptor-like 1 (IL-1 RL1; formerly known as soluble ST2) were compared against established CHD screening methods. Main Outcomes and Measures Performance of DBS tests to detect high-risk CHD using receiver operating characteristic curves; Bland-Altman and Pearson correlation analyses to compare IL-1 RL1 DBS with plasma blood levels. Results A total of 313 newborns were included (mean [SD] gestational age, 39.4 [1.3] weeks; 181 [57.8%] male). Mean (SD) birthweight was 3495 (483) grams. Analyzed DBS samples included 217 CHD cases and 96 controls. Among the CHD cases, 188 participants (89.3%) were high-risk types, of which 73 (38.8%) were suspected prenatally. Of the 188 high-risk cases, 94 (50.0%) passed pulse oximetry screening and 36 (19.1%) were initially discharged after birth without diagnoses. Combining NT-proBNP and IL-1 RL1 tests performed well in comparison with existing screening methods and enabled additional identification of asymptomatic babies with receiver operating characteristic area under the curve 0.95 (95% CI, 0.93-0.98). Conclusions and relevance In this diagnostic study, NT-proBNP and IL-1 RL1 DBS assays identified high-risk CHD in a timely manner, including in asymptomatic newborns, and improved overall screening performance in this cohort from Sweden. Prospective evaluation of this novel approach is warranted.
Collapse
Affiliation(s)
- Henning Clausen
- Medical Faculty, Lund University, Sweden
- Children’s Heart Centre, Skane’s University Hospital, Lund, Sweden
| | - Elin Friberg
- Medical Faculty, Lund University, Sweden
- Children’s Heart Centre, Skane’s University Hospital, Lund, Sweden
| | - Katarina Lannering
- Medical Faculty, Gothenburg University, Gothenburg, Sweden
- Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Aki Koivu
- Revvity, Diagnostics Research & Development, Turku, Finland
| | - Mikko Sairanen
- Revvity, Diagnostics Research & Development, Turku, Finland
| | - Mats Mellander
- Medical Faculty, Gothenburg University, Gothenburg, Sweden
- Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Petru Liuba
- Medical Faculty, Lund University, Sweden
- Children’s Heart Centre, Skane’s University Hospital, Lund, Sweden
| |
Collapse
|
9
|
Cheng TY, Chen YC, Li SJ, Lin FJ, Lu YY, Lee TI, Lee TW, Higa S, Kao YH, Chen YJ. Interleukin-33/ST2 axis involvement in atrial remodeling and arrhythmogenesis. Transl Res 2024; 268:1-12. [PMID: 38244770 DOI: 10.1016/j.trsl.2024.01.006] [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: 09/25/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
Interleukin (IL)-33, a cytokine involved in immune responses, can activate its receptor, suppression of tumorigenicity 2 (ST2), is elevated during atrial fibrillation (AF). However, the role of IL-33/ST2 signaling in atrial arrhythmia is unclear. This study explored the pathological effects of the IL-33/ST2 axis on atrial remodeling and arrhythmogenesis. Patch clamping, confocal microscopy, and Western blotting were used to analyze the electrical characteristics of and protein activity in atrial myocytes (HL-1) treated with recombinant IL-33 protein and/or ST2-neutralizing antibodies for 48 hrs. Telemetric electrocardiographic recordings, Masson's trichrome staining, and immunohistochemistry staining of the atrium were performed in mice receiving tail vein injections with nonspecific immunoglobulin (control), IL-33, and IL-33 combined with anti-ST2 antibody for 2 weeks. IL-33-treated HL-1 cells had a reduced action potential duration, lower L-type Ca2+ current, greater sarcoplasmic reticulum (SR) Ca2+ content, increased Na+/Ca2+ exchanger (NCX) current, elevation of K+ currents, and increased intracellular calcium transient. IL-33-treated HL-1 myocytes had greater activation of the calcium-calmodulin-dependent protein kinase II (CaMKII)/ryanodine receptor 2 (RyR2) axis and nuclear factor kappa B (NF-κB) / NLR family pyrin domain containing 3 (NLRP3) signaling than did control cells. IL-33 treated cells also had greater expression of Nav1.5, Kv1.5, NCX, and NLRP3 than did control cells. Pretreatment with neutralizing anti-ST2 antibody attenuated IL-33-mediated activation of CaMKII/RyR2 and NF-κB/NLRP3 signaling. IL-33-injected mice had more atrial ectopic beats and increased AF episodes, greater atrial fibrosis, and elevation of NF-κB/NLRP3 signaling than did controls or mice treated with IL-33 combined with anti-ST2 antibody. Thus, IL-33 recombinant protein treatment promotes atrial remodeling through ST2 signaling. Blocking the IL-33/ST2 axis might be an innovative therapeutic approach for patients with atrial arrhythmia and elevated serum IL-33.
Collapse
Affiliation(s)
- Tzu-Yu Cheng
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Shao-Jung Li
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Fong-Jhih Lin
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan; School of Medicine, Fu-Jen Catholic University, New Taipei City 24257, Taiwan
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, 1199 Makiminato, Urasoe City, Okinawa 901-2131, Japan
| | - Yu-Hsun Kao
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan; Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan.
| |
Collapse
|
10
|
Zeng Z, Li W, Zhang J, Hu Z, Wu J, Ye G, Luo Y. Highly sensitive and specific graphene oxide-based FRET aptasensor for quantitative detection of human soluble growth stimulating gene protein 2. Talanta 2024; 271:125629. [PMID: 38245955 DOI: 10.1016/j.talanta.2024.125629] [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: 08/12/2023] [Revised: 12/31/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024]
Abstract
Soluble growth stimulation expressed gene 2 (sST2) is a new generation biomarker in the diagnosis and prognosis of heart failure (HF). Here, the sST2-specific aptamers were selected from a random ssDNA library with the full length of 88 nucleotides (nt) via target-immobilized magnetic beads (MB)-based systematic evolution of ligands by exponential enrichment (SELEX) technology. After eight rounds of selection, six aptamers with the most enrichment were selected. Among, the aptamer L1 showed the high-affinity binding to sST2 with the lowest Kd value (77.3 ± 0.05 nM), which was chosen as the optimal aptamer for further molecular docking. Then, the aptamer L1 was used to construct a graphene oxide (GO) - based fluorescence resonance energy transfer (FRET) biosensor for sST2, which exhibits a linear detection range of 0.1-100 μg/ml and a detection limit of 3.7 ng/ml. The aptasensor was applied to detect sST2 in real samples, with a good correlation and agreement with the traditional enzyme-linked immunosorbent assay (ELISA) when quantitative analyzing the sST2 concentration in serum samples from HF patients. The results show that not only an efficient strategy for screening the practicable aptamer, but also a rapid and sensitive detection platform for sST2 were established.
Collapse
Affiliation(s)
- Zhikun Zeng
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Wenfeng Li
- The Second Clinical College of Wuhan University, Wuhan, 430071, Hubei, China
| | - Jixuan Zhang
- The Second Clinical College of Wuhan University, Wuhan, 430071, Hubei, China
| | - Zijian Hu
- The First Clinical College of Wuhan University, Wuhan, 430060, Hubei, China
| | - Junyi Wu
- The Second Clinical College of Wuhan University, Wuhan, 430071, Hubei, China
| | - Guangming Ye
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Yi Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
11
|
Thazhathveettil J, Kumawat AK, Demirel I, Sirsjö A, Paramel GV. Vascular smooth muscle cells in response to cholesterol crystals modulates inflammatory cytokines release and promotes neutrophil extracellular trap formation. Mol Med 2024; 30:42. [PMID: 38519881 PMCID: PMC10960408 DOI: 10.1186/s10020-024-00809-8] [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: 12/03/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The formation and accumulation of cholesterol crystals (CC) at the lesion site is a hallmark of atherosclerosis. Although studies have shown the importance of vascular smooth muscle cells (VSMCs) in the disease atherosclerosis, little is known about the molecular mechanism behind the uptake of CC in VSMCs and their role in modulating immune response. METHODS Human aortic smooth muscle cells were cultured and treated with CC. CC uptake and CC mediated signaling pathway and protein induction were studied using flow cytometry, confocal microscopy, western blot and Olink proteomics. Conditioned medium from CC treated VSMCs was used to study neutrophil adhesion, ROS production and phagocytosis. Neutrophil extracellular traps (NETs) formations were visualized using confocal microscopy. RESULTS VSMCs and macrophages were found around CC clefts in human carotid plaques. CC uptake in VSMCs are largely through micropinocytosis and phagocytosis via PI3K-AkT dependent pathway. The uptake of CC in VSMCs induce the release inflammatory proteins, including IL-33, an alarming cytokine. Conditioned medium from CC treated VSMCs can induce neutrophil adhesion, neutrophil reactive oxygen species (ROS) and neutrophil extracellular traps (NETs) formation. IL-33 neutralization in conditioned medium from CC treated VSMCs inhibited neutrophil ROS production and NETs formation. CONCLUSION We demonstrate that VSMCs due to its vicinity to CC clefts in human atherosclerotic lesion can modulate local immune response and we further reveal that the interaction between CC and VSMCs impart an inflammatory milieu in the atherosclerotic microenvironment by promoting IL-33 dependent neutrophil influx and NETs formation.
Collapse
Affiliation(s)
- Jishamol Thazhathveettil
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden
- School of Medical Sciences, Örebro University, 70182, Örebro, Sweden
| | - Ashok Kumar Kumawat
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden
- School of Medical Sciences, Örebro University, 70182, Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Örebro University, 70182, Örebro, Sweden
| | - Allan Sirsjö
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden
- School of Medical Sciences, Örebro University, 70182, Örebro, Sweden
| | - Geena Varghese Paramel
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 70182, Örebro, Sweden.
- School of Medical Sciences, Örebro University, 70182, Örebro, Sweden.
| |
Collapse
|
12
|
Liu Y, Gao L, Zhao G, Zhang W, Du C, Sun W, Jin L, Lu H, Zhou H. Variations in serum low-density lipoprotein and sST2 among heart failure patients with different ejection fraction groups and their clinical significance. Medicine (Baltimore) 2024; 103:e37357. [PMID: 38428890 PMCID: PMC10906588 DOI: 10.1097/md.0000000000037357] [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: 11/29/2023] [Accepted: 02/02/2024] [Indexed: 03/03/2024] Open
Abstract
OBJECTIVE This study aimed to examine the changes in serum Low Density Lipoprotein Cholesterol (LDL-C) and Soluble Growth Stimulating Expressed Gene 2 Protein (sST2) among Heart Failure (HF) patients with varying ejection fractions and their clinical significance, providing a reference for the clinical assessment of HF severity. METHODS A total of 238 HF patients treated in our hospital's cardiology department from September 2019 to December 2021 were selected; 68 patients hospitalized in the same period were selected as the control group. General information, LDL-C and echocardiographic results of admitted patients were collected. According to LVEF results and the latest European Society of Cardiology standards in 2021, HF patients were categorized into those with HFpEF (n = 95), HFmrEF (n = 60), and HFrEF (n = 83). Meanwhile, venous blood was collected to determine sST2 and NT-proBNP to compare and analyze the changes and clinical significance of sST2 and LDL-C across the groups. RESULTS Compared to the control group, the HF group showed significant differences in age, gender, heart rate, smoking history, history of atrial fibrillation, history of diabetes, LVEDD, LVEF, sST2, and NT-proBNP levels (P < .05), but not in LDL-C levels. Significant differences (P < .05) were also found among the 3 HF groups in terms of age, gender, history of atrial fibrillation, LVEDD, LVEF, LDL-C, sST2, and NT-proBNP levels, with an increase in LVEDD, LDL-C, sST2, and NT-proBNP values as the ejection fraction decreased. ROC curve analysis indicated that the area under the curve (AUC) for sST2 in diagnosing HF was 0.915 (P < .05), with an optimal cutoff value of 23.71 ng/mL, a sensitivity of 76.5%, and a specificity of 95.6%; LDL-C was not a significant diagnostic marker for HF (P > .05). Coronary artery disease, NT-proBNP, and sST2 were identified as risk factors for HF. With each unit increase in coronary artery disease, the risk of HF increased by 36.3%; for NT-proBNP, the risk increased by 1.3% per unit; and for sST2, it increased by 18.3% per unit. CONCLUSION As the ejection fraction decreases in HF patients, serum sST2 and LDL-C values progressively increase, which is clinically significant for predicting the severity of HF. sST2 is an independent risk factor for HF and can enhance the diagnostic accuracy for HF.
Collapse
Affiliation(s)
- Yuanzhi Liu
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - Lijian Gao
- Coronary Artery Disease Center, Department of Cardiology, Fuwai Hospital, CAMS and PUMC, Beijing, China
| | - Guangxian Zhao
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - Wenchen Zhang
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - Chuan Du
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - Wenjing Sun
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - Lei Jin
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - Hongyu Lu
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - He Zhou
- Department of Cardiology, Yanbian University Hospital, Yanji, China
| |
Collapse
|
13
|
Chen P, Zhang J, Du J, Shi D, Zhang H. Predictive value of soluble suppression of tumorigenicity 2 in atrial fibrillation: a systematic review and meta-analysis. Front Cardiovasc Med 2024; 10:1308166. [PMID: 38274310 PMCID: PMC10808625 DOI: 10.3389/fcvm.2023.1308166] [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: 10/14/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Purpose Atrial fibrosis is the main pathological basis for the pathogenesis and progression of atrial fibrillation (AF). Soluble suppression of tumorigenicity 2 (sST2) is involved in fibrosis. Recent studies have explored its predictive value in AF outcomes. We performed this study to assess whether sST2 is an independent biomarker of AF outcomes and explore the potential mechanism. Methods PubMed, Web of Science, EMBASE, and Cochrane Library databases were searched systematically from inception through July 1, 2023, to identify relevant studies. Outcomes of interest included occurrence, recurrence, and major adverse cardiac events (MACEs) of AF. This meta-analysis was reported following the criteria outlined in PRISMA 2020, and the protocol was registered in PROSPERO (number: CRD42023459789). All statistical analyses were performed using the STATA version 16. Result Twenty four studies with 14,755 patients were included in the meta-analysis. The meta-analyses found that sST2 was significantly associated with the risk of occurrence [HR:1.04, 95% CI: 1.02-1.07, P < 0.01; I2 = 67.8%], recurrence [HR:1.09, 95% CI: 1.02-1.16, P < 0.01; I2 = 89.5%], and MACEs (HR:1.60, 95% CI: 1.13-2.27, P < 0.01; I2 = 82.0%) of AF. Furthermore, patients with AF showed higher sST2 than controls without AF (SMD: 0.41, 95% CI: 0.27-0.54, P < 0.01; I2 = 0%), and AF patients with recurrence after catheter ablation (CA) showed significantly higher sST2 than those without recurrence (SMD: 0.81, 95% CI: 0.33-1.28, P < 0.01; I2 = 83.9%). Sensitivity analyses showed that the outcomes were stable. Conclusions Higher sST2 was association with an increased risk of occurrence, recurrence, and MACEs of AF. Assessing sST2 can be used as a potential screening method to predict AF outcomes. Systematic Review Registration PROSPERO (CRD42023459789).
Collapse
Affiliation(s)
- Pengfei Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianpeng Du
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Diseases Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Salvioli S, Basile MS, Bencivenga L, Carrino S, Conte M, Damanti S, De Lorenzo R, Fiorenzato E, Gialluisi A, Ingannato A, Antonini A, Baldini N, Capri M, Cenci S, Iacoviello L, Nacmias B, Olivieri F, Rengo G, Querini PR, Lattanzio F. Biomarkers of aging in frailty and age-associated disorders: State of the art and future perspective. Ageing Res Rev 2023; 91:102044. [PMID: 37647997 DOI: 10.1016/j.arr.2023.102044] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
According to the Geroscience concept that organismal aging and age-associated diseases share the same basic molecular mechanisms, the identification of biomarkers of age that can efficiently classify people as biologically older (or younger) than their chronological (i.e. calendar) age is becoming of paramount importance. These people will be in fact at higher (or lower) risk for many different age-associated diseases, including cardiovascular diseases, neurodegeneration, cancer, etc. In turn, patients suffering from these diseases are biologically older than healthy age-matched individuals. Many biomarkers that correlate with age have been described so far. The aim of the present review is to discuss the usefulness of some of these biomarkers (especially soluble, circulating ones) in order to identify frail patients, possibly before the appearance of clinical symptoms, as well as patients at risk for age-associated diseases. An overview of selected biomarkers will be discussed in this regard, in particular we will focus on biomarkers related to metabolic stress response, inflammation, and cell death (in particular in neurodegeneration), all phenomena connected to inflammaging (chronic, low-grade, age-associated inflammation). In the second part of the review, next-generation markers such as extracellular vesicles and their cargos, epigenetic markers and gut microbiota composition, will be discussed. Since recent progresses in omics techniques have allowed an exponential increase in the production of laboratory data also in the field of biomarkers of age, making it difficult to extract biological meaning from the huge mass of available data, Artificial Intelligence (AI) approaches will be discussed as an increasingly important strategy for extracting knowledge from raw data and providing practitioners with actionable information to treat patients.
Collapse
Affiliation(s)
- Stefano Salvioli
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy
| | - Sara Carrino
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Eleonora Fiorenzato
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy
| | - Alessandro Gialluisi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Angelo Antonini
- Parkinson's Disease and Movement Disorders Unit, Center for Rare Neurological Diseases (ERN-RND), Department of Neurosciences, University of Padova, Padova, Italy; Center for Neurodegenerative Disease Research (CESNE), Department of Neurosciences, University of Padova, Padova, Italy
| | - Nicola Baldini
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Miriam Capri
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milano, Italy
| | - Licia Iacoviello
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy; EPIMED Research Center, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica Delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Napoli, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Scientific Institute of Telese Terme, Telese Terme, Italy
| | | | | |
Collapse
|
15
|
Witkowska A, Staciwa M, Duraj I, Wozniak E, Broncel M, Gorzelak-Pabis P. Interleukin-33/sST2: Dynamic assessment in patients with acute coronary syndrome. Adv Med Sci 2023; 68:195-201. [PMID: 37216709 DOI: 10.1016/j.advms.2023.05.001] [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: 09/15/2022] [Revised: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
PURPOSE Interleukin (IL)-33 and its soluble receptor ST2 (sST2) play a crucial role in the immune response. sST2 has been approved by the Food and Drug Administration as a prognostic biomarker of mortality in chronic heart failure patients, however, the role of IL-33 and sST2 in atherosclerotic cardiovascular disease remains unclear. The aim of this study was to measure serum level of IL-33 and sST2 of patients at the onset of acute coronary syndrome (ACS) and 3 months after primary percutaneous revascularization. PATIENTS AND METHODS Forty patients were divided into ST segment elevation myocardial infarction (STEMI) group, non-ST segment elevation myocardial infarction (NSTEMI) and unstable angina (UA) group. IL-33 and sST2 level were measured with ELISA. Additionally, IL-33 expression in peripheral blood mononuclear cells (PBMCs), was evaluated. RESULTS All ACS patients had a significantly lower level of sST2 3 months after ACS as compared to the baseline (p < 0.039). The STEMI patients had higher serum levels of IL-33 at the moment of ACS as compared to 3 months after the event, with an average decrease of 17.87 pg/ml (p < 0.007). Conversely, sST2 serum levels were still high after 3 months following an ACS in STEMI patients. ROC curve demonstrated that increased IL-33 serum level could be STEMI predictor. CONCLUSIONS The assessment of the baseline and dynamics of changes in IL-33 and sST2 concentrations in patients with ACS may be important for the diagnostic process and may help in understanding of how the immune mechanisms work at the moment of an ACS event.
Collapse
Affiliation(s)
- Anna Witkowska
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland.
| | - Mateusz Staciwa
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Iwona Duraj
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Ewelina Wozniak
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Marlena Broncel
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Paulina Gorzelak-Pabis
- Department of Internal Diseases and Clinical Pharmacology, Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
16
|
Wang C, Hao W. Cardiac arrhythmia and immune response in COVID-19 patients. Allergol Immunopathol (Madr) 2023; 51:63-70. [PMID: 37422781 DOI: 10.15586/aei.v51i4.883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/15/2023] [Indexed: 07/11/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has presented substantial challenges for providing health care due to the numerous complications on the respiratory and cardiovascular systems of people. Cardiac arrhythmia is one of the cardiac complications, and it was observed in COVID-19 patients. Moreover, arrhythmia and cardiac arrest are common in COVID-19 patients in the intensive care unit. The occurrence of cardiac arrhythmia in COVID-19 patients is associated with hypoxia, cytokine storm, myocardial ischemia and inflammatory disease such as congestive heart failure. It is necessary to know the occurrence and mechanisms of tachyarrhythmia and bradyarrhythmia in patients with COVID-19 infection for their proper management. This review provides an overview of the association between COVID-19 and arrhythmias by detailing possible pathophysiological mechanisms.
Collapse
Affiliation(s)
- Chunlian Wang
- Institute for Prevention and Control of Chronic Non-communicable Diseases, Harbin Center for Disease Control and Prevention, Harbin, China
| | - Wenqiang Hao
- The Second Ward of Internal Medicine, The Second Hospital of Heilongjiang Province, Harbin, China; ;
| |
Collapse
|
17
|
Costa D, Pellicano C, Mercuri V, Arnone JM, Rizzo F, Leodori G, Gargiulo P, Rosato E. In acromegalic patients the serum levels of interleukin-33 and Resolvin D1 influence skin perfusion of hands: A pilot study. Microvasc Res 2023; 149:104571. [PMID: 37321455 DOI: 10.1016/j.mvr.2023.104571] [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: 05/11/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
AIM Acromegaly is a rare chronic disease, caused by the over-secretion of growth hormone (GH), that creates a pro-inflammatory state, but the exact mechanisms by which GH or insulin-like growth factor 1 (IGF-I) act on inflammatory cells are not fully understood. Aim of the study was to evaluate Interleukin-33 (IL33) and D-series resolvins 1 (RvD1) and the skin perfusion of hands in patients with acromegaly (AP) and healthy controls (HC). METHODS IL33 and RvD1 have been assessed in 20 AP and 20 HC. Nailfold videocapillaroscopy (NVC) was performed and skin perfusion of hands was assessed by laser speckle contrast analysis (LASCA) in both populations. RESULTS IL33 was significantly higher in AP compared to HC [73.08 pg/ml (IQR 47.11-100.80 pg/ml) vs 41.5 4 pg/ml (IQR 20.16-55.49 pg/ml), p < 0.05] and RvD1 was significantly lower in AP than HC [36.1 pg/ml (IQR 27.88-66.21 pg/ml) vs 60.01 pg/ml (IQR 46.88-74.69 pg/ml), p < 0.05]. At LASCA, peripheral blood perfusion (PBP) was significantly lower in AP compared to HC [56.66 pU (IQR 46.29-65.44 pU) vs 87 pU (IQR 80-98 pU), p < 0.001]. The median values of ROI1 and ROI3 were significantly lower in AP compared to HC [112.81 pU (IQR 83.36-121.69 pU) vs 131 pU (IQR 108-135 pU), p < 0.05] and [59.78 pU (IQR 46.84-79.75 pU) vs 85 pU (IQR 78-98 pU), p < 0.05], respectively. The proximal-distal gradient (PDG) was observed in 8 of 20 (40 %) AP. CONCLUSION Serum IL33 is higher in AP compared to HC; conversely, RvD1 is lower in AP compared to HC. Reduction of PBP of hands was present in AP compared to HC, probably due to endothelial dysfunction.
Collapse
Affiliation(s)
- Denise Costa
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Chiara Pellicano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Valeria Mercuri
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Jacopo Maria Arnone
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Flavio Rizzo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giorgia Leodori
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Patrizia Gargiulo
- Department of Experimental Medicine, Endocrinology-Pituitary Disease, Sapienza University of Rome, Rome, Italy
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
18
|
Sciatti E, Merlo A, Scangiuzzi C, Limonta R, Gori M, D’Elia E, Aimo A, Vergaro G, Emdin M, Senni M. Prognostic Value of sST2 in Heart Failure. J Clin Med 2023; 12:3970. [PMID: 37373664 PMCID: PMC10299183 DOI: 10.3390/jcm12123970] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
In recent years, there has been growing interest in the risk stratification for heart failure, and the use of multiple biomarkers to identify different pathophysiological processes associated with this condition. One such biomarker is soluble suppression of tumorigenicity-2 (sST2), which has shown some potential for integration into clinical practice. sST2 is produced by both cardiac fibroblasts and cardiomyocytes in response to myocardial stress. Other sources of sST2 are endothelial cells of the aorta and coronary arteries and immune cells such as T cells. Indeed, ST2 is also associated with inflammatory and immune processes. We aimed at reviewing the prognostic value of sST2 in both chronic and acute heart failure. In this setting, we also provide a flowchart about its potential use in clinical practice.
Collapse
Affiliation(s)
- Edoardo Sciatti
- Cardiology Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (M.G.); (E.D.); (M.S.)
| | - Anna Merlo
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (A.M.); (C.S.); (R.L.)
| | - Claudio Scangiuzzi
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (A.M.); (C.S.); (R.L.)
| | - Raul Limonta
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (A.M.); (C.S.); (R.L.)
| | - Mauro Gori
- Cardiology Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (M.G.); (E.D.); (M.S.)
| | - Emilia D’Elia
- Cardiology Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (M.G.); (E.D.); (M.S.)
| | - Alberto Aimo
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, 56127 Pisa, Italy; (A.A.); (G.V.); (M.E.)
- Fondazione Toscana G. Monasterio, 56124 Pisa, Italy
| | - Giuseppe Vergaro
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, 56127 Pisa, Italy; (A.A.); (G.V.); (M.E.)
- Fondazione Toscana G. Monasterio, 56124 Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant’Anna, 56127 Pisa, Italy; (A.A.); (G.V.); (M.E.)
- Fondazione Toscana G. Monasterio, 56124 Pisa, Italy
| | - Michele Senni
- Cardiology Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy; (M.G.); (E.D.); (M.S.)
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (A.M.); (C.S.); (R.L.)
| |
Collapse
|
19
|
Dimosiari A, Patoulias D, Kitas GD, Dimitroulas T. Do Interleukin-1 and Interleukin-6 Antagonists Hold Any Place in the Treatment of Atherosclerotic Cardiovascular Disease and Related Co-Morbidities? An Overview of Available Clinical Evidence. J Clin Med 2023; 12:jcm12041302. [PMID: 36835838 PMCID: PMC9962740 DOI: 10.3390/jcm12041302] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Cardiovascular disease (CVD) constitutes a real pandemic of the 21st century. According to data from the Centers for Disease Control and Prevention, one person dies every 34 min due to some form of CVD in the United States. Apart from the extremely high morbidity and mortality accompanying CVD, the economic burden seems to be unbearable even for developed countries in the Western World. The role of inflammation in the development and progression of CVD appears to be crucial, while, various inflammatory pathways, such as the Nod-like receptor protein 3 (NLRP3) inflammasome-interleukin (IL)-1/IL-6 pathway of the innate immunity, have attracted scientific interest during the last decade, as a potential treatment target in primary and/or secondary prevention of CVD. Whereas there is a significant amount of evidence, stemming mainly from observational studies, concerning the cardiovascular safety of IL-1 and IL-6 antagonists in patients with rheumatic diseases, evidence from relevant randomized controlled trials (RCTs) is rather scarce and conflicting, especially for patients without underlying rheumatic disease. In this review, we summarize and critically present the currently available evidence, both from RCTs and observational studies, concerning the place that IL-1 and IL-6 antagonists may hold in the treatment of CVD.
Collapse
Affiliation(s)
- Athina Dimosiari
- Second Department of Internal Medicine, European Interbalkan Medical Center, 57001 Thessaloniki, Greece
| | - Dimitrios Patoulias
- Second Department of Internal Medicine, European Interbalkan Medical Center, 57001 Thessaloniki, Greece
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, General Hospital Hippokration, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Correspondence:
| | - George D. Kitas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, General Hospital Hippokration, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
20
|
Inflammageing and Cardiovascular System: Focus on Cardiokines and Cardiac-Specific Biomarkers. Int J Mol Sci 2023; 24:ijms24010844. [PMID: 36614282 PMCID: PMC9820990 DOI: 10.3390/ijms24010844] [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: 12/02/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
The term "inflammageing" was introduced in 2000, with the aim of describing the chronic inflammatory state typical of elderly individuals, which is characterized by a combination of elevated levels of inflammatory biomarkers, a high burden of comorbidities, an elevated risk of disability, frailty, and premature death. Inflammageing is a hallmark of various cardiovascular diseases, including atherosclerosis, hypertension, and rapid progression to heart failure. The great experimental and clinical evidence accumulated in recent years has clearly demonstrated that early detection and counteraction of inflammageing is a promising strategy not only to prevent cardiovascular disease, but also to slow down the progressive decline of health that occurs with ageing. It is conceivable that beneficial effects of counteracting inflammageing should be most effective if implemented in the early stages, when the compensatory capacity of the organism is not completely exhausted. Early interventions and treatments require early diagnosis using reliable and cost-effective biomarkers. Indeed, recent clinical studies have demonstrated that cardiac-specific biomarkers (i.e., cardiac natriuretic peptides and cardiac troponins) are able to identify, even in the general population, the individuals at highest risk of progression to heart failure. However, further clinical studies are needed to better understand the usefulness and cost/benefit ratio of cardiac-specific biomarkers as potential targets in preventive and therapeutic strategies for early detection and counteraction of inflammageing mechanisms and in this way slowing the progressive decline of health that occurs with ageing.
Collapse
|
21
|
The Therapeutic Role of SGLT-2 Inhibitors in Acute Heart Failure: From Pathophysiologic Mechanisms to Clinical Evidence with Pooled Analysis of Relevant Studies across Safety and Efficacy Endpoints of Interest. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122062. [PMID: 36556427 PMCID: PMC9782870 DOI: 10.3390/life12122062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
(1) Background: Sodium-glucose co-transporter-2 (SGLT-2) inhibitors constitute a novel drug class with remarkable cardiovascular benefits for patients with chronic heart failure (HF). Recently, this class has been utilized in acute HF as an additional treatment option to classic diuretics, which remain the cornerstone of treatment. (2) Methods: We attempted to identify those pathophysiologic mechanisms targeted by SGLT-2 inhibitors, which could be of benefit to patients with acute HF. We then conducted a comprehensive review of the literature within the PubMed database in order to identify relevant studies, both randomized controlled trials (RCTs) and observational studies, assessing the safety and efficacy of SGLT-2 inhibitors in acute HF. (3) Results: SGLT-2 inhibitors induce significant osmotic diuresis and natriuresis, decrease interstitial fluid volume and blood pressure, improve left ventricular (LV) function, ameliorate LV remodeling and prevent atrial arrhythmia occurrence, mechanisms that seem to be beneficial in acute HF. However, currently available studies, including six RCTs and two real-world studies, provide conflicting results concerning the true efficacy of SGLT-2 inhibitors, including "hard" surrogate endpoints. (4) Conclusions: Current evidence appears insufficient to substantiate the use of SGLT-2 inhibitors in acute HF. Further trials are required to shed more light on this issue.
Collapse
|
22
|
Tembhre MK, Sriwastva MK, Hote MP, Srivastava S, Solanki P, Imran S, Lakshmy R, Sharma A, Jaiswal K, Upadhyay AD. Interleukin-33 Induces Neutrophil Extracellular Trap (NET) Formation and Macrophage Necroptosis via Enhancing Oxidative Stress and Secretion of Proatherogenic Factors in Advanced Atherosclerosis. Antioxidants (Basel) 2022; 11:antiox11122343. [PMID: 36552551 PMCID: PMC9774908 DOI: 10.3390/antiox11122343] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Interleukin-33 (IL-33) acts as an 'alarmin', and its role has been demonstrated in driving immune regulation and inflammation in many human diseases. However, the precise mechanism of action of IL-33 in regulating neutrophil and macrophage functioning is not defined in advanced atherosclerosis (aAT) patients. Further, the role of IL-33 in neutrophil extracellular trap (NET) formation in aAT and its consequent effect on macrophage function is not known. In the present study, we recruited n = 52 aAT patients and n = 52 control subjects. The neutrophils were isolated from both groups via ficoll/percoll-based density gradient centrifugation. The effect of IL-33 on the NET formation ability of the neutrophils was determined in both groups. Monocytes, isolated via a positive selection method, were used to differentiate them into macrophages from each of the study subjects and were challenged by IL-33-primed NETs, followed by the measurement of oxidative stress by calorimetric assay and the expression of the proinflammatory molecules by quantitative PCR (qPCR). Transcript and protein expression was determined by qPCR and immunofluorescence/ELISA, respectively. The increased expression of IL-33R (ST-2) was observed in the neutrophils, along with an increased serum concentration of IL-33 in aAT compared to the controls. IL-33 exacerbates NET formation via specifically upregulating CD16 expression in aAT. IL-33-primed NETs/neutrophils increased the cellular oxidative stress levels in the macrophages, leading to enhanced macrophage necroptosis and the release of atherogenic factors and matrix metalloproteinases (MMPs) in aAT compared to the controls. These findings suggested a pathogenic effect of the IL-33/ST-2 pathway in aAT patients by exacerbating NET formation and macrophage necroptosis, thereby facilitating the release of inflammatory factors and the release of MMPs that may be critical for the destabilization/rupture of atherosclerotic plaques in aAT. Targeting the IL-33/ST-2-NETs axis may be a promising therapeutic target for preventing plaque instability/rupture and its adverse complications in aAT.
Collapse
Affiliation(s)
- Manoj Kumar Tembhre
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
- Correspondence: ; Tel.: +91-880-050-2994
| | | | - Milind Padmakar Hote
- Department of Cardiothoracic & Vascular Surgery, C. T. Centre, AIIMS, New Delhi 110029, India
| | - Shikha Srivastava
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, USA
| | - Priyanka Solanki
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Shafaque Imran
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Ramakrishnan Lakshmy
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Alpana Sharma
- Department of Biochemistry, AIIMS, New Delhi 110029, India
| | - Kailash Jaiswal
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | | |
Collapse
|