1
|
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
|
2
|
Battista S, Bima P, Forno D, Luzzi D, Pizzolato E, Ianniello A, Ponzetto F, Rumbolo F, Settanni F, Mengozzi G, Morello F, Lupia E. Plasma soluble suppression of tumorigenesis 2 measured in the emergency department for diagnosis and outcome prediction of sepsis: A single-center prospective study. Clin Chim Acta 2024; 553:117710. [PMID: 38141937 DOI: 10.1016/j.cca.2023.117710] [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: 10/27/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND AND AIMS The diagnostic and prognostic performance of soluble Suppression of Tumorigenicity 2 (sST2) in suspected septic patients presenting to the Emergency Department (ED) is largely unknown. MATERIALS AND METHODS Patients were included in this prospective study if there was high suspicion of sepsis. The plasma level of sST2 was measured during initial ED evaluation. Outcomes were the evaluation of (1) sST2 diagnostic performance (alone and in combination with procalcitonin [PCT]), and (2) sST2 ability to predict 30-day and 90-day all-cause mortality. RESULTS Among 569 patients included, 481 (84.5 %) had sepsis or septic shock. Plasma sST2 levels were more elevated in septic patients (159 [71-331] vs 50 [31-103] ng/mL, P < 0.001). The AUC of sST2 for sepsis diagnosis was lower than the AUC of PCT (0.76 vs 0.85, P = 0.03). The best cut-off for sST2 was 61.7 ng/mL, with a sensitivity of 79.9 % and a specificity of 70.6 %. sST2 was able to correctly reclassify septic patients with PCT <0.5 (NRI 28.9 % [P = 0.02]). sST2 level was an independent predictor of 30-day mortality in a model including clinical variables (aHR 2.03 [1.24-3.33], C-index 0.69). CONCLUSION sST2 could be a useful adjunct in diagnosing sepsis and in all-cause mortality prediction.
Collapse
Affiliation(s)
- Stefania Battista
- Emergency Medicine Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Paolo Bima
- Emergency Medicine Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy; Cardiovascular Research Institute Basel, Basel, Switzerland
| | - Daniela Forno
- Emergency Medicine Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Demetrio Luzzi
- Emergency Medicine Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Elisa Pizzolato
- Emergency Medicine Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Alice Ianniello
- Department of Medical Sciences, University of Turin, Turin, Italy; Clinical Biochemistry Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Federico Ponzetto
- Department of Medical Sciences, University of Turin, Turin, Italy; Clinical Biochemistry Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Francesca Rumbolo
- Department of Medical Sciences, University of Turin, Turin, Italy; Clinical Biochemistry Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Fabio Settanni
- Department of Medical Sciences, University of Turin, Turin, Italy; Clinical Biochemistry Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Giulio Mengozzi
- Department of Medical Sciences, University of Turin, Turin, Italy; Clinical Biochemistry Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy
| | - Fulvio Morello
- Emergency Medicine Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Enrico Lupia
- Emergency Medicine Unit, "Città della Salute e della Scienza" University Hospital, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
3
|
Zhou Y, Xu Z, Liu Z. Role of IL-33-ST2 pathway in regulating inflammation: current evidence and future perspectives. J Transl Med 2023; 21:902. [PMID: 38082335 PMCID: PMC10714644 DOI: 10.1186/s12967-023-04782-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Interleukin (IL)-33 is an alarmin of the IL-1 superfamily localized to the nucleus of expressing cells, such as endothelial cells, epithelial cells, and fibroblasts. In response to cellular damage or stress, IL-33 is released and activates innate immune responses in some immune and structural cells via its receptor interleukin-1 receptor like-1 (IL-1RL1 or ST2). Recently, IL-33 has become a hot topic of research because of its role in pulmonary inflammation. The IL-33-ST2 signaling pathway plays a pro-inflammatory role by activating the type 2 inflammatory response, producing type 2 cytokines and chemokines. Elevated levels of IL-33 and ST2 have been observed in chronic pulmonary obstructive disease (COPD). Notably, IL-33 is present in COPD induced by cigarette smoke or acute inflammations. The role of IL-33 in sepsis is becoming increasingly prominent, and understanding its significance in the treatment of sepsis associated with high mortality is critical. In addition to its pro-inflammatory effects, the IL-33-ST2 axis appears to play a role in bacterial clearance and tissue repair. In this review, we focused on the role of the IL-33-ST2 axis in sepsis, asthma, and COPD and summarized the therapeutic targets associated with this axis, providing a basis for future treatment.
Collapse
Affiliation(s)
- Yilu Zhou
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhendong Xu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zhiqiang Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
4
|
Chatterton C, Romero R, Jung E, Gallo DM, Suksai M, Diaz-Primera R, Erez O, Chaemsaithong P, Tarca AL, Gotsch F, Bosco M, Chaiworapongsa T. A biomarker for bacteremia in pregnant women with acute pyelonephritis: soluble suppressor of tumorigenicity 2 or sST2. J Matern Fetal Neonatal Med 2023; 36:2183470. [PMID: 36997168 PMCID: PMC10352993 DOI: 10.1080/14767058.2023.2183470] [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: 06/11/2022] [Accepted: 02/15/2023] [Indexed: 04/01/2023]
Abstract
Objective: Sepsis is a leading cause of maternal death, and its diagnosis during the golden hour is critical to improve survival. Acute pyelonephritis in pregnancy is a risk factor for obstetrical and medical complications, and it is a major cause of sepsis, as bacteremia complicates 15-20% of pyelonephritis episodes in pregnancy. The diagnosis of bacteremia currently relies on blood cultures, whereas a rapid test could allow timely management and improved outcomes. Soluble suppression of tumorigenicity 2 (sST2) was previously proposed as a biomarker for sepsis in non-pregnant adults and children. This study was designed to determine whether maternal plasma concentrations of sST2 in pregnant patients with pyelonephritis can help to identify those at risk for bacteremia.Study design: This cross-sectional study included women with normal pregnancy (n = 131) and pregnant women with acute pyelonephritis (n = 36). Acute pyelonephritis was diagnosed based on a combination of clinical findings and a positive urine culture. Patients were further classified according to the results of blood cultures into those with and without bacteremia. Plasma concentrations of sST2 were determined by a sensitive immunoassay. Non-parametric statistics were used for analysis.Results: The maternal plasma sST2 concentration increased with gestational age in normal pregnancies. Pregnant patients with acute pyelonephritis had a higher median (interquartile range) plasma sST2 concentration than those with a normal pregnancy [85 (47-239) ng/mL vs. 31 (14-52) ng/mL, p < .001]. Among patients with pyelonephritis, those with a positive blood culture had a median plasma concentration of sST2 higher than that of patients with a negative blood culture [258 (IQR: 75-305) ng/mL vs. 83 (IQR: 46-153) ng/mL; p = .03]. An elevated plasma concentration of sST2 ≥ 215 ng/mL had a sensitivity of 73% and a specificity of 95% (area under the receiver operating characteristic curve, 0.74; p = .003) with a positive likelihood ratio of 13.8 and a negative likelihood ratio of 0.3 for the identification of patients who had a positive blood culture.Conclusion: sST2 is a candidate biomarker to identify bacteremia in pregnant women with pyelonephritis. Rapid identification of these patients may optimize patient care.
Collapse
Affiliation(s)
- Carolyn Chatterton
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Dahiana M. Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Gynecology and Obstetrics, Universidad del Valle, Cali, Colombia
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer Sheva, Israel
| | - Piya Chaemsaithong
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
5
|
Kanninen T, Jung E, Gallo DM, Diaz-Primera R, Romero R, Gotsch F, Suksai M, Bosco M, Chaiworapongsa T. Soluble suppression of tumorigenicity-2 in pregnancy with a small-for-gestational-age fetus and with preeclampsia. J Matern Fetal Neonatal Med 2023; 36:2153034. [PMID: 36521862 PMCID: PMC10291739 DOI: 10.1080/14767058.2022.2153034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Preeclampsia and fetal growth disorders are pregnancy-specific conditions that share common pathophysiological mechanisms. Yet, why some patients develop preeclampsia while others experience fetal growth restriction, or a combination of both clinical presentations, is unknown. We propose that the difference in severity of the maternal inflammatory response can contribute to the clinical phenotypes of preeclampsia vs. small for gestational age (SGA). To assess this hypothesis, we measured maternal plasma concentrations of the soluble isoform of suppression of tumorigenicity-2 (sST2), a member of the interleukin-1 receptor family that buffers proinflammatory responses. Previous reports showed that serum sST2 concentrations rise in the presence of intravascular inflammation and Th1-type immune responses and are significantly higher in patients with preeclampsia compared to those with normal pregnancy. The behavior of sST2 in pregnancies complicated by SGA has not been reported. This study was conducted to compare sST2 plasma concentrations in normal pregnancies, in those with preeclampsia, and in those with an SGA fetus. METHODS This retrospective cross-sectional study included women with an SGA fetus (n = 52), women with preeclampsia (n = 106), and those with normal pregnancy (n = 131). Maternal plasma concentrations of sST2 were determined by enzyme-linked immunosorbent assay. Doppler velocimetry of the uterine and umbilical arteries was available in a subset of patients with SGA (42 patients and 43 patients, respectively). RESULTS (1) Women with an SGA fetus had a significantly higher median plasma concentration of sST2 than normal pregnant women (p = .008); (2) women with preeclampsia had a significantly higher median plasma concentration of sST2 than those with normal pregnancy (p < .001) and those with an SGA fetus (p < .001); (3) patients with SGA and abnormal uterine artery Doppler velocimetry had a higher median plasma concentration of sST2 than controls (p < .01) and those with SGA and normal uterine artery Doppler velocimetry (p = .02); (4) there was no significant difference in the median plasma sST2 concentration between patients with SGA who had normal uterine artery Doppler velocimetry and controls (p = .4); (5) among patients with SGA, those with abnormal and those with normal umbilical artery Doppler velocimetry had higher median plasma sST2 concentrations than controls (p = .001 and p = .02, respectively); and (6) there was no significant difference in the median plasma sST2 concentrations between patients with SGA who did and those who did not have abnormal umbilical artery Doppler velocimetry (p = .06). CONCLUSIONS Preeclampsia and disorders of fetal growth are conditions characterized by intravascular inflammation, as reflected by maternal plasma concentrations of sST2. The severity of intravascular inflammation is highest in patients with preeclampsia.
Collapse
Affiliation(s)
- Tomi Kanninen
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dahiana M Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Gynecology and Obstetrics, Universidad del Valle, Cali, Colombia
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
- Detroit Medical Center, Detroit, MI, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
6
|
Horner E, Lord JM, Hazeldine J. The immune suppressive properties of damage associated molecular patterns in the setting of sterile traumatic injury. Front Immunol 2023; 14:1239683. [PMID: 37662933 PMCID: PMC10469493 DOI: 10.3389/fimmu.2023.1239683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Associated with the development of hospital-acquired infections, major traumatic injury results in an immediate and persistent state of systemic immunosuppression, yet the underlying mechanisms are poorly understood. Detected in the circulation in the minutes, days and weeks following injury, damage associated molecular patterns (DAMPs) are a heterogeneous collection of proteins, lipids and DNA renowned for initiating the systemic inflammatory response syndrome. Suggesting additional immunomodulatory roles in the post-trauma immune response, data are emerging implicating DAMPs as potential mediators of post-trauma immune suppression. Discussing the results of in vitro, in vivo and ex vivo studies, the purpose of this review is to summarise the emerging immune tolerising properties of cytosolic, nuclear and mitochondrial-derived DAMPs. Direct inhibition of neutrophil antimicrobial activities, the induction of endotoxin tolerance in monocytes and macrophages, and the recruitment, activation and expansion of myeloid derived suppressor cells and regulatory T cells are examples of some of the immune suppressive properties assigned to DAMPs so far. Crucially, with studies identifying the molecular mechanisms by which DAMPs promote immune suppression, therapeutic strategies that prevent and/or reverse DAMP-induced immunosuppression have been proposed. Approaches currently under consideration include the use of synthetic polymers, or the delivery of plasma proteins, to scavenge circulating DAMPs, or to treat critically-injured patients with antagonists of DAMP receptors. However, as DAMPs share signalling pathways with pathogen associated molecular patterns, and pro-inflammatory responses are essential for tissue regeneration, these approaches need to be carefully considered in order to ensure that modulating DAMP levels and/or their interaction with immune cells does not negatively impact upon anti-microbial defence and the physiological responses of tissue repair and wound healing.
Collapse
Affiliation(s)
- Emily Horner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Janet M. Lord
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Jon Hazeldine
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| |
Collapse
|
7
|
Weber B, Voth M, Rottluff K, Marzi I, Henrich D, Leppik L. Evaluation of IL-33R and Galectin-3 as New Biomarkers of Cardiac Damage after Polytrauma-Association with Cardiac Comorbidities and Risk Factors. J Clin Med 2022; 11:6350. [PMID: 36362577 PMCID: PMC9659247 DOI: 10.3390/jcm11216350] [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: 09/23/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 09/08/2024] Open
Abstract
Polytrauma is one of the disorders with the greatest economic impact on healthcare in society and one predictor for poor outcome is cardiac damage. Interleukin 33 receptors (IL-33R) and galectin-3 are two new potential cardiac trauma biomarkers that are the subjects of this investigation. Additionally, this study assesses pre-existing cardiac damage or risk factors as predictors of cardiac damage after polytrauma. This retrospective study includes 107 polytraumatized patients with an ISS ≥16 admitted in a Level 1 Trauma Centre. Plasma samples were taken at admission. IL-33R and galectin-3 concentrations were detected in plasma samples by ELISA. Both did not correlate with the cardiac damage measured by troponin. Next to troponin, IL-33R was increased in patients with pre-existing cardiac comorbidities. In the subgroup of patients with cardiac comorbidities, the BMI and the initial blood sugar level were significantly increased compared to patients without cardiac comorbidities. Galectin-3 and IL-33R were shown to not correlate with cardiac damage. However, our data suggests that IL-33R protein should be revised in future studies as a marker of cardiac comorbidities. Further, our data indicate that patients with cardiac comorbidities represent a separate group of polytrauma patients characterized by higher concentrations of troponin, IL-33R, BMI and initial sugar level.
Collapse
Affiliation(s)
- Birte Weber
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, 60596 Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
8
|
Xu C, Du L, Chen F, Tang K, Tang L, Shi J, Xiao L, Zeng Z, Liang Y, Guo Y. Increased expression of IL1-RL1 is associated with type 2 and type 1 immune pathways in asthma. BMC Immunol 2022; 23:23. [PMID: 35578178 PMCID: PMC9112580 DOI: 10.1186/s12865-022-00499-z] [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: 12/08/2021] [Accepted: 05/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background Asthma is a common chronic airway disease in the world. The purpose of this study was to explore the expression of IL1-RL1 in sputum and its correlation with Th1 and Th2 cytokines in asthma. Methods We recruited 132 subjects, detected IL1-RL1 protein level in sputum supernatant by ELISA, and analyzed the correlation between the expression level of IL1-RL1 and fraction of exhaled nitric oxide (FeNO), IgE, peripheral blood eosinophil count (EOS#), and Th2 cytokines (IL-4, IL-5, IL-10, IL-13, IL-33 and TSLP) and Th1 cytokines (IFN-γ, IL-2, IL-8). The diagnostic value of IL1-RL1 was evaluated by ROC curve. The expression of IL1-RL1 was further confirmed by BEAS-2B cell in vitro. Results Compared with the healthy control group, the expression of IL1-RL1 in sputum supernatant, sputum cells and serum of patients with asthma increased. The AUC of ROC curve of IL1-RL1 in sputum supernatant and serum were 0.6840 (p = 0.0034), and 0.7009 (p = 0.0233), respectively. IL1-RL1 was positively correlated with FeNO, IgE, EOS#, Th2 cytokines (IL-4, IL-5, IL-10, IL-13, IL-33 and TSLP) and Th1 cytokines (IFN-γ, IL-2, IL-8) in induced sputum supernatant. Four weeks after inhaled glucocorticoids (ICS) treatment, the expression of IL1-RL1 in sputum supernatant and serum was increased. In vitro, the expression of IL1-RL1 in BEAS-2B was increased after stimulated by IL-4 or IL-13 for 24 h. Conclusion The expression of IL1-RL1 in sputum supernatant, sputum cells and serum of patients with asthma was increased, and was positively correlated with some inflammatory markers in patients with asthma. IL1-RL1 may be used as a potential biomarker for the diagnosis and treatment of asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-022-00499-z.
Collapse
Affiliation(s)
- Changyi Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Lijuan Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Fengjia Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Kun Tang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Lu Tang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Jia Shi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Lisha Xiao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Zhimin Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| | - Yuxia Liang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| | - Yubiao Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
9
|
Ilieșiu AM, Hodorogea AS, Balahura AM, Bădilă E. Non-Invasive Assessment of Congestion by Cardiovascular and Pulmonary Ultrasound and Biomarkers in Heart Failure. Diagnostics (Basel) 2022; 12:962. [PMID: 35454010 PMCID: PMC9024731 DOI: 10.3390/diagnostics12040962] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Worsening chronic heart failure (HF) is responsible for recurrent hospitalization and increased mortality risk after discharge, irrespective to the ejection fraction. Symptoms and signs of pulmonary and systemic congestion are the most common cause for hospitalization of acute decompensated HF, as a consequence of increased cardiac filling pressures. The elevated cardiac filling pressures, also called hemodynamic congestion, may precede the occurrence of clinical congestion by days or weeks. Since HF patients often have comorbidities, dyspnoea, the main symptom of HF, may be also caused by respiratory or other illnesses. Recent studies underline the importance of the diagnosis and treatment of hemodynamic congestion before HF symptoms worsen, reducing hospitalization and improving prognosis. In this paper we review the role of integrated evaluation of biomarkers and imaging technics, i.e., echocardiography and pulmonary ultrasound, for the diagnosis, prognosis and treatment of congestion in HF patients.
Collapse
Affiliation(s)
- Adriana Mihaela Ilieșiu
- Cardiology and Internal Medicine Department, Theodor Burghele Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Andreea Simona Hodorogea
- Cardiology and Internal Medicine Department, Theodor Burghele Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Ana-Maria Balahura
- Internal Medicine Department, Bucharest Clinical Emergency Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (E.B.)
| | - Elisabeta Bădilă
- Internal Medicine Department, Bucharest Clinical Emergency Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.B.); (E.B.)
| |
Collapse
|
10
|
Galeone A, Salem JE, Lebreton G, Coutance G, Nguyen L, Hulot JS, Atassi F, Bega M, Leprince P, Varnous S. Suppression of tumorigenicity-2 (ST2) is a promising biomarker in heart transplantation. Clin Transplant 2022; 36:e14616. [PMID: 35188995 DOI: 10.1111/ctr.14616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/15/2022] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND To evaluate the association between donors' and recipients' serum levels of soluble ST2 (sST2) and recipients' outcome after heart transplantation (HT). METHODS Blood samples were collected in 50 heart donors before organ procurement and in 50 recipients before HT (D0), a week after HT (D7) and at every first year's endomyocardial biopsy (EMB); sST2 levels were evaluated by ELISA. RESULTS Donors who sustained a cardiac arrest, had significantly higher sST2 levels. Recipients on national high emergency waiting list had significantly higher preoperative sST2 levels compared to recipients who did not. Recipients with postoperative sepsis or continuous renal replacement therapy had significantly higher sST2 levels at D7. Recipients who needed a postoperative ECMO for allograft dysfunction had significantly higher sST2 levels in their corresponding donors. Recipients who died during the hospitalization after the transplantation had significantly higher sST2 levels at D7 compared to recipients who did not. No difference was observed in sST2 levels in recipients who had mild allograft rejection and recipient who did not. CONCLUSIONS Higher sST2 levels in donors are associated to allograft dysfunction requiring ECMO in recipients; higher postoperative sST2 levels in recipients are associated with in-hospital mortality.
Collapse
Affiliation(s)
- Antonella Galeone
- Department of Thoracic and Cardiovascular Surgery, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Joe-Elie Salem
- Department of Pharmacology and Clinical Investigation Center, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Guillaume Lebreton
- Department of Thoracic and Cardiovascular Surgery, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Guillaume Coutance
- Department of Thoracic and Cardiovascular Surgery, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Lee Nguyen
- Department of Pharmacology and Clinical Investigation Center, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Jean Sébastien Hulot
- Sorbonne Université, INSERM, Research unit on cardiovascular, metabolic and nutrition diseases, (UMRS-1166), Institute of Cardiometabolism and Nutrition Paris, Paris, France
| | - Fabrice Atassi
- Sorbonne Université, INSERM, Research unit on cardiovascular, metabolic and nutrition diseases, (UMRS-1166), Institute of Cardiometabolism and Nutrition Paris, Paris, France
| | - Marco Bega
- Department of Thoracic and Cardiovascular Surgery, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Pascal Leprince
- Department of Thoracic and Cardiovascular Surgery, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Shaida Varnous
- Department of Thoracic and Cardiovascular Surgery, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| |
Collapse
|
11
|
Laggner M, Lingitz MT, Copic D, Direder M, Klas K, Bormann D, Gugerell A, Moser B, Radtke C, Hacker S, Mildner M, Ankersmit HJ, Haider T. Severity of thermal burn injury is associated with systemic neutrophil activation. Sci Rep 2022; 12:1654. [PMID: 35102298 PMCID: PMC8803945 DOI: 10.1038/s41598-022-05768-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Burn injuries elicit a unique and dynamic stress response which can lead to burn injury progression. Though neutrophils represent crucial players in the burn-induced immunological events, the dynamic secretion pattern and systemic levels of neutrophil-derived factors have not been investigated in detail so far. Serum levels of neutrophil elastase (NE), myeloperoxidase (MPO), citrullinated histone H3 (CitH3), and complement factor C3a were quantified in burn victims over 4 weeks post injury. Furthermore, the potential association with mortality, degree of burn injury, and inhalation trauma was evaluated. In addition, leukocyte, platelet, neutrophil, and lymphocyte counts were assessed. Lastly, we analyzed the association of neutrophil-derived factors with clinical severity scoring systems. Serum levels of NE, MPO, CitH3, and C3a were remarkably elevated in burn victims compared to healthy controls. Leukocyte and neutrophil counts were significantly increased on admission day and day 1, while relative lymphocytes were decreased in the first 7 days post burn trauma. Though neutrophil-derived factors did not predict mortality, patients suffering from 3rd degree burn injuries displayed increased CitH3 and NE levels. Accordingly, CitH3 and NE were elevated in cases with higher abbreviated burn severity indices (ABSI). Taken together, our data suggest a role for neutrophil activation and NETosis in burn injuries and burn injury progression. Targeting exacerbated neutrophil activation might represent a new therapeutic option for severe cases of burn injury.
Collapse
Affiliation(s)
- Maria Laggner
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Marie-Therese Lingitz
- Division of General Anesthesia and Intensive Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Dragan Copic
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Martin Direder
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Katharina Klas
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Daniel Bormann
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Alfred Gugerell
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Stefan Hacker
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090, Vienna, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Landesklinikum Wiener Neustadt, 2700, Wiener Neustadt, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090, Vienna, Austria.
| | - Thomas Haider
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
12
|
Abstract
Our understanding of the functions of the IL-1 superfamily cytokine and damage-associated molecular pattern IL-33 continues to evolve with our understanding of homeostasis and immunity. The early findings that IL-33 is a potent driver of type 2 immune responses promoting parasite expulsion, but also inflammatory diseases like allergy and asthma, have been further supported. Yet, as the importance of a type 2 response in tissue repair and homeostasis has emerged, so has the fundamental importance of IL-33 to these processes. In this review, we outline an evolving understanding of IL-33 immunobiology, paying particular attention to how IL-33 directs a network of ST2+ regulatory T cells, reparative and regulatory macrophages, and type 2 innate lymphoid cells that are fundamental to tissue development, homeostasis, and repair. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Gaelen K. Dwyer
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Louise M. D'Cruz
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
13
|
Wendt R, Lingitz MT, Laggner M, Mildner M, Traxler D, Graf A, Krotka P, Moser B, Hoetzenecker K, Kalbitz S, Lübbert C, Beige J, Ankersmit HJ. Clinical Relevance of Elevated Soluble ST2, HSP27 and 20S Proteasome at Hospital Admission in Patients with COVID-19. BIOLOGY 2021; 10:1186. [PMID: 34827178 PMCID: PMC8615143 DOI: 10.3390/biology10111186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 12/12/2022]
Abstract
Although, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) represents one of the biggest challenges in the world today, the exact immunopathogenic mechanism that leads to severe or critical Coronavirus Disease 2019 (COVID-19) has remained incompletely understood. Several studies have indicated that high systemic plasma levels of inflammatory cytokines result in the so-called "cytokine storm", with subsequent development of microthrombosis, disseminated intravascular coagulation, and multiorgan-failure. Therefore, we reasoned those elevated inflammatory molecules might act as prognostic factors. Here, we analyzed 245 serum samples of patients with COVID-19, collected at hospital admission. We assessed the levels of heat shock protein 27 (HSP27), soluble suppressor of tumorigenicity-2 (sST2) and 20S proteasome at hospital admission and explored their associations with overall-, 30-, 60-, 90-day- and in-hospital mortality. Moreover, we investigated their association with the risk of ventilation. We demonstrated that increased serum sST2 was uni- and multivariably associated with all endpoints. Furthermore, we also identified 20S proteasome as independent prognostic factor for in-hospital mortality (sST2, AUC = 0.73; HSP27, AUC = 0.59; 20S proteasome = 0.67). Elevated sST2, HSP27, and 20S proteasome levels at hospital admission were univariably associated with higher risk of invasive ventilation (OR = 1.8; p < 0.001; OR = 1.1; p = 0.04; OR = 1.03, p = 0.03, respectively). These findings could help to identify high-risk patients early in the course of COVID-19.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Infectious Diseases, Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Delitzscher Str. 141, 04129 Leipzig, Germany; (R.W.); (S.K.); (C.L.); (J.B.)
| | - Marie-Therese Lingitz
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.-T.L.); (M.L.); (M.M.); (D.T.); (B.M.)
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Maria Laggner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.-T.L.); (M.L.); (M.M.); (D.T.); (B.M.)
| | - Michael Mildner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.-T.L.); (M.L.); (M.M.); (D.T.); (B.M.)
- Department of Dermatology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Denise Traxler
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.-T.L.); (M.L.); (M.M.); (D.T.); (B.M.)
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alexandra Graf
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Spitalg. 23, 1090 Vienna, Austria; (A.G.); (P.K.)
| | - Pavla Krotka
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Spitalg. 23, 1090 Vienna, Austria; (A.G.); (P.K.)
| | - Bernhard Moser
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.-T.L.); (M.L.); (M.M.); (D.T.); (B.M.)
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Konrad Hoetzenecker
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Sven Kalbitz
- Department of Infectious Diseases, Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Delitzscher Str. 141, 04129 Leipzig, Germany; (R.W.); (S.K.); (C.L.); (J.B.)
| | - Christoph Lübbert
- Department of Infectious Diseases, Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Delitzscher Str. 141, 04129 Leipzig, Germany; (R.W.); (S.K.); (C.L.); (J.B.)
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine II, Leipzig University Medical Center, Liebigstr. 20, 04103 Leipzig, Germany
| | - Joachim Beige
- Department of Infectious Diseases, Tropical Medicine, Nephrology and Rheumatology, St. Georg Hospital, Delitzscher Str. 141, 04129 Leipzig, Germany; (R.W.); (S.K.); (C.L.); (J.B.)
- Department of Internal Medicine II, Martin-Luther-University Halle-Wittenberg, 06108 Halle/Saale, Germany
| | - Hendrik Jan Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.-T.L.); (M.L.); (M.M.); (D.T.); (B.M.)
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| |
Collapse
|
14
|
Rezar R, Paar V, Seelmaier C, Pretsch I, Schwaiger P, Kopp K, Kaufmann R, Felder TK, Prinz E, Gemes G, Pistulli R, Hoppe UC, Wernly B, Lichtenauer M. Soluble suppression of tumorigenicity 2 as outcome predictor after cardiopulmonary resuscitation: an observational prospective study. Sci Rep 2021; 11:21756. [PMID: 34741120 PMCID: PMC8571342 DOI: 10.1038/s41598-021-01389-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/27/2021] [Indexed: 11/09/2022] Open
Abstract
Prognostication after cardiopulmonary resuscitation (CPR) is complex. Novel biomarkers like soluble suppression of tumorigenicity 2 (sST2) may provide an objective approach. A total of 106 post-CPR patients were included in this single-center observational prospective study. Serum sST2 levels were obtained 24 h after admission. Individuals were assigned to two groups: patients below and above the overall cohort’s median sST2 concentration. Primary outcome was a combined endpoint at 6 months (death or Cerebral Performance Category > 2); secondary endpoint 30-day mortality. A uni- and multivariate logistic regression analysis were conducted. Elevated sST2-levels were associated with an increased risk for the primary outcome (OR 1.011, 95% CI 1.004–1.019, p = 0.004), yet no patients with poor neurological outcome were observed at 6 months. The optimal empirical cut-off for sST2 was 46.15 ng/ml (sensitivity 81%, specificity 53%, AUC 0.69). Levels above the median (> 53.42 ng/ml) were associated with higher odds for both endpoints (death or CPC > 2 after 6 months: 21% vs. 49%, OR 3.59, 95% CI 1.53–8.45, p = 0.003; death after 30 days: 17% vs. 43.3%, OR 3.75, 95% CI 1.52–9.21, p = 0.003). A positive correlation of serum sST2 after CPR with mortality at 30 days and 6 months after cardiac arrest could be demonstrated.
Collapse
Affiliation(s)
- Richard Rezar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria.
| | - Vera Paar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Clemens Seelmaier
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Ingrid Pretsch
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Philipp Schwaiger
- Department of Anaesthesiology, Perioperative Medicine and Intensive Care Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Kristen Kopp
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Reinhard Kaufmann
- Department of Radiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Thomas K Felder
- Department of Laboratory Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Erika Prinz
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Geza Gemes
- Department of Anaesthesiology and Intensive Care Medicine, Krankenhaus Der Barmherzigen Brüder Graz, Graz, Austria
| | - Rudin Pistulli
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Uta C Hoppe
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Bernhard Wernly
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria.,Department of Anaesthesiology, Perioperative Medicine and Intensive Care Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria.,Center for Public Health and Healthcare Research, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
15
|
Ambari AM, Setianto B, Santoso A, Radi B, Dwiputra B, Susilowati E, Tulrahmi F, Wind A, Cramer MJM, Doevendans P. Randomised controlled trial into the role of ramipril in fibrosis reduction in rheumatic heart disease: the RamiRHeD trial protocol. BMJ Open 2021; 11:e048016. [PMID: 34518254 PMCID: PMC8438922 DOI: 10.1136/bmjopen-2020-048016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Rheumatic heart disease (RHD) is a major burden in developing countries and accounts for 80% of all people living with the disease, where it causes most cardiovascular morbidity and mortality in children and young adults. Chronic inflammation and fibrosis of heart valve tissue due to chronic inflammation in RHD will cause calcification and thickening of the impacted heart valves, especially the mitral valve. This fibrogenesis is enhanced by the production of angiotensin II by increased transforming growth factor β expression and later by the binding of interleukin-33, which is known to have antihypertrophic and antifibrotic effects, to soluble sST2. sST2 binding to this non-natural ligand worsens fibrosis. Therefore, we hypothesise that ACE inhibitors (ACEIs) would improve rheumatic mitral valve stenosis. METHODS AND ANALYSIS This is a single-centre, double-blind, placebo-controlled, randomised clinical trial with a pre-post test design. Patients with rheumatic mitral stenosis and valve dysfunction will be planned for cardiac valve replacement operation and will be given ramipril 5 mg or placebo for a minimum of 12 weeks before the surgery. The expression of ST2 in the mitral valve is considered to be representative of cardiac fibrosis. Mitral valve tissue will be stained by immunohistochemistry to ST2. Plasma ST2 will be measured by ELISA. This study is conducted in the Department of Cardiology and Vascular Medicine, Universitas Indonesia, National Cardiac Center Harapan Kita Hospital, Jakarta, Indonesia, starting on 27 June 2019. ETHICS AND DISSEMINATION The performance and dissemination of this study were approved by the ethics committee of National Cardiovascular Center Harapan Kita with ethical code LB.02.01/VII/286/KEP.009/2018. TRIAL REGISTRATION NUMBER NCT03991910.
Collapse
Affiliation(s)
- Ade Meidian Ambari
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Budhi Setianto
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Anwar Santoso
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Basuni Radi
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Bambang Dwiputra
- Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, West Jakarta, Jakarta, Indonesia
| | - Eliana Susilowati
- Research Assistant of Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
| | - Fadilla Tulrahmi
- Research Assistant of Department of Cardiovascular Prevention and Rehabilitation, National Cardiovascular Center Harapan Kita, West Jakarta, Jakarta, Indonesia
| | - Annemiek Wind
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Pieter Doevendans
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Central Military Hospital, Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
16
|
Ataklte F, Vasan RS. Heart failure risk estimation based on novel biomarkers. Expert Rev Mol Diagn 2021; 21:655-672. [PMID: 34014781 DOI: 10.1080/14737159.2021.1933446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Despite advances in medical care, heart failure (HF)-associated morbidity and mortality remains high. Consequently, there is increased effort to find better ways for predicting, screening, and prognosticating HF in order to facilitate effective primary and secondary prevention.Areas covered: In this review, we describe the various biomarkers associated with different etiologic pathways implicated in HF, and discuss their roles in screening, diagnosing, prognosticating and predicting HF. We explore the emerging role of multi-omic approaches. We performed electronic searches in databases (PubMed and Google Scholar) through December 2020, using the following key terms: biomarker, novel, heart failure, risk, prediction, and estimation.Circulating BNP and troponin concentrations have been established in clinical care as key biomarkers for diagnosing and prognosticating HF. Emerging biomarkers (such as galectin-3 and ST-2) have gained further recognition for use in evaluating prognosis of HF patients. Promising biomarkers that are yet to be part of clinical recommendations include biomarkers of cardiorenal disease.Expert opinion: Increasing recognition of the complex and interdependent nature of pathophysiological pathways of HF has led to the application of multi-marker approaches including multi-omic high throughput assays. These newer approaches have the potential for new therapeutic discoveries and improving precision medicine in HF.
Collapse
Affiliation(s)
- Feven Ataklte
- Department of Internal Medicine, Boston Medical Center and Boston University School of Medicine, Boston, MA, USA
| | - Ramachandran S Vasan
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.,Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA.,Framingham Heart Study, Framingham, MA, USA.,Boston University Center for Computing and Data Sciences, Boston, MA, USA
| |
Collapse
|
17
|
Veraar C, Koschutnik M, Nitsche C, Laggner M, Polak D, Bohle B, Mangold A, Moser B, Mascherbauer J, Ankersmit HJ. Inflammatory immune response in recipients of transcatheter aortic valves. JTCVS OPEN 2021; 6:85-96. [PMID: 36003560 PMCID: PMC9390500 DOI: 10.1016/j.xjon.2021.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 01/17/2023]
Abstract
Objective Transcatheter aortic valve implantation (TAVI) is rapidly replacing cardiac surgery due to its minimal invasiveness and practicality. Midterm immunological studies on the biocompatibility of galactose-alpha-1,3-galactose (α-Gal)–carrying bioprosthetic heart valves for TAVI are not available. In this study we investigated whether bioprosthetic heart valves employed for TAVI augment an α-Gal–specific antibody-dependent and antibody-independent immune response 3 months after TAVI implantation. Methods This prospective observational study included 27 patients with severe aortic valve stenosis undergoing TAVI and 10 patients with severe mitral valve regurgitation treated with a transcatheter MitraClip (Abbott Laboratories, Abbott Park, Ill) procedure. Blood samples were drawn before and 90 days after treatment at a routine checkup. Serum samples were analyzed using enzyme-linked immunosorbent assay. Serum concentrations of α-Gal–specific immunoglobulin (Ig) G, IgG subclasses and IgE, complement factor 3a, NETosis-specific citrullinated H3, and the systemic inflammation markers soluble suppression of tumorigenicity and interleukin 33 were evaluated. Results Three months after TAVI, we found significantly increased serum concentrations of α-Gal–specific IgG3, complement factor complement factor 3a, citrullinated H3 levels, and soluble suppression of tumorigenicity (P = .002, P = .001, P = .025, and P = .039, respectively). Sensitization of α-Gal–specific IgE antibodies occurred in 55% of all patients after TAVI. Conclusions Our results indicate that TAVI elicits a midterm, specific humoral immune response against α-Gal and causes an unspecific humoral inflammation compared with patients undergoing MitraClip implantation. This observation will lead to a better understanding of postintervention morbidity and the long-term durability of bioprostheses and indicates that caution is appropriate when designing implantation strategies for younger patients.
Collapse
Affiliation(s)
- Cecilia Veraar
- Division of Cardiothoracic and Vascular Anaesthesia and Intensive Care Medicine, Department of Anaesthesiology, General Intensive Care, and Pain Medicine, Medical University of Vienna, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Vienna, Austria
| | - Matthias Koschutnik
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christian Nitsche
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Maria Laggner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Dominika Polak
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Andreas Mangold
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Julia Mascherbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine III, University Hospital St. Pölten, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Hendrik J. Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Address for reprints: Hendrik J. Ankersmit, MD, MBA, Department of Thoracic Surgery, Laboratory for Cardiac and Thoracic Diagnosis, Regeneration, and Applied Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
18
|
Sundnes O, Ottestad W, Schjalm C, Lundbäck P, la Cour Poulsen L, Mollnes TE, Haraldsen G, Eken T. Rapid systemic surge of IL-33 after severe human trauma: a prospective observational study. Mol Med 2021; 27:29. [PMID: 33771098 PMCID: PMC8004436 DOI: 10.1186/s10020-021-00288-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 03/08/2021] [Indexed: 01/12/2023] Open
Abstract
Background Alarmins are considered proximal mediators of the immune response after tissue injury. Understanding their biology could pave the way for development of new therapeutic targets and biomarkers in human disease, including multiple trauma. In this study we explored high-resolution concentration kinetics of the alarmin interleukin-33 (IL-33) early after human trauma. Methods Plasma samples were serially collected from 136 trauma patients immediately after hospital admission, 2, 4, 6, and 8 h thereafter, and every morning in the ICU. Levels of IL-33 and its decoy receptor sST2 were measured by immunoassays. Results We observed a rapid and transient surge of IL-33 in a subset of critically injured patients. These patients had more widespread tissue injuries and a greater degree of early coagulopathy. IL-33 half-life (t1/2) was 1.4 h (95% CI 1.2–1.6). sST2 displayed a distinctly different pattern with low initial levels but massive increase at later time points. Conclusions We describe for the first time early high-resolution IL-33 concentration kinetics in individual patients after trauma and correlate systemic IL-33 release to clinical data. These findings provide insight into a potentially important axis of danger signaling in humans. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00288-1.
Collapse
Affiliation(s)
- Olav Sundnes
- K.G Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Rikshospitalet, N-0027, Oslo, Norway.,Department of Dermatology, Oslo University Hospital, Oslo, Norway
| | - William Ottestad
- Department of Anaesthesiology, Division of Emergencies and Critical Care, Oslo University Hospital Ullevål, Oslo, Norway.,Division of Critical Care, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Camilla Schjalm
- K.G Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Peter Lundbäck
- K.G Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Rikshospitalet, N-0027, Oslo, Norway
| | - Lars la Cour Poulsen
- K.G Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Oslo University Hospital, Rikshospitalet, N-0027, Oslo, Norway
| | - Tom Eirik Mollnes
- K.G Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Immunology, Oslo University Hospital, Oslo, Norway.,Reserach Laboratory, Nordland Hospital, Bodø, and K.G.Jebsen TREC, University of Tromsø, Tromsø, Norway.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Guttorm Haraldsen
- K.G Jebsen Inflammation Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway. .,Department of Pathology, Oslo University Hospital, Rikshospitalet, N-0027, Oslo, Norway.
| | - Torsten Eken
- Department of Anaesthesiology, Division of Emergencies and Critical Care, Oslo University Hospital Ullevål, Oslo, Norway.,Division of Critical Care, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
19
|
Early sST2 Liberation after Implantation of a Left Ventricular Assist Device in Patients with Advanced Heart Failure. J Immunol Res 2020; 2020:5826176. [PMID: 36301686 PMCID: PMC7781699 DOI: 10.1155/2020/5826176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 11/24/2020] [Accepted: 12/05/2020] [Indexed: 11/18/2022] Open
Abstract
Background The use of left ventricular assist device (LVAD) has increased considerably over the past decade; however, there is limited literature to assist in patient selection and monitoring. The frequency of adverse events remains high. We examined the early expression of circulating soluble ST2 (sST2), a biomarker with immunosuppressive and profibrotic activity, and assessed the risk of death at 1 year in patients receiving LVAD implant. Methods We prospectively enrolled 20 heart failure patients and measured sST2, IL-33, and IL-6 serum concentrations over three weeks after LVAD implantation. We compared the kinetics of IL-6, sST2, and IL-33 release in survivors with those of nonsurvivors using mixed model two-way analysis of variance for repeated measures. We also collected data on hemodynamic parameters (i.e., cardiac output) and frequency of infections during the hospital stay. Results LVAD therapy led to an immediate and significant improvement of the hemodynamic parameters in 1-year survivors and nonsurvivors alike. The 1-year survival rate was 65%. IL-6 concentrations showed a significant (p = 0.03) peak at admission to the intensive care unit following LVAD implantation, whereas sST2 levels were massively increased (p < 0.0003) on day 1. While 1-year survivors had persistently lower sST2 values compared to nonsurvivors during the first 3 weeks after LVAD implantation (p = 0.012), no differences were observed in the temporal pattern of IL-6 or IL-33. The odds of detecting Candida species in the bronchoalveolar lavage fluid were 14 times higher in nonsurvivors than in survivors (OR 13.7, CI 1.4-127, p = 0.02). Conclusion In patients implanted with LVAD, circulating sST2 levels and frequency of Candida colonisation were associated with higher mortality. Awareness of this early immune response can guide physicians in risk-benefit analysis.
Collapse
|
20
|
Cyriaque V, Géron A, Billon G, Nesme J, Werner J, Gillan DC, Sørensen SJ, Wattiez R. Metal-induced bacterial interactions promote diversity in river-sediment microbiomes. FEMS Microbiol Ecol 2020; 96:5826176. [PMID: 32343356 DOI: 10.1093/femsec/fiaa076] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/27/2020] [Indexed: 01/05/2023] Open
Abstract
Anthropogenic metal contamination results in long-term environmental selective pressure with unclear impacts on bacterial communities, which comprise key players in ecosystem functioning. Since metal contamination poses serious toxicity and bioaccumulation issues, assessing their impact on environmental microbiomes is important to respond to current environmental and health issues. Despite elevated metal concentrations, the river sedimentary microbiome near the MetalEurop foundry (France) shows unexpected higher diversity compared with the upstream control site. In this work, a follow-up of the microbial community assembly during a metal contamination event was performed in microcosms with periodic renewal of the supernatant river water. Sediments of the control site were gradually exposed to a mixture of metals (Cd, Cu, Pb and Zn) in order to reach similar concentrations to MetalEurop sediments. Illumina sequencing of 16S rRNA gene amplicons was performed. Metal-resistant genes, czcA and pbrA, as well as IncP plasmid content, were assessed by quantitative PCR. The outcomes of this study support previous in situ observations showing that metals act as community assembly managers, increasing diversity. This work revealed progressive adaptation of the sediment microbiome through the selection of different metal-resistant mechanisms and cross-species interactions involving public good-providing bacteria co-occurring with the rest of the community.
Collapse
Affiliation(s)
- Valentine Cyriaque
- Proteomics and Microbiology Laboratory, Research Institute for Biosciences, UMONS, 20 Place du Parc, 7000 Mons, Belgium
| | - Augustin Géron
- Proteomics and Microbiology Laboratory, Research Institute for Biosciences, UMONS, 20 Place du Parc, 7000 Mons, Belgium.,Division of Biological and Environmental Sciences, Faculty of Natural Sciences, University of Stirling, Stirling,FK9 4LA, UK
| | - Gabriel Billon
- Univ. Lille, CNRS, UMR 8516 - LASIRE - LAboratoire de Spectroscopie pour les Interactions, la Réactivité et l'Environnement, F-59000 Lille, France
| | - Joseph Nesme
- Section of Microbiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Johannes Werner
- Department of Biological Oceanography, Leibniz Institute of Baltic Sea Research, D-18119 Rostock, Germany
| | - David C Gillan
- Proteomics and Microbiology Laboratory, Research Institute for Biosciences, UMONS, 20 Place du Parc, 7000 Mons, Belgium
| | - Søren J Sørensen
- Section of Microbiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Ruddy Wattiez
- Proteomics and Microbiology Laboratory, Research Institute for Biosciences, UMONS, 20 Place du Parc, 7000 Mons, Belgium
| |
Collapse
|
21
|
Ruiz-Castilla M, Dos Santos B, Vizcaíno C, Baena J, Guilabert P, Marin-Corral J, Masclans JR, Roca O, Barret JP. Soluble suppression of tumorigenicity-2 predicts pneumonia in patients with inhalation injury: Results of a pilot study. Burns 2020; 47:906-913. [PMID: 33143991 DOI: 10.1016/j.burns.2020.10.010] [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: 01/28/2020] [Revised: 08/25/2020] [Accepted: 10/08/2020] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Several mechanisms play a role in the development of pneumonia after inhalation injury. Our aim was to analyze whether higher concentrations of inflammatory markers or of biomarkers of epithelial injury are associated with a higher incidence of pneumonia in patients with inhalation injury. MATERIAL AND METHODS Secondary analysis of a single-center prospective observational cohort pilot study, performed over a two-year period (2015-2017) at the Burns Unit of the Plastic and Reconstructive Surgery Department of Vall d'Hebron University Hospital. All patients aged 18 with suspected inhalation injury undergoing admission to the Burns Unit were included. Plasma biomarkers of the lung epithelium (RAGE and SP-D), inflammation markers (IL6, IL8), and IL33, as well as soluble suppression of tumorigenicity-2 (sST2) levels, were measured within the first 24 h of admission. RESULTS Twenty-four patients with inhalation injury were included. Eight (33.3%) developed pneumonia after a median of 7 (4-8) days of hospital stay. Patients with pneumonia presented higher plasma concentrations of sST2 (2853 [2356-3351] ng/mL vs 1352 [865-1839] ng/mL; p < 0.001), IL33 (1.95 [1.31-2.59] pg/mL vs 1.26 [1.07-1.45] pg/mL; p = 0.002) and IL8 (325.7 [221.6-430.0] pg/mL vs 174.1 [95.2-253.0] pg/mL; p = 0.017) on day 1 of inclusion. Plasma sST2 concentration in the first 24 h demonstrated excellent diagnostic accuracy for predicting the occurrence of pneumonia in patients with smoke inhalation (AUROC 0.929 [95%CI 0.818-1.000]). A cutoff point of ≥2825 ng/mL for sST2 had a sensitivity of 75% and a specificity of 100%. The risk ratio of pneumonia in patients with sST2 ≥ 2825 ng/mL was 7.14 ([95% CI 1.56-32.61]; p = 0.016). CONCLUSIONS Plasma sST2 in the first 24 h of admission predicts the occurrence of pneumonia in patients with inhalation injury.
Collapse
Affiliation(s)
- Mireia Ruiz-Castilla
- Plastic and Reconstructive Surgery Department, Hospital Quirónsalud Barcelona, Barcelona, Spain; Plastic and Reconstructive Surgery Department and Burns Unit, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute, Barcelona, Spain.
| | - Bruce Dos Santos
- Plastic and Reconstructive Surgery Department and Burns Unit, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Claudia Vizcaíno
- Critical Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Jacinto Baena
- Critical Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Patricia Guilabert
- Anesthesiology Department, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Judith Marin-Corral
- Critical Care Department, Parc de Salut Mar (Hospital del Mar) de Barcelona, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Joan R Masclans
- Critical Care Department, Parc de Salut Mar (Hospital del Mar) de Barcelona, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Ciber Enfermedades Respiratorias (Ciberes), Instituto de Salud Carlos III, Madrid, Spain
| | - Oriol Roca
- Critical Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute, Barcelona, Spain; Ciber Enfermedades Respiratorias (Ciberes), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan P Barret
- Plastic and Reconstructive Surgery Department and Burns Unit, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute, Barcelona, Spain
| |
Collapse
|
22
|
Nicolai O, Pötschke C, Schmoeckel K, Darisipudi MN, van der Linde J, Raafat D, Bröker BM. Antibody Production in Murine Polymicrobial Sepsis-Kinetics and Key Players. Front Immunol 2020; 11:828. [PMID: 32425951 PMCID: PMC7205023 DOI: 10.3389/fimmu.2020.00828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022] Open
Abstract
Although antigen-specific priming of antibody responses is impaired during sepsis, there is nevertheless a strong increase in IgM and IgG serum concentrations. Using colon ascendens stent peritonitis (CASP), a mouse model of polymicrobial abdominal sepsis, we observed substantial increases in IgM as well as IgG of all subclasses, starting at day 3 and peaking 2 weeks after sepsis induction. The dominant source of antibody-secreting cells was by far the spleen, with a minor contribution of the mesenteric lymph nodes. Remarkably, sepsis induction in splenectomized mice did not change the dynamics of the serum IgM/IgG reaction, indicating that the marginal zone B cells, which almost exclusively reside in the spleen, are dispensable in such a setting. Hence, in systemic bacterial infection, the function of the spleen as dominant niche of antibody-producing cells can be compensated by extra-splenic B cell populations as well as other lymphoid organs. Depletion of CD4+ T cells did not affect the IgM response, while it impaired IgG generation of all subclasses with the exception of IgG3. Taken together, our data demonstrate that the robust class-switched antibody response in sepsis encompasses both T cell-dependent and -independent components.
Collapse
Affiliation(s)
- Oliver Nicolai
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christian Pötschke
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Schmoeckel
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Murthy N Darisipudi
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Julia van der Linde
- Department of General Surgery, Visceral, Thoracic and Vascular Surgery, University Medicine Greifswald, Greifswald, Germany
| | - Dina Raafat
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany.,Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Barbara M Bröker
- Immunology Department, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
23
|
Homsak E, Gruson D. Soluble ST2: A complex and diverse role in several diseases. Clin Chim Acta 2020; 507:75-87. [PMID: 32305537 DOI: 10.1016/j.cca.2020.04.011] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022]
Abstract
The Suppression of Tumorigenicity 2 protein (ST2) is a member of the interleukin (IL) 1 receptor family with transmembrane (ST2L) and soluble (sST2) isoforms that are (over)expressed in several cells in different conditions and following various triggers (e.g. inflammation, stress). The ligand of ST2 is IL-33, which on binding to ST2L results in nuclear signalling and immunomodulatory action in various cells (tumour, immune, heart). sST2, that is released in the circulation, functions as a »decoy« receptor of IL-33 and inhibits IL-33/ST2L signalling and beneficial effects. The importance and role of the ST2/IL-33 axis and sST2 have been evaluated and confirmed in several inflammatory, cancer and cardiac diseases. sST2 is involved in homeostasis/pathogenesis of these diseases, as the counterbalance/response on IL-33/ST2L axis activation, which is triggered and expressed during developing fibrosis, tissue damage/inflammation and remodelling. In clinical studies, sST2 has been recognised as an important prognostic marker in patients with cardiac disease, including patients with chronic kidney disease where specific characteristics of sST2 enable better assessment of the risk of End-Stage Renal Disease patients on dialysis. sST2 is also recognised as an important marker for monitoring treatment in heart failure patients. However, accurate measurement and interpretation of ST2 concentration in serum/plasma samples for routine and research applications require the use of appropriate methods and recognition of essential characteristics of both the methods and the analyte that may influence the result. sST2, as one of the most promising disease biomarkers, is deserving of further study and wider application in clinical practice.
Collapse
Affiliation(s)
- Evgenija Homsak
- Department of Laboratory Diagnostics, University Medical Centre Maribor, Maribor, Slovenia.
| | - Damien Gruson
- Department of Laboratory Medicine, Cliniques Universitaires St-Luc and Universite Catholique de Lovain, Brussels, Belgium
| |
Collapse
|
24
|
Zach V, Bähr FL, Edelmann F. Suppression of Tumourigenicity 2 in Heart Failure With Preserved Ejection Fraction. Card Fail Rev 2020; 6:1-7. [PMID: 32257387 PMCID: PMC7111301 DOI: 10.15420/cfr.2019.10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF), with steadily increasing incidence rates and mortality in an ageing population, represents a major challenge. Evidence suggests that more than half of all patients with a diagnosis of HF suffer from HF with preserved ejection fraction (HFpEF). Emerging novel biomarkers to improve and potentially guide the treatment of HFpEF are the subject of discussion. One of these biomarkers is suppression of tumourigenicity 2 (ST2), a member of the interleukin (IL)-1 receptor family, binding to IL-33. Its two main isoforms – soluble ST2 (sST2) and transmembrane ST2 (ST2L) – show opposite effects in cardiovascular diseases. While the ST2L/IL-33 interaction is considered as being cardioprotective, sST2 antagonises this beneficial effect by competing for binding to IL-33. Recent studies show that elevated levels of sST2 are associated with increased mortality in HF with reduced ejection fraction. Nevertheless, the significance of sST2 in HFpEF remains uncertain. This article aims to give an overview of the current evidence on sST2 in HFpEF with an emphasis on prognostic value, clinical association and interaction with HF treatment. The authors conclude that sST2 is a promising biomarker in HFpEF. However, further research is needed to fully understand underlying mechanisms and ultimately assess its full value.
Collapse
Affiliation(s)
- Veronika Zach
- Department of Internal Medicine and Cardiology, Charité University Medicine Berlin Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin Berlin, Germany
| | - Felix Lucas Bähr
- Department of Internal Medicine and Cardiology, Charité University Medicine Berlin Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin Berlin, Germany
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology, Charité University Medicine Berlin Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin Berlin, Germany.,Berlin Institute of Health Berlin, Germany
| |
Collapse
|
25
|
Hasan A, Aldhahi W. Soluble Suppression of Tumorigenicity 2 is Directly Correlated with Glycated Hemoglobin in Individuals with an Average glycemia in the Normal/Prediabetes Range. Diabetes Metab Syndr Obes 2020; 13:2711-2718. [PMID: 32801817 PMCID: PMC7413703 DOI: 10.2147/dmso.s251135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/05/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Cardiovascular disease can be detected in individuals with prediabetes. The purpose of this study was to determine whether soluble suppression of tumorigenicity 2 (sST2), which is elevated in cardiovascular disease and/or type 2 diabetes, is correlated with glycated haemoglobin in individuals with glycemia in the normal/prediabetes range. PATIENTS AND METHODS The anthropometric, biochemical and metabolic parameters were measured in 30 adults, and the plasma levels of sST2 were quantified. RESULTS sST2 was directly correlated with glycated hemoglobin in individuals with glycemia in the normal/prediabetes range. Participants who were at the higher end of glycated hemoglobin (5.8-6.4%) had significantly higher sST2 compared to those at the lower end (≤5.5%). Moreover, sST2 was directly correlated with homeostatic model assessment of insulin resistance (HOMA-IR), alkaline phosphatase, and waist circumference. However, the correlation between sST2 and HOMA-IR or waist circumference was lost after adjusting for age, gender or body mass index. CONCLUSION Circulating sST2 may be used to establish a cut-off value for cardiometabolic risk/disease in individuals with glycemia in the normal/prediabetes range.
Collapse
Affiliation(s)
- Amal Hasan
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Kuwait City, Kuwait
- Correspondence: Amal Hasan Email
| | - Waleed Aldhahi
- Medical Division, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Medicine, Mubarak Al-Kabeer Hospital, Kuwait City, Kuwait
| |
Collapse
|
26
|
Kamardinov DK, Songurov RN, Ioshina VI, Buziashvili YI. [Soluble ST2 - as a biomarker, a tool for risk stratification and therapeutic target in patients with chronic heart failure]. ACTA ACUST UNITED AC 2019; 60:111-121. [PMID: 32345207 DOI: 10.18087/cardio.2020.2.n816] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/02/2019] [Indexed: 11/18/2022]
Abstract
This review focuses on possibilities of using soluble ST2 as a HF marker for diagnostics, stratification of risk of adverse events, and for evaluation of prognosis and treatment effectiveness in patients with CHF. Circulating biomarkers are an essential element of algorithms for diagnostics, stratification of risk, and evaluation of prognosis in patients with HF. The recognized "gold standard", natriuretic peptides, has several well-known limitations, and multiple new candidate biomarkers have appeared in recent years. Soluble ST2, a marker of "mechanical myocardial stress", is considered as one of the most promising new biomarkers. This review discusses possibilities of using it in clinical practice in CHF patients.
Collapse
Affiliation(s)
- D K Kamardinov
- Bakulev National Medical Research Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation
| | - R N Songurov
- Bakulev National Medical Research Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation
| | - V I Ioshina
- Bakulev National Medical Research Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation
| | - Yu I Buziashvili
- Bakulev National Medical Research Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation
| |
Collapse
|
27
|
Skvortsov AA, Narusov OY, Muksinova MD. [Soluble ST2 - biomarker for prognosis and monitoring in decompensated heart failure]. ACTA ACUST UNITED AC 2019; 59:18-27. [PMID: 31884937 DOI: 10.18087/cardio.n765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/10/2019] [Indexed: 11/18/2022]
Abstract
The review aims to appraise the value of determining the concentrations of the new biomarker sST2 for assessing prognosis and monitoring treatment effectiveness of patients with decompensated heart failure during an episode of decompensation and during long-term follow-up after discharge from the hospital. The article analyses in detail the expedience of sST2 measurement in a patient with ADHF on admission and discharge from the hospital and the changes in the biomarker level during the period of active treatment for risk-stratification in patients, presents the optimal threshold values of sST2, which should be oriented when selecting patients with high and very high risk. The importance of subsequent monitoring of the marker concentration during long-term observation in emphasized to predict the risk of death, HF re-decompensation / HF rehospitalization. The potential benefits of choosing sST2 as the optimal marker for serial measurement during long-term follow-up, as well as evaluating the treatment effectiveness in patients with HF, compared to the "classical" variant - natriuretic peptides are shown.
Collapse
Affiliation(s)
- A A Skvortsov
- FSBO National Medical research center of cardiology of the Ministry of healthcare of the Russian Federation
| | - O Yu Narusov
- FSBO National Medical research center of cardiology of the Ministry of healthcare of the Russian Federation
| | - M D Muksinova
- FSBO National Medical research center of cardiology of the Ministry of healthcare of the Russian Federation
| |
Collapse
|
28
|
Torrungruang K, Katudat D, Mahanonda R, Sritara P, Udomsak A. Periodontitis is associated with elevated serum levels of cardiac biomarkers—Soluble ST2 and C‐reactive protein. J Clin Periodontol 2019; 46:809-818. [DOI: 10.1111/jcpe.13149] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/30/2019] [Accepted: 05/20/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Kitti Torrungruang
- Department of Microbiology, Faculty of Dentistry Chulalongkorn University Bangkok Thailand
| | | | - Rangsini Mahanonda
- Department of Periodontology, Faculty of Dentistry Chulalongkorn University Bangkok Thailand
| | - Piyamitr Sritara
- Cardiology Division, Department of Internal Medicine, Faculty of Medicine Ramathibodi Hospital Mahidol University Bangkok Thailand
| | - Artit Udomsak
- Medical and Health Department Electricity Generating Authority of Thailand Nonthaburi Thailand
| |
Collapse
|
29
|
Vorlat A, De Hous N, Vervaecke AJ, Vermeulen T, Van Craenenbroeck E, Heidbuchel H, Rodrigus I, Van Donink W, Ancion A, Van Cleemput J, Van Hoof VO, Claeys MJ. Biomarkers and Donor Selection in Heart Transplantation. Transplant Proc 2019; 51:1673-1678. [PMID: 31307770 DOI: 10.1016/j.transproceed.2019.04.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/19/2019] [Accepted: 04/05/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Previously, we showed that B-type natriuretic peptide (BNP) measured in the donor was related to cardiac performance after cardiac transplantation. The present study assesses the value of 3 biomarkers in the selection of donor hearts in a larger cohort. METHODS Blood samples were prospectively obtained in 105 brain-dead patients scheduled for heart donation. BNP, soluble suppressor of tumorigenicity 2 (ST2), and troponin of heart donors were correlated with hemodynamic parameters early after transplantation as well as with the mortality of the recipients. RESULTS A significant inverse relationship was found between donor BNP measured at the time of donation and recipient cardiac index and cardiac output at day 13 post-transplantation (r = -0.31, P = .005, and r = -0.34, P = .0016, respectively). Logistic regression analysis-including BNP, ST2, and troponin-showed that donor BNP was a predictor of a poor cardiac index (< 2.2 L/min/m2) in the recipient (P = .04). A donor BNP > 132 pg/mL has a sensitivity of 56% (95% confidence interval 21-86) and a specificity of 86% (95% confidence interval 77-93) to predict poor cardiac performance in the recipient. When the donor BNP is ≤ 132 pg/mL, the risk of a poor cardiac function in the recipient is very low (negative predictive value 94%). Mortality at 30 days was also correlated to donor BNP (r = 0.29, P = .0029). Long-term survival of the recipient was not correlated to the biomarkers measured in the donor. CONCLUSION Donor BNP, but not donor ST2 or high-sensitivity troponin, provides information on the donor heart and early post-transplant performance, including 1-month mortality.
Collapse
Affiliation(s)
- Anne Vorlat
- Department of Cardiology, University Hospital of Antwerp, Edegem, Belgium.
| | - Nicolas De Hous
- Department of Cardiology, University Hospital of Antwerp, Edegem, Belgium
| | | | - Tom Vermeulen
- Department of Cardiology, University Hospital of Antwerp, Edegem, Belgium
| | | | - Hein Heidbuchel
- Department of Cardiology, University Hospital of Antwerp, Edegem, Belgium
| | - Inez Rodrigus
- Department of Cardiac Surgery, University Hospital of Antwerp, Edegem, Belgium
| | - Walter Van Donink
- Department of Cardiac Surgery, University Hospital of Antwerp, Edegem, Belgium
| | - Arnaud Ancion
- Department of Cardiology, University Hospital Sart Tilman, Liege, Belgium
| | - Johan Van Cleemput
- Department of Cardiology, University Hospital of Leuven, Leuven, Belgium
| | - Viviane O Van Hoof
- Department of Clinical Chemistry, Antwerp University Hospital, Edegem, Belgium; Translational Pathophysiological Research Group, University of Antwerp, Edegem, Belgium
| | - Marc J Claeys
- Department of Cardiology, University Hospital of Antwerp, Edegem, Belgium
| |
Collapse
|
30
|
Elevations in Circulating sST2 Levels Are Associated With In-Hospital Mortality and Adverse Clinical Outcomes After Blunt Trauma. J Surg Res 2019; 244:23-33. [PMID: 31279260 DOI: 10.1016/j.jss.2019.05.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/09/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Soluble suppression of tumorigenicity 2 (sST2), a decoy receptor for interleukin (IL)-33, has emerged as a novel biomarker in various disease processes. Recent studies have elucidated the role of the sST2/IL-33 complex in modulating the balance of Th1/Th2 immune responses after tissue stress. However, the role of sST2 as a biomarker after traumatic injury remains unclear. To address this, we evaluated serum sST2 correlations with mortality and in-hospital adverse outcomes as endpoints in blunt trauma patients. METHODS We retrospectively analyzed clinical and biobank data of 493 blunt trauma victims 472 survivors (mean age: 48.4 ± 0.87; injury severity score [ISS]: 19.6 ± 0.48) and 19 nonsurvivors (mean age: 58.8 ± 4.5; ISS: 23.3 ± 2.1) admitted to the intensive care unit. Given the confounding impact of age on the inflammatory response, we derived a propensity-matched survivor subgroup (n = 19; mean age: 59 ± 3; ISS: 23.4 ± 2) using an IBM SPSS case-control matching algorithm. Serial blood samples were obtained from all patients (3 samples within the first 24 h and then once daily from day [D] 1 to D5 after injury). sST2 and twenty-nine inflammatory biomarkers were assayed using enzyme-linked immunosorbent assay and Luminex, respectively. Two-way analysis of variance on ranks was used to compare groups (P < 0.05). Spearman rank correlation was performed to determine the association of circulating sST2 levels with biomarker levels and in-hospital clinical outcomes. RESULTS Circulating sST2 levels of the nonsurvivor cohort were statistically significantly elevated at 12 h after injury and remained elevated up to D5 when compared either to the overall 472 survivor cohort or a matched 19 survivor subcohort. Admission sST2 levels obtained from the first blood draw after injury in the survivor cohort correlated positively with admission base deficit (correlation coefficient [CC] = 0.1; P = 0.02), international normalized ratio (CC = 0.1, P = 0.03), ISS (CC = 0.1, P = 0.008), and the average Marshall multiple organ dysfunction score between D2 and D5 (CC = 0.1, P = 0.04). Correlations with ISS revealed a positive correlation of ISS with plasma sST2 levels across the mild ISS (CC = 0.47, P < 0.001), moderate ISS (CC = 0.58, P < 0.001), and severe ISS groups (CC = 0.63, P < 0.001). Analysis of biomarker correlations in the matched survivor group over the initial 24 h after injury showed that sST2 correlates strongly and positively with IL-4 (CC = 0.65, P = 0.002), IL-5 (CC = 0.57, P = 0.01), IL-21 (CC = 0.52, P = 0.02), IL-2 (CC = 0.51, P = 0.02), soluble IL-2 receptor-α (CC = 0.5, P = 0.02), IL-13 (CC = 0.49, P = 0.02), and IL-17A (CC = 0.48, P = 0.03). This was not seen in the matched nonsurvivor group. sST2/IL-33 ratios were significantly elevated in nonsurvivors and patients with severe injury based on ISS ≥ 25. CONCLUSIONS Elevations in serum sST2 levels are associated with poor clinical trajectories and mortality after blunt trauma. High sST2 coupled with low IL-33 associates with severe injury, mortality, and worse clinical outcomes. These findings suggest that sST2 could serve as an early prognostic biomarker in trauma patients and that sustained elevations of sST2 could contribute to a detrimental suppression of IL-33 bioavailability in patients with high injury severity.
Collapse
|
31
|
Frisbee AL, Saleh MM, Young MK, Leslie JL, Simpson ME, Abhyankar MM, Cowardin CA, Ma JZ, Pramoonjago P, Turner SD, Liou AP, Buonomo EL, Petri WA. IL-33 drives group 2 innate lymphoid cell-mediated protection during Clostridium difficile infection. Nat Commun 2019; 10:2712. [PMID: 31221971 PMCID: PMC6586630 DOI: 10.1038/s41467-019-10733-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
Clostridium difficile (C. difficile) incidence has tripled over the past 15 years and is attributed to the emergence of hypervirulent strains. While it is clear that C. difficile toxins cause damaging colonic inflammation, the immune mechanisms protecting from tissue damage require further investigation. Through a transcriptome analysis, we identify IL-33 as an immune target upregulated in response to hypervirulent C. difficile. We demonstrate that IL-33 prevents C. difficile-associated mortality and epithelial disruption independently of bacterial burden or toxin expression. IL-33 drives colonic group 2 innate lymphoid cell (ILC2) activation during infection and IL-33 activated ILC2s are sufficient to prevent disease. Furthermore, intestinal IL-33 expression is regulated by the microbiota as fecal microbiota transplantation (FMT) rescues antibiotic-associated depletion of IL-33. Lastly, dysregulated IL-33 signaling via the decoy receptor, sST2, predicts C. difficile-associated mortality in human patients. Thus, IL-33 signaling to ILC2s is an important mechanism of defense from C. difficile colitis. Here, Frisbee et al. show that hypervirulent Clostridium difficile induces IL-33 expression in the gut and IL-33 reduces mortality and morbidity via group 2 innate lymphoid cells. Furthermore, serum levels of the soluble IL-33 decoy receptor, sST2, are associated with enhanced disease severity in human C. difficile patients.
Collapse
Affiliation(s)
- Alyse L Frisbee
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Mahmoud M Saleh
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Mary K Young
- Department of Medicine, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Jhansi L Leslie
- Department of Medicine, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Morgan E Simpson
- Department of Pathology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Mayuresh M Abhyankar
- Department of Medicine, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Carrie A Cowardin
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Jennie Z Ma
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Patcharin Pramoonjago
- Department of Pathology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Stephen D Turner
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | | | - Erica L Buonomo
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - William A Petri
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA. .,Department of Medicine, University of Virginia Health System, Charlottesville, VA, 22908, USA. .,Department of Pathology, University of Virginia Health System, Charlottesville, VA, 22908, USA.
| |
Collapse
|
32
|
Fields JK, Günther S, Sundberg EJ. Structural Basis of IL-1 Family Cytokine Signaling. Front Immunol 2019; 10:1412. [PMID: 31281320 PMCID: PMC6596353 DOI: 10.3389/fimmu.2019.01412] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/04/2019] [Indexed: 01/13/2023] Open
Abstract
Interleukin-1 (IL-1) family cytokines are key signaling molecules in both the innate and adaptive immune systems, mediating inflammation in response to a wide range of stimuli. The basic mechanism of signal initiation is a stepwise process in which an agonist cytokine binds its cognate receptor. Together, this cytokine-receptor complex recruits an often-common secondary receptor. Intracellularly, the Toll/IL-1 Receptor (TIR) domains of the two receptors are brought into close proximity, initiating an NF-κB signal transduction cascade. Due to the potent inflammatory response invoked by IL-1 family cytokines, several physiological mechanisms exist to inhibit IL-1 family signaling, including antagonist cytokines and decoy receptors. The numerous cytokines and receptors in the IL-1 superfamily are further classified into four subfamilies, dependent on their distinct cognate receptors—the IL-1, IL-33, and IL-36 subfamilies share IL-1RAcP as their secondary receptor, while IL-18 subfamily utilizes a distinct secondary receptor. Here, we describe how structural biology has informed our understanding of IL-1 family cytokine signaling, with a particular focus on molecular mechanisms of signaling complex formation and antagonism at the atomic level, as well as how these findings have advanced therapeutics to treat some chronic inflammatory diseases that are the result of dysregulated IL-1 signaling.
Collapse
Affiliation(s)
- James K Fields
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, United States.,Program in Molecular Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Eric J Sundberg
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
33
|
Najjar E, Faxén UL, Hage C, Donal E, Daubert JC, Linde C, Lund LH. ST2 in heart failure with preserved and reduced ejection fraction. SCAND CARDIOVASC J 2019; 53:21-27. [DOI: 10.1080/14017431.2019.1583363] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Emil Najjar
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrika Ljung Faxén
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Perioperative Medicine and Intensive Care Function, Karolinska University Hospital, Stockholm, Sweden
| | - Camilla Hage
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Erwan Donal
- Département de Cardiologie & CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France
| | - Jean-Claude Daubert
- Département de Cardiologie & CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France
| | - Cecilia Linde
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Lars H. Lund
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
34
|
Ge Y, Huang M, Yao YM. Recent advances in the biology of IL-1 family cytokines and their potential roles in development of sepsis. Cytokine Growth Factor Rev 2018; 45:24-34. [PMID: 30587411 DOI: 10.1016/j.cytogfr.2018.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/16/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Abstract
The IL-1 family comprises two anti-inflammatory cytokines (IL-37, IL-38), two receptor antagonists (IL-1ra, IL-36ra), and seven ligand agonists (IL-1α, IL-1β, IL-33, IL-36α, IL-36β, IL-36γ). The members of this family exert pleiotropic effects on intercellular signaling, leading to pro- or anti-inflammatory responses. They initiate potent inflammatory and immune responses by binding to specific receptors in the IL-1 receptor family, and their activities are repressed by naturally occurring inhibitors. Various immune cells produce and are regulated by these crucial molecules, which appear to be involved in the pathogenesis of diverse diseases including cancer as well as inflammatory and autoimmune disorders. Recent decades have seen substantial progress in understanding how the IL-1 family contributes to the development of sepsis. In this review, we will briefly introduce the IL-1 family and discuss its critical role in inflammatory and immune responses. The potential significance of IL-1 members in sepsis will also be explored, together with the clinical implications for treating this dangerous condition.
Collapse
Affiliation(s)
- Yun Ge
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Man Huang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
35
|
Pascual-Figal DA, Pérez-Martínez MT, Asensio-Lopez MC, Sanchez-Más J, García-García ME, Martinez CM, Lencina M, Jara R, Januzzi JL, Lax A. Pulmonary Production of Soluble ST2 in Heart Failure. Circ Heart Fail 2018; 11:e005488. [DOI: 10.1161/circheartfailure.118.005488] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Domingo A. Pascual-Figal
- Department of Cardiology, Hospital Virgen de la Arrixaca, University of Murcia, Spain (D.A.P.-F., M.T.P.-M., M.C.A.-L., A.L.)
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (D.A.P.-F.)
- CIBERCV, Madrid, Spain (D.A.P.-F.)
| | - Maria T. Pérez-Martínez
- Department of Cardiology, Hospital Virgen de la Arrixaca, University of Murcia, Spain (D.A.P.-F., M.T.P.-M., M.C.A.-L., A.L.)
- IMIB-Arrixaca, University of Murcia, Spain (M.T.P.-M., M.C.A.-L., C.M.M., A.L.)
| | - Maria C. Asensio-Lopez
- Department of Cardiology, Hospital Virgen de la Arrixaca, University of Murcia, Spain (D.A.P.-F., M.T.P.-M., M.C.A.-L., A.L.)
| | - Jesús Sanchez-Más
- Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera, Moncada, Valencia, Spain (J.S.-M.)
| | - Maria E. García-García
- Servicio de Medicina Intensiva, Hospital Virgen de la Arrixaca, Hospital Virgen de la Arrixaca, Murcia, Spain (M.E.G.-G., R.J.)
| | - Carlos M. Martinez
- IMIB-Arrixaca, University of Murcia, Spain (M.T.P.-M., M.C.A.-L., C.M.M., A.L.)
| | - Miriam Lencina
- Department of Pathological Anatomy, Hospital Virgen de la Arrixaca, Murcia, Spain (M.L.)
| | - Ruben Jara
- Servicio de Medicina Intensiva, Hospital Virgen de la Arrixaca, Hospital Virgen de la Arrixaca, Murcia, Spain (M.E.G.-G., R.J.)
| | - James L. Januzzi
- Division of Cardiology, Massachusetts General Hospital, Baim Institute for Clinical Research, Boston (J.L.J.)
| | - Antonio Lax
- IMIB-Arrixaca, University of Murcia, Spain (M.T.P.-M., M.C.A.-L., C.M.M., A.L.)
| |
Collapse
|
36
|
Jha D, Goenka L, Ramamoorthy T, Sharma M, Dhandapani VE, George M. Prognostic role of soluble ST2 in acute coronary syndrome with diabetes. Eur J Clin Invest 2018; 48:e12994. [PMID: 29992539 DOI: 10.1111/eci.12994] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/15/2018] [Accepted: 07/07/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Acute coronary syndrome (ACS) patients are at an increased risk of major adverse cardiovascular events (MACE). The objective of our study was to assess whether cardiac biomarker like soluble ST2 (sST2) can predict MACE among ACS patients with diabetes. MATERIALS AND METHODS A total of 122 patients with ACS were included in the study. sST2 level in blood plasma samples was quantified using enzyme-linked immunosorbent assay (ELISA). Prognostic utility of sST2 for the primary outcome of MACE which included mortality, rehospitalization due to chest pain, unstable angina, recurrent myocardial infarction (MI) and stroke, was assessed during follow-up. RESULTS The median follow-up period was of 180 days. ROC (receiver operating characteristic) curve demonstrated that elevated levels of sST2 were able to predict mortality, and MACE in ACS patients, along with increased risk of occurrence of MACE and mortality in ACS patients having diabetes. Kaplan-Meier plots revealed a significant increase in the occurrence of MACE in diabetic ACS patients (P = 0.006; by log-rank test). Cox regression analysis revealed that sST2 is not an independent predictor of mortality and MACE in ACS patients having diabetes; however, high sST2 level was found to be a predictor of MACE in all ACS subjects in the fully adjusted model with a hazard ratio (HR) of 5.8 (P = 0.032). CONCLUSION The current study indicates that elevated levels of sST2 might be a suitable biomarker to evaluate the risk of future adverse cardiovascular events in ACS patients with diabetes.
Collapse
Affiliation(s)
- Durga Jha
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, TamilNadu, India
| | - Luxitaa Goenka
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, TamilNadu, India
| | - Thilagavathi Ramamoorthy
- Division of Biostatistics, School of Public Health, SRM University, Kattankulathur, TamilNadu, India
| | - Masum Sharma
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, TamilNadu, India
| | - Vellala E Dhandapani
- Department of Cardiology, SRM Medical College Hospital & Research Centre, Kattankulathur, TamilNadu, India
| | - Melvin George
- Department of Clinical Pharmacology, SRM Medical College Hospital & Research Centre, Kattankulathur, TamilNadu, India
| |
Collapse
|
37
|
Babic ZM, Zunic FZ, Pantic JM, Radosavljevic GD, Jovanovic IP, Arsenijevic NN, Lukic ML. IL-33 receptor (ST2) deficiency downregulates myeloid precursors, inflammatory NK and dendritic cells in early phase of sepsis. J Biomed Sci 2018; 25:56. [PMID: 30001716 PMCID: PMC6044035 DOI: 10.1186/s12929-018-0455-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Sepsis is a life-threatening disease mediated by profound disturbances in systemic inflammatory response to infection. IL-33 is multifunctional regulator of numerous aspects of innate and adaptive immune response. The aim of this article was to further evaluate the role of IL-33 receptor (ST2) in different pathways of innate immunity during early polymicrobial sepsis. METHODS Polymicrobial sepsis was induced using cecal ligation and puncture (CLP) model in ST2 deficient (ST2-/-) and wild type BALB/c mice. Peritoneal and spleen cells were isolated for further phenotyping. Apoptosis was determined by immunohistochemistry and flow cytometry. RESULTS Deletion of ST2 leads to increased susceptibility to early manifestations of sepsis as evaluated by clinical signs and survival. These are accompanied by decrease in the total number of neutrophils, eosinophils and mast cells in peritoneal cavity 12 h after CLP. In early sepsis there was also low number of precursors of myeloid cells in particular CD11b+Ly6G+Ly6Clow cells in spleen of ST2-/- mice. Although the number of NK cells in the spleen was similar, there were significant differences in the presence of inflammatory IFN-γ and IL-17 producing NK cells. Further, ST2 deletion affects the phenotype and maturation of dendritic cell in sepsis. The total number of dendritic cells in the spleen was lower as well as IL-12 expressing dendritic cells. Finally, there was higher frequency of active caspase-3 positive and early apoptotic cells, in particular CD11c positive cells, in spleen of septic ST2-/- mice. CONCLUSION Taken together, our data provide the evidence that ST2 deficiency in early phase of sepsis downregulates myeloid precursors, inflammatory NK and dendritic cells.
Collapse
Affiliation(s)
- Zivan M. Babic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Filip Z. Zunic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jelena M. Pantic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, Kragujevac, 34000 Serbia
| | - Gordana D. Radosavljevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, Kragujevac, 34000 Serbia
| | - Ivan P. Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, Kragujevac, 34000 Serbia
| | - Nebojsa N. Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, Kragujevac, 34000 Serbia
| | - Miodrag L. Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, Kragujevac, 34000 Serbia
| |
Collapse
|
38
|
Abstract
Suppression of tumorigenicity 2 (ST2) is a member of the interleukin (IL)-1 receptor family, whose role was originally established in the context of inflammatory and autoimmune diseases. More recently, testing for ST2 has been used in the setting of cardiovascular disease. The soluble form of ST2 is a decoy receptor that inhibits beneficial cardioprotective effects of IL-33; such inhibition results in cardiac hypertrophy, myocardial fibrosis, and ventricular dysfunction. Measurement of soluble ST2 has utility for assessing heart failure severity and prognosis. In this review, we examine the role of soluble ST2 in both acute and chronic heart failure.
Collapse
Affiliation(s)
- Cian P McCarthy
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - James L Januzzi
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 32 Fruit Street, Yawkey 5984, Boston, MA 02114, USA; Baim Institute for Clinical Research, Cardiometabolic Trials, 930 Commonwealth Avenue, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Mitchell PD, Salter BM, Oliveria JP, El-Gammal A, Tworek D, Smith SG, Sehmi R, Gauvreau GM, O Apos Byrne PM. IL-33 and Its Receptor ST2 after Inhaled Allergen Challenge in Allergic Asthmatics. Int Arch Allergy Immunol 2018; 176:133-142. [PMID: 29694974 DOI: 10.1159/000488015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/12/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous murine models have demonstrated interleukin (IL)-33 to be an important mediator of type-2 inflammation and to promote airway hyperresponsiveness in allergic asthma. A number of inflammatory cells produce IL-33 and eosinophils express ST2 mRNA. The relationship between IL-33 and eosinophils in allergic asthma, however, remains unclear. OBJECTIVE The aim of this work was to evaluate in vitro the effect of allergen inhalation on IL-33 levels and expression of its receptor (ST2L) on eosinophils in allergic asthmatics, and the effect of IL-33 stimulation on eosinophil activity. METHODS Plasma and sputum IL-33, soluble ST2 (sST2) levels, and ST2L expression on eosinophils were measured in 10 healthy controls and 10 allergic asthmatics. Asthmatics underwent allergen and diluent inhalation challenges. Blood and sputum samples were collected to measure IL-33, sST2, and ST2L eosinophil expression before and 24 h after allergen inhalation. Purified blood eosinophils from allergic asthmatics were incubated overnight with IL-33 to assess ST2 and intracellular IL-5 expression. RESULTS Baseline levels of IL-33 in sputum and sST2 in plasma and sputum were similar in allergic asthmatics compared to healthy controls. In addition, there was no difference in blood or sputum eosinophil ST2L expression in healthy controls versus allergic asthmatics. Eosinophil ST2L expression was significantly increased 24 h postallergen inhalation in allergic asthmatics. In vitro stimulation of human eosinophils with IL-33 and LPS significantly increased eosinophil ST2L expression and IL-33 stimulation increased intracellular IL-5 expression, which was attenuated by treatment with sST2 and ST2 blockade. CONCLUSION AND CLINICAL RELEVANCE In mild asthmatics, there was a significant upregulation of ST2 surface expression on eosinophils from blood and sputum following allergen inhalation challenge. In vitro, IL-33 stimulation of eosinophils increases both ST2 membrane expression and IL-5 production. These results support a role for IL-33 in causing allergen-induced eosinophilia. Blockade of IL-33 and ST2 signaling may present a novel therapeutic avenue for asthma treatment.
Collapse
|
40
|
Li L, Sun Z, Xu C, Wu J, Liu G, Cui H, Chen H. Adenovirus-mediated overexpression of sST2 attenuates cardiac injury in the rat with severe acute pancreatitis. Life Sci 2018; 202:167-174. [PMID: 29653119 DOI: 10.1016/j.lfs.2018.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/29/2018] [Accepted: 04/08/2018] [Indexed: 12/12/2022]
Abstract
AIMS Severe acute pancreatitis (SAP) is a serious disease associated with systematic inflammation and multiple organs dysfunction. Soluble ST2 (sST2), a member of the Toll interleukin (IL)-1 receptor (TIR) superfamily, has been demonstrated to exert immune-regulatory and anti-inflammatory properties in several inflammation-related diseases. In this study, we investigated whether transfer of sST2 gene by adenovirus vector could attenuate sodium taurocholate-induced SAP and associated cardiac injury. MAIN METHODS A rat model of SAP was induced by retrograde injection of 5% sodium taurocholate (1 ml/kg) into the biliopancreatic duct. Rats in the treatment groups were intravenously injected with adenovirus expressing sST2 (Ad-sST2, 1 × 109 particles/rat) or green fluorescent protein (Ad-GFP) via the tail vein 48 h before SAP induction. Histological changes in the pancreatic and heart tissues, and parameters for evaluating SAP and associated cardiac injury were determined at 24 h after SAP. KEY FINDINGS Sodium taurocholate induced obvious pathological changes in pancreas and elevated serum levels of amylase and lipase. Furthermore, SAP animals exhibited significant cardiac impairment, evidenced by decreased cardiac function, increased myocardial apoptosis and cardiac-related enzymes including creatine kinase isoenzyme, lactate dehydrogenase, and Troponin T. Administration of Ad-sST2 markedly improved the structure of pancreas and heart tissues, and reversed the alterations in serum amylase, lipase and cardiac-related enzymes. In addition, Ad-sST2 treatment downregulated pro-inflammatory cytokines production, demonstrating the anti-inflammatory property of sST2. SIGNIFICANCE Our results suggest that administration of Ad-sST2 significantly attenuated the severity of SAP and associated cardiac damage, and the cardioprotective effect is associated with its anti-inflammatory action.
Collapse
Affiliation(s)
- Lei Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, People's Republic of China; Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Zhongwei Sun
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, People's Republic of China
| | - Caiming Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, People's Republic of China
| | - Jun Wu
- Department of Ultrasound, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Geliang Liu
- Department of Urology Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Hongzhang Cui
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, People's Republic of China
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, People's Republic of China.
| |
Collapse
|
41
|
Schmoeckel K, Mrochen DM, Hühn J, Pötschke C, Bröker BM. Polymicrobial sepsis and non-specific immunization induce adaptive immunosuppression to a similar degree. PLoS One 2018; 13:e0192197. [PMID: 29415028 PMCID: PMC5802895 DOI: 10.1371/journal.pone.0192197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/19/2018] [Indexed: 11/18/2022] Open
Abstract
Sepsis is frequently complicated by a state of profound immunosuppression, in its extreme form known as immunoparalysis. We have studied the role of the adaptive immune system in the murine acute peritonitis model. To read out adaptive immunosuppression, we primed post-septic and control animals by immunization with the model antigen TNP-ovalbumin in alum, and measured the specific antibody-responses via ELISA and ELISpot assay as well as T-cell responses in a proliferation assay after restimulation. Specific antibody titers, antibody affinity and plasma cell counts in the bone marrow were reduced in post-septic animals. The antigen-induced splenic proliferation was also impaired. The adaptive immunosuppression was positively correlated with an overwhelming general antibody response to the septic insult. Remarkably, antigen “overload” by non-specific immunization induced a similar degree of adaptive immunosuppression in the absence of sepsis. In both settings, depletion of regulatory T cells before priming reversed some parameters of the immunosuppression. In conclusion, our data show that adaptive immunosuppression occurs independent of profound systemic inflammation and life-threatening illness.
Collapse
Affiliation(s)
- Katrin Schmoeckel
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Daniel M. Mrochen
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Jochen Hühn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Christian Pötschke
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Barbara M. Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
- * E-mail:
| |
Collapse
|
42
|
Zhang Z, Liu X, Wang H, Qu Z, Crew R, Zhang N, Jiang Y. Increased soluble ST2 and IL‑4 serum levels are associated with disease severity in patients with membranous nephropathy. Mol Med Rep 2017; 17:2778-2786. [PMID: 29207152 DOI: 10.3892/mmr.2017.8130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 11/13/2017] [Indexed: 11/05/2022] Open
Abstract
The interleukin (IL)‑33/suppression of tumorigenicity 2 (ST2) axis regulates Th2 reactivity, and ST2 is the receptor for IL‑33. In this study, the roles of IL‑33 and soluble ST2 (sST2) in the pathogenesis of membranous nephropathy (MN), and their association with disease severity were evaluated. Serum levels of IL‑33 and sST2 in 93 patients, and 34 healthy controls (HCs) were measured by enzyme‑linked immunosorbent assays. Clinical characteristics were recorded and the estimated glomerular filtration rates (eGFRs) were computed. In addition, the association between serum IL‑33 and sST2 levels, and clinical measurements in patients with MN was analyzed. No difference in the serum levels of IL‑33 was identified between the patients with MN and HCs. However, the serum levels of sST2 were considerably higher in the MN patients compared with in the HCs at every stage. Higher concentrations of serum IL‑2, IL‑4, IL‑10, IL‑17A, and IFN‑γ were measured in the MN patients compared with in the HCs. Serum sST2 concentrations were negatively correlated with IL‑4 concentrations in the patients with MN. Furthermore, serum sST2 levels were negatively correlated with the eGFRs and serum calcium levels. Serum sST2 levels, but not IL‑33 levels, were positively correlated with the 24‑h urine protein and serum phosphorus levels. Following treatment, serum sST2 levels were considerably reduced, whereas serum IL‑4 and IL‑10 levels were significantly increased. These data suggest that sST2 and IL‑4, but not IL‑33, contribute to the pathogenesis of MN.
Collapse
Affiliation(s)
- Zhihui Zhang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaolei Liu
- Key Laboratory of Zoonoses Research, Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haifeng Wang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhihui Qu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Rebecca Crew
- University of Oklahoma Health Sciences Center, Oklahoma, OK 73104, USA
| | - Nan Zhang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
43
|
Vk C, Ty L, Wf L, Ywy WS, An S, S Z, A M. Leptospirosis in human: Biomarkers in host immune responses. Microbiol Res 2017; 207:108-115. [PMID: 29458845 DOI: 10.1016/j.micres.2017.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/23/2017] [Accepted: 11/25/2017] [Indexed: 02/07/2023]
Abstract
Leptospirosis remains one of the most widespread zoonotic diseases caused by spirochetes of the genus Leptospira, which accounts for high morbidity and mortality globally. Leptospiral infections are often found in tropical and subtropical regions, with people exposed to contaminated environments or animal reservoirs are at high risk of getting the infection. Leptospirosis has a wide range of clinical manifestations with non-specific signs and symptoms and often misdiagnosed with other acute febrile illnesses at early stage of infection. Despite being one of the leading causes of zoonotic morbidity worldwide, there is still a gap between pathogenesis and human immune responses during leptospiral infection. It still remains obscure whether the severity of the infection is caused by the pathogenic properties of the Leptospira itself, or it is a consequence of imbalance host immune factors. Hence, in this review, we seek to summarize the past and present milestone findings on the biomarkers of host immune response aspects during human leptospiral infection, including cytokine and other immune mediators. A profound understanding of the interlink between virulence factors and host immune responses during human leptospirosis is imperative to identify potential biomarkers for diagnostic and prognostic applications as well as designing novel immunotherapeutic strategies in future.
Collapse
Affiliation(s)
- Chin Vk
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia.
| | - Lee Ty
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; School of Foundation Studies, Perdana University, 43400, Serdang, Malaysia.
| | - Lim Wf
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia; Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| | - Wan Shahriman Ywy
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia; Department of Medical Laboratory Technology, Faculty of Health Sciences, Universiti Teknologi MARA, Cawangan Selangor Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| | - Syafinaz An
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia.
| | - Zamberi S
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM, Serdang, Selangor, Malaysia.
| | - Maha A
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
44
|
Lee YT, Gong M, Chau A, Wong WT, Bazoukis G, Wong SH, Lampropoulos K, Xia Y, Li G, Wong MCS, Liu T, Wu WKK, Tse G. Pentraxin-3 as a marker of sepsis severity and predictor of mortality outcomes: A systematic review and meta-analysis. J Infect 2017; 76:1-10. [PMID: 29174966 DOI: 10.1016/j.jinf.2017.10.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 10/28/2017] [Accepted: 10/30/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Pentraxin-3 (PTX-3) is a multi-functional pattern recognition molecule produced by various cell types of peripheral tissues in different infections. It is raised in sepsis, but its values in predicting disease severity or mortality outcomes have been controversial. Therefore, we conducted a systematic review and meta-analysis of these associations. METHODS PubMed and Embase were searched until July 18, 2017 for studies that evaluated the relationship between PTX-3 levels and disease severity or mortality in sepsis. RESULTS A total of 23 and 10 entries were retrieved from both databases, respectively, of which 16 studies were included in the final meta-analysis. A total of 3001 patients (56% male, mean age 63 ± 15 years; mean follow-up duration of 207 days) were analysed. PTX-3 was significantly higher in patients with more severe sepsis compared to those with less severe sepsis (standard mean difference = 18.5 ng/mL, standard error: 4.5 ng/mL, P < 0.0001) and higher in non-survivors compared to survivors (standard mean difference = 40.3 ng/mL, standard error: 6.8 ng/mL, P < 0.0001). Elevated PTX-3 levels significantly increased the risk of all-cause mortality (hazard ratio: 1.91, 95% CI: 1.53 to 2.46, P < 0.0001). CONCLUSIONS PTX-3 significantly predicts disease severity and mortality in sepsis.
Collapse
Affiliation(s)
- Yee Ting Lee
- Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, SAR, China
| | - Mengqi Gong
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Alex Chau
- University of British Columbia, Canada
| | - Wing Tak Wong
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China
| | - George Bazoukis
- Second Department of Cardiology, Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Athens, Greece
| | - Sunny Hei Wong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Konstantinos Lampropoulos
- Second Department of Cardiology, Laboratory of Cardiac Electrophysiology, Evangelismos General Hospital of Athens, Athens, Greece
| | - Yunlong Xia
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Martin C S Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - William K K Wu
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; Department of Anaesthesia and Intensive Care, State Key Laboratory of Digestive Disease, LKS Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China.
| | - Gary Tse
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China.
| | | |
Collapse
|
45
|
Predictive value of low interleukin-33 in critically ill patients. Cytokine 2017; 103:109-113. [PMID: 28974430 DOI: 10.1016/j.cyto.2017.09.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 02/08/2023]
Abstract
Patients admitted to a medical intensive care unit (ICU) are characterized by an activated immune system and exhibit a high mortality rate irrespective of the underlying cause of admission. Interleukin (IL)-33 has been shown to be protective in experimental sepsis models and it has been demonstrated that circulating levels of its "decoy" receptor soluble ST2 (sST2) are associated with outcome in critically ill patients. The aim of the present study was to investigate whether circulating IL-33 is associated with 30-day mortality in patients admitted to a medical ICU. In this prospective, observational study, both IL-33 and sST2 levels were assessed in 223 consecutive patients at ICU admission using specific enzyme-linked immunosorbent assays (ELISAs). During the 30-day follow-up, 58 patients (26%) died. Circulating IL-33 was detectable in 166 patients and in 57 patients, serum IL-33 was below the detection limit. Both detectable IL-33 and sST2 below the median were strong predictors of survival in critically ill patients independent of acute physiology and chronic health evaluation II (APACHE II) score. IL-33 and sST2 predicted risk independent from each other. Patients with both, non-detectable levels of IL-33 and sST2 levels above the median, showed a dramatically increased mortality risk (HR 6.9 95% CI 3.0-16.2; p<0.001). Low levels of IL-33 and increased levels of sST2 predict mortality risk in critically ill patients independent from each other and APACHE II score. Both together showed additive predictive value suggesting a pathogenic role of the IL-33/ST2 system in critically ill patients.
Collapse
|
46
|
Association of interleukin 1 receptor-like 1 gene polymorphisms with eosinophilic phenotype in Japanese adults with asthma. Respir Investig 2017; 55:338-347. [PMID: 29153414 DOI: 10.1016/j.resinv.2017.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 07/27/2017] [Accepted: 08/08/2017] [Indexed: 01/26/2023]
Abstract
BACKGROUND IL1RL1 (ST2) is involved in Th2 inflammation including eosinophil activation. Single nucleotide polymorphisms (SNPs) of the IL1RL1 gene are associated with asthma development and increased peripheral blood eosinophil counts. However, the association between IL1RL1 SNPs and eosinophilic phenotype among adults with asthma remains unexplored. METHODS In a primary cohort of 110 adult Japanese patients with stable asthma, we examined the associations between IL1RL1 SNPs and clinical measurements including forced expiratory volume (FEV1), airway reversibility of FEV1, exhaled nitric oxide (FeNO), serum soluble-ST2 (sST2) levels, peripheral blood eosinophil differentials and serum total IgE level. The findings in the primary cohort were confirmed in a validation cohort of 126 adult Japanese patients with stable asthma. RESULTS Patients with minor alleles in 3 SNPs (rs17026974, rs1420101, and rs1921622) had high FeNO, blood eosinophil differentials, and reversibility of FEV1, but low levels of serum sST2 and FEV1. Minor alleles of rs1041973 were associated with low serum sST2 levels alone. In the validation cohort, minor alleles of rs1420101 were associated with high FeNO and blood eosinophil differentials, whereas minor alleles of rs17026974 and rs1921622 were associated with high blood eosinophil differentials and FeNO, respectively. Multivariate analyses revealed that the minor allele of rs1420101 additively contributed to the FeNO, blood eosinophil differentials, and reversibility of FEV1. CONCLUSIONS The minor alleles of IL1RL1 SNPs were associated with high FeNO and peripheral blood eosinophilia among adult Japanese patients with stable asthma. IL1RL1 SNPs may characterize the eosinophilic phenotype with greater eosinophilic inflammation in the Japanese asthma cohort.
Collapse
|
47
|
Emerging Roles of IL-33/ST2 Axis in Renal Diseases. Int J Mol Sci 2017; 18:ijms18040783. [PMID: 28387719 PMCID: PMC5412367 DOI: 10.3390/ijms18040783] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Renal diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), have a great impact on health care systems worldwide. Similar to cardiovascular diseases, renal diseases are inflammatory diseases involving a variety of cytokines. Primary causes of renal injury include ischemia, uremic toxins, bacteremia, or nephrotoxicity. Inflammation represents an important component following kidney injury. Interleukin (IL)-33 is a member of the IL-1 cytokine family, which is widely expressed in epithelial barrier tissues and endothelial cells, and mediates both tissue inflammation and repair responses. IL-33 is released as a nuclear alarmin in response to tissue damage and triggers innate and adaptive immune responses by binding to its receptor, suppression of tumorigenicity 2 (ST2). Recent evidence from clinical and experimental animal studies indicates that the IL-33/ST2 axis is involved in the pathogenesis of CKD, renal graft injury, systemic lupus nephritis, and AKI. In this review, we discuss the pathological and tissue reparative roles of the IL-33/ST2 pathway in different types of renal diseases.
Collapse
|
48
|
Meta-Analysis of Soluble Suppression of Tumorigenicity-2 and Prognosis in Acute Heart Failure. JACC-HEART FAILURE 2017; 5:287-296. [DOI: 10.1016/j.jchf.2016.12.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/31/2016] [Accepted: 12/29/2016] [Indexed: 11/23/2022]
|
49
|
Romero R, Chaemsaithong P, Tarca AL, Korzeniewski SJ, Maymon E, Pacora P, Panaitescu B, Chaiyasit N, Dong Z, Erez O, Hassan SS, Chaiworapongsa T. Maternal plasma-soluble ST2 concentrations are elevated prior to the development of early and late onset preeclampsia - a longitudinal study. J Matern Fetal Neonatal Med 2017; 31:418-432. [PMID: 28114842 DOI: 10.1080/14767058.2017.1286319] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The objectives of this study were to determine (1) the longitudinal profile of plasma soluble ST2 (sST2) concentrations in patients with preeclampsia and those with uncomplicated pregnancies; (2) whether the changes in sST2 occur prior to the diagnosis of preeclampsia; and (3) the longitudinal sST2 profile of women with early or late preeclampsia. MATERIALS AND METHODS This longitudinal nested case-control study included singleton pregnancies in the following groups: (1) uncomplicated pregnancies (n = 160); and (2) those complicated by early (<34 weeks, n = 9) and late (≥34 weeks, n = 31) preeclampsia. sST2 concentrations were determined by enzyme-linked immunosorbent assays. Mixed-effects models were used for the longitudinal analysis. RESULTS (1) Plasma sST2 concentration profiles across gestation differed significantly among cases and controls (p < 0.0001); (2) women with early preeclampsia had higher mean sST2 concentrations than controls at >22 weeks of gestation; cases with late preeclampsia had higher mean concentrations at >33 weeks of gestation (both p < 0.05); and (3) these changes started approximately 6 weeks prior to clinical diagnosis. CONCLUSIONS Maternal plasma sST2 concentrations are elevated 6 weeks prior to the clinical diagnosis of preeclampsia. An increase in the maternal plasma concentration of sST2 may contribute to an exaggerated intravascular inflammatory response and/or the Th1/Th2 imbalance in some cases.
Collapse
Affiliation(s)
- Roberto Romero
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,b Department of Obstetrics and Gynecology , University of Michigan , Ann Arbor , MI , USA.,c Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,d Center for Molecular Medicine and Genetics , Wayne State University , Detroit , MI , USA
| | - Piya Chaemsaithong
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Adi L Tarca
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Steven J Korzeniewski
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,c Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Eli Maymon
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Percy Pacora
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Bogdan Panaitescu
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Noppadol Chaiyasit
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Zhong Dong
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Offer Erez
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Sonia S Hassan
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Tinnakorn Chaiworapongsa
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|
50
|
Yang HS, Hur M, Kim H, Magrini L, Marino R, Di Somma S. Soluble Suppression of Tumorigenicity 2 and Echocardiography in Sepsis. Ann Lab Med 2017; 36:590-4. [PMID: 27578513 PMCID: PMC5011113 DOI: 10.3343/alm.2016.36.6.590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 05/22/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022] Open
Abstract
Soluble suppression of tumorigenicity 2 (sST2) has emerged as a biomarker of cardiac stretch or remodeling, and has demonstrated a role in acutely decompensated heart failure. However, its role in sepsis-induced cardiac dysfunction is still unknown. We explored whether sST2 serum concentration reflects either systolic or diastolic dysfunction as measured by Doppler echocardiography. In a total of 127 patients with sepsis, correlations between sST2 and blood pressure, left ventricular (LV) ejection fraction, LV diastolic filling (ratio of early transmitral flow velocity to early diastolic mitral annulus velocity), and resting pulmonary arterial pressure were evaluated. Correlations between sST2 and other sepsis biomarkers (high-sensitivity C-reactive protein [hs-CRP] and procalcitonin) were also examined. sST2 showed a moderate correlation with mean arterial pressure (r=-0.3499) but no correlation with LV ejection fraction, diastolic filling, or resting pulmonary hypertension. It showed moderate correlations with hs-CRP and procalcitonin (r=0.2608 and r=0.3829, respectively). sST2 might have a role as a biomarker of shock or inflammation, but it cannot reflect echocardiographic findings of LV ejection fraction or diastolic filling in sepsis.
Collapse
Affiliation(s)
- Hyun Suk Yang
- Department of Cardiovascular Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Mina Hur
- Department of Cardiovascular Medicine, Konkuk University School of Medicine, Seoul, Korea.
| | - Hanah Kim
- Department of Laboratory Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Laura Magrini
- Department of Medical-Surgery Sciences and Translational Medicine, University La Sapienza Rome, Sant'Andrea Hospital, Rome, Italy
| | - Rossella Marino
- Department of Medical-Surgery Sciences and Translational Medicine, University La Sapienza Rome, Sant'Andrea Hospital, Rome, Italy
| | - Salvatore Di Somma
- Department of Medical-Surgery Sciences and Translational Medicine, University La Sapienza Rome, Sant'Andrea Hospital, Rome, Italy.
| | | |
Collapse
|