1
|
Tan ESJ, Choi H, DeFilippi CR, Oon YY, Chan SP, Gong L, Lunaria JB, Liew OW, Chong JPC, Tay ELW, Soo WM, Yip JWL, Yong QW, Lee EM, Daniel Yeo PS, Ding ZP, Tang HC, Ewe SH, Chin CWL, Chai SC, Goh PP, Ling LF, Ong HY, Richards AM, Ling LH. Circulating Plasma Proteins in Aortic Stenosis: Associations With Severity, Myocardial Response, and Clinical Outcomes. J Am Heart Assoc 2024; 13:e035486. [PMID: 39344657 DOI: 10.1161/jaha.124.035486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Echocardiographic indexes of aortic stenosis may not comprehensively reflect disease morbidity. Plasma proteomic profiling may add prognostic value in these patients. METHODS AND RESULTS Proximity extension assays (Olink) of 183 circulating cardiovascular and inflammatory proteins were performed in a prospective follow-up study of 122 asymptomatic/minimally symptomatic patients (mean±SD age, 69.1±10.9 years; 61% men) with moderate to severe aortic stenosis and preserved left ventricular ejection fraction. Protein signatures of higher-risk echocardiographic subgroups were determined. Associations of proteins with the primary composite outcome (heart failure hospitalization, progression to New York Heart Association class III-IV, or all-cause mortality) were evaluated using competing risk analyses, with aortic valve replacement being the competing risk. Network analysis unveiled mutually exclusive communities of proteins and echocardiographic parameters, connected only through NT-proBNP (N-terminal pro-B-type natriuretic peptide). Members of the tumor necrosis factor receptor superfamily (TNFRSF1A, TNFRSF1B, and TNFRSF14), and trefoil factor-3 were major hub proteins among the circulating biomarkers. Left ventricular global longitudinal strain >-15% was associated with higher levels of proteins, primarily of inflammation and immune regulation, whereas aortic valve area <1 cm2, E/e' >15, and left atrial reservoir strain <20% were associated with higher levels of NT-proBNP. Of 14 proteins associated with the primary end point, phospholipase-C, C-X-C motif chemokine-9, and interleukin-10 receptor subunit β demonstrated the highest hazard ratios after adjusting for clinical factors (q<0.05). CONCLUSIONS Plasma proteins involved in inflammation and immune regulation were differentially expressed in patients with aortic stenosis with reduced left ventricular global longitudinal strain, and associated with adverse clinical outcomes. Their incorporation into aortic stenosis risk stratification warrants further assessment.
Collapse
Affiliation(s)
- Eugene S J Tan
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Hyungwon Choi
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| | | | - Yen-Yee Oon
- Sarawak Heart Centre Kota Samarahan Sarawak Malaysia
| | - Siew-Pang Chan
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Lingli Gong
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Josephine B Lunaria
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Oi-Wah Liew
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| | - Jenny Pek-Ching Chong
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| | - Edgar Lik-Wui Tay
- National University Heart Centre Singapore Singapore
- Asian Heart and Vascular Centre Singapore Singapore
| | - Wern-Miin Soo
- National University Heart Centre Singapore Singapore
| | - James Wei-Luen Yip
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | | | | | - Poh Shuan Daniel Yeo
- Tan Tock Seng Hospital Singapore Singapore
- Apex Heart Clinic Gleneagles Hospital Singapore Singapore
| | | | | | | | | | | | | | | | | | - A Mark Richards
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
- Christchurch Heart Institute, University of Otago Christchurch New Zealand
| | - Lieng-Hsi Ling
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| |
Collapse
|
2
|
Eligini S, Savini C, Ghilardi S, Mallia A, Vieceli Dalla Sega F, Fortini F, Mikus E, Munno M, Modafferi G, Agostoni P, Tremoli E, Banfi C. Immature Surfactant Protein B Increases in the Serum of Patients with Calcific Severe Aortic Stenosis. Int J Mol Sci 2024; 25:6418. [PMID: 38928127 PMCID: PMC11204170 DOI: 10.3390/ijms25126418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Valvular disease is a complex pathological condition that impacts countless individuals around the globe. Due to limited treatments, it is crucial to understand its mechanisms to identify new targets. Valve disease may result in pulmonary venous hypertension, which is linked to compromised functioning of the alveolar and capillary membranes and hindered gas exchange. Nonetheless, the correlation between surfactant proteins (SPs) and valve disease remains unexplored. A total of 44 patients were enrolled in this study, with 36 undergoing aortic valve replacement and 8 needing a second aortic valve substitution due to bioprosthetic valve degeneration. Ten healthy subjects were also included. The results showed that patients who underwent both the first valve replacement and the second surgery had significantly higher levels of immature SP-B (proSP-B) compared to control subjects. The levels of the extra-lung collectin SP-D were higher in patients who needed a second surgery due to bioprosthetic valve degeneration, while SP-A levels remained unchanged. The research also showed that there was no reciprocal relationship between inflammation and SP-D as the levels of inflammatory mediators did not differ between groups. The present study demonstrates that circulating proSP-B serves as a reliable marker of alveolar-capillary membrane damage in patients with valvular heart disease.
Collapse
Affiliation(s)
- Sonia Eligini
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.E.); (S.G.); (A.M.); (M.M.); (G.M.)
| | - Carlo Savini
- Maria Cecilia Hospital, GVM Care and Research, 48033 Cotignola, Italy; (C.S.); (F.V.D.S.); (F.F.); (E.M.); (E.T.)
- Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum, Università di Bologna, 40126 Bologna, Italy
| | - Stefania Ghilardi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.E.); (S.G.); (A.M.); (M.M.); (G.M.)
| | - Alice Mallia
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.E.); (S.G.); (A.M.); (M.M.); (G.M.)
- Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Università di Pavia, 27100 Pavia, Italy
| | - Francesco Vieceli Dalla Sega
- Maria Cecilia Hospital, GVM Care and Research, 48033 Cotignola, Italy; (C.S.); (F.V.D.S.); (F.F.); (E.M.); (E.T.)
| | - Francesca Fortini
- Maria Cecilia Hospital, GVM Care and Research, 48033 Cotignola, Italy; (C.S.); (F.V.D.S.); (F.F.); (E.M.); (E.T.)
| | - Elisa Mikus
- Maria Cecilia Hospital, GVM Care and Research, 48033 Cotignola, Italy; (C.S.); (F.V.D.S.); (F.F.); (E.M.); (E.T.)
| | - Marco Munno
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.E.); (S.G.); (A.M.); (M.M.); (G.M.)
| | - Gloria Modafferi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.E.); (S.G.); (A.M.); (M.M.); (G.M.)
| | - Piergiuseppe Agostoni
- Heart Failure Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy;
- Department of Clinical and Community Sciences, University of Milan, 20122 Milan, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care and Research, 48033 Cotignola, Italy; (C.S.); (F.V.D.S.); (F.F.); (E.M.); (E.T.)
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.E.); (S.G.); (A.M.); (M.M.); (G.M.)
| |
Collapse
|
3
|
Zhang T, Han X, Zhang H, Li X, Zhou X, Feng S, Guo C, Song F, Tao T, Yin C, Xia J. Identification of molecular markers for predicting the severity of heart failure after AMI: An Olink precision proteomic study. Clin Chim Acta 2024; 555:117825. [PMID: 38331209 DOI: 10.1016/j.cca.2024.117825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/06/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Acute myocardial infarction (AMI) still has a high incidence of varying degrees of heart failure (HF). The aim of this study is to identify new molecular markers for predicting the severity of HF after AMI. METHODS We analyzed demographic indicators, past medical history, clinical indicators, major adverse cardiac events (MACEs) and molecular markers in patients with different Killip classifications after AMI. Olink proteomics was used to explore new molecular markers for predicting different severity of HF after AMI. RESULTS Neutrophil count was the independent risk factors for in-hospital MACEs. Nineteen differentially expressed proteins (DEPs) increased significantly with increasing Killip classification. Five DEPs were also found to have an AUC (95 % CI) value greater than 0.8: GDF-15, NT-pro BNP, TNF-R2, TNF-R1 and TFF3. CONCLUSIONS Neutrophil count, GDF-15, TNF-R2, TNF-R1 and TFF3 were closely related to the Killip classification of HF after AMI, which suggests that the inflammatory response plays an important role in the severity of HF after AMI and that regulating inflammation might become a new target for controlling HF.
Collapse
Affiliation(s)
- Tianxing Zhang
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xuexue Han
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Hao Zhang
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xue Li
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xingzhu Zhou
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shuhui Feng
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Chenglong Guo
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Fei Song
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Tianqi Tao
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Chunlin Yin
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jinggang Xia
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
4
|
de Bakker M, Loncq de Jong M, Petersen T, de Lange I, Akkerhuis KM, Umans VA, Rizopoulos D, Boersma E, Brugts JJ, Kardys I. Sex-specific cardiovascular protein levels and their link with clinical outcome in heart failure. ESC Heart Fail 2024; 11:594-600. [PMID: 38009274 PMCID: PMC10804167 DOI: 10.1002/ehf2.14578] [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: 06/19/2023] [Revised: 10/02/2023] [Accepted: 10/29/2023] [Indexed: 11/28/2023] Open
Abstract
AIMS This study aims to provide insight into sex-specific cardiovascular protein profiles and their associations with adverse outcomes, which may contribute to a better understanding of heart failure (HF) pathophysiology and the optimal use of circulating proteins for prognostication in women and men. METHODS AND RESULTS In 250 stable patients with HF with reduced ejection fraction (HFrEF), we performed trimonthly blood sampling (median follow-up: 26 [17-30] months). We selected all baseline samples and two samples closest to the primary endpoint (PEP; composite of cardiovascular death, heart transplantation, left ventricular assist device implantation, and HF hospitalization) or one sample closest to censoring and applied the Olink Cardiovascular III panel. We used linear regression to study sex-based differences in baseline levels and joint models to study differences in the prognostic value of serially measured proteins. In 66 women and 184 men (mean age of 66 and 67 years, respectively), 21% and 28% reached the PEP, respectively. Mean baseline levels of fatty acid-binding protein 4, secretoglobin family 3A member 2, paraoxonase 3, and trefoil factor 3 were higher in women (Pinteraction : 0.001, 0.007, 0.018, and 0.049, respectively), while matrix metalloproteinase-3, interleukin 1 receptor-like 1, and myoglobin were higher in men (Pinteraction : <0.001, 0.001, and 0.049, respectively), independent of clinical characteristics. No significant differences between sexes were observed in the longitudinal associations of proteins with the PEP. Only peptidoglycan recognition protein 1 showed a suggestive interaction with sex for the primary outcome (Pinteraction = 0.028), without multiple testing correction, and was more strongly associated with adverse outcome in women {hazard ratio [HR] 3.03 [95% confidence interval (CI), 1.42 to 6.68], P = 0.008} compared with men [HR 1.18 (95% CI, 0.84 to 1.66), P = 0.347]. CONCLUSIONS Although multiple cardiovascular-related proteins show sex differences at baseline, temporal associations with the adverse outcome do not differ between women and men with HFrEF.
Collapse
Affiliation(s)
- Marie de Bakker
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
| | - Mylène Loncq de Jong
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
| | - Teun Petersen
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
- Department of BiostatisticsErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Iris de Lange
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
| | - K. Martijn Akkerhuis
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
| | - Victor A. Umans
- Department of CardiologyNorthwest ClinicsAlkmaarThe Netherlands
| | - Dimitris Rizopoulos
- Department of BiostatisticsErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
- Department of EpidemiologyErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Eric Boersma
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
| | - Jasper J. Brugts
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
| | - Isabella Kardys
- Department of CardiologyErasmus MC Cardiovascular Institute, University Medical Center RotterdamRoom Na‐316, P.O. Box 20403000 CARotterdamThe Netherlands
| |
Collapse
|
5
|
Wang L, Niu X. Immunoregulatory Roles of Osteopontin in Diseases. Nutrients 2024; 16:312. [PMID: 38276550 PMCID: PMC10819284 DOI: 10.3390/nu16020312] [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: 11/17/2023] [Revised: 01/07/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Osteopontin (OPN) is a multifunctional protein that plays a pivotal role in the immune system. It is involved in various biological processes, including cell adhesion, migration and survival. The study of the immunomodulatory effects of OPN is of paramount importance due to its potential therapeutic applications. A comprehensive understanding of how OPN regulates the immune response could pave the way for the development of novel treatments for a multitude of diseases, including autoimmune disorders, infectious diseases and cancer. Therefore, in the following paper, we provide a systematic overview of OPN and its immunoregulatory roles in various diseases, laying the foundation for the development of OPN-based therapies in the future.
Collapse
Affiliation(s)
- Lebei Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoyin Niu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| |
Collapse
|
6
|
Abstract
Pulmonary surfactant is a critical component of lung function in healthy individuals. It functions in part by lowering surface tension in the alveoli, thereby allowing for breathing with minimal effort. The prevailing thinking is that low surface tension is attained by a compression-driven squeeze-out of unsaturated phospholipids during exhalation, forming a film enriched in saturated phospholipids that achieves surface tensions close to zero. A thorough review of past and recent literature suggests that the compression-driven squeeze-out mechanism may be erroneous. Here, we posit that a surfactant film enriched in saturated lipids is formed shortly after birth by an adsorption-driven sorting process and that its composition does not change during normal breathing. We provide biophysical evidence for the rapid formation of an enriched film at high surfactant concentrations, facilitated by adsorption structures containing hydrophobic surfactant proteins. We examine biophysical evidence for and against the compression-driven squeeze-out mechanism and propose a new model for surfactant function. The proposed model is tested against existing physiological and pathophysiological evidence in neonatal and adult lungs, leading to ideas for biophysical research, that should be addressed to establish the physiological relevance of this new perspective on the function of the mighty thin film that surfactant provides.
Collapse
Affiliation(s)
- Fred Possmayer
- Department of Biochemistry, Western University, London, Ontario N6A 3K7, Canada
- Department of Obstetrics/Gynaecology, Western University, London, Ontario N6A 3K7, Canada
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manon, Honolulu, Hawaii 96822, United States
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96826, United States
| | - Ruud A W Veldhuizen
- Department of Physiology & Pharmacology, Western University, London, Ontario N6A 5C1, Canada
- Department of Medicine, Western University, London, Ontario N6A 3K7, Canada
- Lawson Health Research Institute, London, Ontario N6A 4V2, Canada
| | - Nils O Petersen
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada
| |
Collapse
|
7
|
Siegbahn A, Eriksson N, Assarsson E, Lundberg M, Ballagi A, Held C, Stewart RAH, White HD, Åberg M, Wallentin L. Development and validation of a quantitative Proximity Extension Assay instrument with 21 proteins associated with cardiovascular risk (CVD-21). PLoS One 2023; 18:e0293465. [PMID: 37963145 PMCID: PMC10645335 DOI: 10.1371/journal.pone.0293465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Treatment of cardiovascular diseases (CVD) is a substantial burden to healthcare systems worldwide. New tools are needed to improve precision of treatment by optimizing the balance between efficacy, safety, and cost. We developed a high-throughput multi-marker decision support instrument which simultaneously quantifies proteins associated with CVD. METHODS AND FINDINGS Candidate proteins independently associated with different clinical outcomes were selected from clinical studies by the screening of 368 circulating biomarkers. We then custom-designed a quantitative PEA-panel with 21 proteins (CVD-21) by including recombinant antigens as calibrator samples for normalization and absolute quantification of the proteins. The utility of the CVD-21 tool was evaluated in plasma samples from a case-control cohort of 4224 patients with chronic coronary syndrome (CCS) using multivariable Cox regression analyses and machine learning techniques. The assays in the CVD-21 tool gave good precision and high sensitivity with lower level of determination (LOD) between 0.03-0.7 pg/ml for five of the biomarkers. The dynamic range for the assays was sufficient to accurately quantify the biomarkers in the validation study except for troponin I, which in the modeling was replaced by high-sensitive cardiac troponin T (hs-TnT). We created seven different multimarker models, including a reference model with NT-proBNP, hs-TnT, GDF-15, IL-6, and cystatin C and one model with only clinical variables, for the comparison of the discriminative value of the CVD-21 tool. All models with biomarkers including hs-TnT provided similar discrimination for all outcomes, e.g. c-index between 0.68-0.86 and outperformed models using only clinical variables. Most important prognostic biomarkers were MMP-12, U-PAR, REN, VEGF-D, FGF-23, TFF3, ADM, and SCF. CONCLUSIONS The CVD-21 tool is the very first instrument which with PEA simultaneously quantifies 21 proteins with associations to different CVD. Novel pathophysiologic and prognostic information beyond that of established biomarkers were identified by a number of proteins.
Collapse
Affiliation(s)
- Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | | | | | | | - Claes Held
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Ralph A. H. Stewart
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Harvey D. White
- Green Lane Cardiovascular Service, Te Whatu Ora Health New Zealand, Te Toka Tumai Auckland and University of Auckland, Auckland, New Zealand
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
Eidizadeh A, Schnelle M, Leha A, Edelmann F, Nolte K, Werhahn SM, Binder L, Wachter R. Biomarker profiles in heart failure with preserved vs. reduced ejection fraction: results from the DIAST-CHF study. ESC Heart Fail 2022; 10:200-210. [PMID: 36184749 PMCID: PMC9871664 DOI: 10.1002/ehf2.14167] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/29/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
AIMS Chronic heart failure (HF) is a common disease and one of the leading causes of death worldwide. Heart failure with preserved ejection fraction (HFpEF) and with reduced ejection fraction (HFrEF) are different diseases with distinct as well as comparable pathophysiologies and diverse responses to therapeutic agents. We aimed to identify possible pathobiochemical signalling pathways and biomarkers in HFpEF and HFrEF by using a broad proteomic approach. METHODS AND RESULTS A total of 180 biomarkers in the plasma of a representative subgroup (71 years old) of HFpEF (70% female) with a left ventricular ejection fraction (LVEF) ≥ 50% and HFrEF (18% female) with an LVEF ≤ 40% patients (n = 127) from the Prevalence and Clinical Course of Diastolic Dysfunction and Diastolic Heart Failure (DIAST-CHF) trial were examined and compared with a healthy control group (n = 40; 48% female). We were able to identify 35 proteins that were expressed significantly different in both HF groups compared with the control group. We determine 29 unique proteins expressed in HFpEF and 33 unique proteins in HFrEF. Significantly up-regulated trefoil factor 3 (TFF3) and down-regulated contactin-1 could be identified as previously unknown biomarkers for HF. However, TFF3 is also a predictive factor for the occurrence of a cardiovascular event in HFpEF patients. In HFpEF, serine protease 27 was found at reduced levels for the first time, which could offer a new therapeutic target. Additionally, network analyses showed a special role of platelet-derived growth factor subunit A, Dickkopf-related protein 1, and tumour necrosis factor receptor superfamily member 6 in HFpEF patients, whereas perlecan and junctional adhesion molecule A stood out in the HFrEF group. Overall, signalling pathways of metabolic processes, cellular stress, and iron metabolism seemed to be important for HFrEF, whereas for HFpEF, oxygen stress, haemostasis, cell renewal, cell migration, and cell proliferation are in the foreground. CONCLUSIONS The identified proteins and signalling pathways offer new therapeutic and diagnostic approaches for patients with chronic HF.
Collapse
Affiliation(s)
- Abass Eidizadeh
- Institute for Clinical Chemistry/Interdisciplinary UMG LaboratoryUniversity Medical Center GöttingenGöttingenGermany
| | - Moritz Schnelle
- Institute for Clinical Chemistry/Interdisciplinary UMG LaboratoryUniversity Medical Center GöttingenGöttingenGermany,DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Andreas Leha
- DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany,Department of Medical StatisticsUniversity Medical Center GöttingenGöttingenGermany
| | - Frank Edelmann
- Department of Internal Medicine and CardiologyCharité‐Universitätsmedizin Berlin, Campus Virchow KlinikumBerlinGermany,DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany,Berlin Institute of HealthBerlinGermany
| | - Kathleen Nolte
- Clinic of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | | | - Lutz Binder
- Institute for Clinical Chemistry/Interdisciplinary UMG LaboratoryUniversity Medical Center GöttingenGöttingenGermany,DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany
| | - Rolf Wachter
- DZHK (German Centre for Cardiovascular Research), Partner Site GöttingenGöttingenGermany,Clinic of Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany,Clinic and Policlinic for CardiologyUniversity Hospital LeipzigLeipzigGermany
| |
Collapse
|
9
|
Berezin AE. Cell-free long noncoding RNAs as predictive biomarkers for cardiovascular diseases. Int J Cardiol 2022; 359:115-117. [PMID: 35439586 DOI: 10.1016/j.ijcard.2022.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022]
|
10
|
Israr MZ, Zhan H, Salzano A, Voors AA, Cleland JG, Anker SD, Metra M, van Veldhuisen DJ, Lang CC, Zannad F, Samani NJ, Ng LL, Suzuki T. Surrogate markers of gut dysfunction are related to heart failure severity and outcome-from the BIOSTAT-CHF consortium. Am Heart J 2022; 248:108-119. [PMID: 35278373 DOI: 10.1016/j.ahj.2022.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND The contribution of gut dysfunction to heart failure (HF) pathophysiology is not routinely assessed. We sought to investigate whether biomarkers of gut dysfunction would be useful in assessment of HF (eg, severity, adverse outcomes) and risk stratification. METHODS A panel of gut-related biomarkers including metabolites of the choline/carnitine- pathway (acetyl-L-carnitine, betaine, choline, γ-butyrobetaine, L-carnitine and trimethylamine-N-oxide [TMAO]) and the gut peptide, Trefoil factor-3 (TFF-3), were investigated in 1,783 patients with worsening HF enrolled in the systems BIOlogy Study to TAilored Treatment in Chronic Heart Failure (BIOSTAT-CHF) cohort and associations with HF severity and outcomes, and use in risk stratification were assessed. RESULTS Metabolites of the carnitine-TMAO pathway (acetyl-L-carnitine, γ-butyrobetaine, L-carnitine, and TMAO) and TFF-3 were associated with the composite outcome of HF hospitalization or all-cause mortality at 3 years (hazards ratio [HR] 2.04-2.93 [95% confidence interval {CI} 1.30-4.71] P≤ .002). Combining the carnitine-TMAO metabolites with TFF-3, as a gut dysfunction panel, showed a graded association; a greater number of elevated markers was associated with higher New York Heart Association class (P< .001), higher plasma concentrations of B-type natriuretic peptide (P< .001), and worse outcome (HR 1.90-4.58 [95% CI 1.19-6.74] P≤ 0.008). Addition of gut dysfunction biomarkers to the contemporary BIOSTAT HF risk model also improved prediction for the aforementioned composite outcome (C-statistics P≤ .011, NRI 13.5-21.1 [95% CI 2.7-31.9] P≤ .014). CONCLUSIONS A panel of biomarkers of gut dysfunction showed graded association with severity of HF and adverse outcomes. Biomarkers as surrogate markers are potentially useful for assessment of gut dysfunction to HF pathophysiology and in risk stratification.
Collapse
Affiliation(s)
- Muhammad Zubair Israr
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Hong Zhan
- Tellgen Corporation, Shanghai, China
| | - Andrea Salzano
- IRCCS SDN, Diagnostic and Nuclear Research Institute, Naples, Italy
| | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John G Cleland
- Robertson Centre for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow and National Heart and Lung Institute, Imperial College, London, UK
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Metra
- Department of Medical and Surgical Specialties, Institute of Cardiology, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Chim C Lang
- School of Medicine Centre for Cardiovascular and Lung Biology, Division of Medical Sciences, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Faiez Zannad
- Inserm CIC 1433, Université de Lorraine, CHU de Nancy, Nancy, France
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Leicester, UK; The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | | |
Collapse
|
11
|
Girerd N, Cleland J, Anker SD, Byra W, Lam CSP, Lapolice D, Mehra MR, van Veldhuisen DJ, Bresso E, Lamiral Z, Greenberg B, Zannad F. Inflammation and remodeling pathways and risk of cardiovascular events in patients with ischemic heart failure and reduced ejection fraction. Sci Rep 2022; 12:8574. [PMID: 35595781 PMCID: PMC9123183 DOI: 10.1038/s41598-022-12385-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
Patients with heart failure (HF) and coronary artery disease (CAD) have a high risk for cardiovascular (CV) events including HF hospitalization, stroke, myocardial infarction (MI) and sudden cardiac death (SCD). The present study evaluated associations of proteomic biomarkers with CV outcome in patients with CAD and HF with reduced ejection fraction (HFrEF), shortly after a worsening HF episode. We performed a case-control study within the COMMANDER HF international, double-blind, randomized placebo-controlled trial investigating the effects of the factor-Xa inhibitor rivaroxaban. Patients with the following first clinical events: HF hospitalization, SCD and the composite of MI or stroke were matched with corresponding controls for age, sex and study drug. Plasma concentrations of 276 proteins with known associations with CV and cardiometabolic mechanisms were analyzed. Results were corrected for multiple testing using false discovery rate (FDR). In 485 cases and 455 controls, 49 proteins were significantly associated with clinical events of which seven had an adjusted FDR < 0.001 (NT-proBNP, BNP, T-cell immunoglobulin and mucin domain containing 4 (TIMD4), fibroblast growth factor 23 (FGF-23), growth differentiation factor-15 (GDF-15), pulmonary surfactant-associated protein D (PSP-D) and Spondin-1 (SPON1)). No significant interactions were identified between the type of clinical event (MI/stroke, SCD or HFH) and specific biomarkers (all interaction FDR > 0.20). When adding the biomarkers significantly associated with the above outcome to a clinical model (including NT-proBNP), the C-index increase was 0.057 (0.033-0.082), p < 0.0001 and the net reclassification index was 54.9 (42.5 to 67.3), p < 0.0001. In patients with HFrEF and CAD following HF hospitalization, we found that NT-proBNP, BNP, TIMD4, FGF-23, GDF-15, PSP-D and SPON1, biomarkers broadly associated with inflammation and remodeling mechanistic pathways, were strong but indiscriminate predictors of a variety of individual CV events.
Collapse
Affiliation(s)
- Nicolas Girerd
- Université de Lorraine, Centre d'Investigation Clinique-Plurithématique Inserm CIC-P 1433, Inserm U1116, CHRU Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - John Cleland
- Robertson Centre for Biostatistics and Clinical Trials, University of Glasgow, Glasgow, Scotland
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - William Byra
- Janssen Research and Development, Raritan, NJ, USA
| | - Carolyn S P Lam
- National Heart Centre Singapore, Duke-National University of Singapore, Singapore, Singapore
| | | | - Mandeep R Mehra
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Emmanuel Bresso
- Université de Lorraine, Centre d'Investigation Clinique-Plurithématique Inserm CIC-P 1433, Inserm U1116, CHRU Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Zohra Lamiral
- Université de Lorraine, Centre d'Investigation Clinique-Plurithématique Inserm CIC-P 1433, Inserm U1116, CHRU Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Barry Greenberg
- Cardiology Division, Department of Medicine, University of California, La Jolla, San Diego, USA
| | - Faiez Zannad
- Université de Lorraine, Centre d'Investigation Clinique-Plurithématique Inserm CIC-P 1433, Inserm U1116, CHRU Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France.
| |
Collapse
|
12
|
Dakhel A, Memon AA, Zarrouk M, Ågren-Witteschus S, Sundquist J, Sundquist K, Gottsäter A. Novel cardiovascular biomarkers associated with peripheral arterial disease in men screened for abdominal aortic aneurysm. VASA 2022; 51:167-173. [DOI: 10.1024/0301-1526/a000999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Summary: Background: Peripheral arterial disease (PAD) is a common atherosclerotic disease with severity ranging from asymptomatic to chronic limb threatening ischemia. The aim of the present cross-sectional study was to identify novel biomarkers associated with PAD. Patients and methods: Levels of 91 cardiovascular specific proteins in plasma samples were measured by the Proseek Multiplex CVD III96x96 panel from a cohort consisting of 267 65-year-old men recruited from a screening program for abdominal aortic aneurysm (AAA) Levels of protein biomarkers were compared in men with and without PAD (defined as an ankle brachial index of <0.9) and their diagnostic potential was calculated by receiver-operating characteristic analysis. Results: The prevalence of PAD was 14.2% (38/267). After adjustment for multiple comparisons, levels of the following 11 biomarkers remained significantly higher ( p<0.0001) in patients with PAD: secretoglobin family 3A member 2, osteoprotegerin, urokinase-type plasminogen activator surface receptor, serum macrophage chemokine ligand 16, matrix metalloproteinase 9, p-selectin, growth differentiation factor 15, elafin, cystatin B, trefoil factor 3, and fatty acid-binding protein 4. Multivariable logistic regression analysis (adjusted for smoking, use of antihypertensive and lipid-lowering medication, and metformin) showed that 11 biomarkers were significantly associated with higher risk of PAD with odds ratios ranging from 1.6 to 2.4. Area under curve calculated by receiver operating characteristic curve analysis (diagnostic value) for each protein biomarker ranged from 0.63 to 0.74. Conclusions: We have identified multiple proteins with a potential to be diagnostic biomarkers for PAD, and further research is warranted to clarify their potential predictive and prognostic value.
Collapse
Affiliation(s)
- Ardwan Dakhel
- Department of Vascular Diseases, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Ashfaque A. Memon
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Moncef Zarrouk
- Department of Vascular Diseases, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Jan Sundquist
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Kristina Sundquist
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Anders Gottsäter
- Department of Acute and Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
13
|
Williams SA, Ostroff R, Hinterberg MA, Coresh J, Ballantyne CM, Matsushita K, Mueller CE, Walter J, Jonasson C, Holman RR, Shah SH, Sattar N, Taylor R, Lean ME, Kato S, Shimokawa H, Sakata Y, Nochioka K, Parikh CR, Coca SG, Omland T, Chadwick J, Astling D, Hagar Y, Kureshi N, Loupy K, Paterson C, Primus J, Simpson M, Trujillo NP, Ganz P. A proteomic surrogate for cardiovascular outcomes that is sensitive to multiple mechanisms of change in risk. Sci Transl Med 2022; 14:eabj9625. [PMID: 35385337 DOI: 10.1126/scitranslmed.abj9625] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A reliable, individualized, and dynamic surrogate of cardiovascular risk, synoptic for key biologic mechanisms, could shorten the path for drug development, enhance drug cost-effectiveness and improve patient outcomes. We used highly multiplexed proteomics to address these objectives, measuring about 5000 proteins in each of 32,130 archived plasma samples from 22,849 participants in nine clinical studies. We used machine learning to derive a 27-protein model predicting 4-year likelihood of myocardial infarction, stroke, heart failure, or death. The 27 proteins encompassed 10 biologic systems, and 12 were associated with relevant causal genetic traits. We independently validated results in 11,609 participants. Compared to a clinical model, the ratio of observed events in quintile 5 to quintile 1 was 6.7 for proteins versus 2.9 for the clinical model, AUCs (95% CI) were 0.73 (0.72 to 0.74) versus 0.64 (0.62 to 0.65), c-statistics were 0.71 (0.69 to 0.72) versus 0.62 (0.60 to 0.63), and the net reclassification index was +0.43. Adding the clinical model to the proteins only improved discrimination metrics by 0.01 to 0.02. Event rates in four predefined protein risk categories were 5.6, 11.2, 20.0, and 43.4% within 4 years; median time to event was 1.71 years. Protein predictions were directionally concordant with changed outcomes. Adverse risks were predicted for aging, approaching an event, anthracycline chemotherapy, diabetes, smoking, rheumatoid arthritis, cancer history, cardiovascular disease, high systolic blood pressure, and lipids. Reduced risks were predicted for weight loss and exenatide. The 27-protein model has potential as a "universal" surrogate end point for cardiovascular risk.
Collapse
Affiliation(s)
| | | | | | - Josef Coresh
- Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | - Christian E Mueller
- Cardiovascular Research Institute, University of Basel, Basel 4001, Switzerland
| | - Joan Walter
- Cardiovascular Research Institute, University of Basel, Basel 4001, Switzerland.,Institute of Diagnostic and Interventional Radiology, University Hospital Zürich, University of Zürich, Zürich 7491, Switzerland
| | - Christian Jonasson
- Jebsen Centre for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Svati H Shah
- Division of Cardiology, Duke Department of Medicine, and Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Roy Taylor
- Newcastle Magnetic Resonance Centre, University of Newcastle upon Tyne, Newcastle upon Tyne NE1 7RU, UK
| | - Michael E Lean
- School of Medicine, Nursing and Dentistry, University of Glasgow, Glasgow G12 8QQ, UK
| | | | - Hiroaki Shimokawa
- Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan.,Graduate School, International University of Health and Welfare, Narita 286-8686, Japan
| | - Yasuhiko Sakata
- Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| | - Kotaro Nochioka
- Tohoku University Graduate School of Medicine, Sendai 980-8576, Japan
| | | | - Steven G Coca
- Mt Sinai Clinical and Translational Science Research Unit, Icahn School of Medicine at Mount Sinai, New York, NY 11766, USA
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital and University of Oslo, Oslo 1478, Norway
| | | | | | | | | | | | | | | | | | | | - Peter Ganz
- Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
14
|
Ceelen D, Voors AA, Tromp J, van Veldhuisen DJ, Dickstein K, de Boer RA, Lang CC, Anker SD, Ng LL, Metra M, Ponikowski P, Figarska SM. Pathophysiological pathways related to high plasma GDF-15 concentrations in patients with heart failure. Eur J Heart Fail 2022; 24:308-320. [PMID: 34989084 PMCID: PMC9302623 DOI: 10.1002/ejhf.2424] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/24/2021] [Accepted: 01/03/2022] [Indexed: 11/11/2022] Open
Abstract
AIMS Elevated concentrations of Growth Differentiation factor 15 (GDF-15) in patients with heart failure (HF) have been consistently associated with worse clinical outcomes, but what disease mechanisms high GDF-15 concentrations represent remains unclear. Here, we aim to identify activated pathophysiological pathways related to elevated GDF-15 expression in patients with HF. METHODS AND RESULTS In 2279 patients with HF, we measured circulating levels of 363 biomarkers. Then, we performed a pathway over-representation analysis to identify key biological pathways between patients in the highest and lowest GDF-15 concentration quartiles. Data were validated in an independent cohort of 1705 patients with HF. In both cohorts, the strongest up-regulated biomarkers in those with high GDF-15 were fibroblast growth factor 23 (FGF-23), death receptor 5 (TRAIL-R2), WNT1-inducible-signaling pathway protein 1 (WISP-1), TNF Receptor Superfamily Member 11a (TNFRSF11A), leukocyte immunoglobulin-like receptor subfamily B member 4 (LILRB4), and Trefoil Factor 3 (TFF3). Pathway over-representation analysis revealed that high GDF-15 patients had increased activity of pathways related to inflammatory processes, notably positive regulation of chemokine production; response to interleukin 6 (IL-6); tumour necrosis factor (TNF) and death receptor activity; and positive regulation of T cell differentiation and inflammatory response. Furthermore, we found pathways involved in regulation of insulin-like growth factor (IGF) receptor signalling and regulatory pathways of tissue, bones, and branching structures. GDF-15 quartiles significantly predicted all-cause mortality and HF hospitalization. CONCLUSION Patients with HF and high plasma concentrations of GDF-15 are characterized by increased activation of inflammatory pathways and pathways related to IGF-1 regulation and bone/tissue remodelling.
Collapse
Affiliation(s)
- Daan Ceelen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jasper Tromp
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,National Heart Centre Singapore, Singapore
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kenneth Dickstein
- University of Bergen, Bergen, Norway.,Stavanger University Hospital, Stavanger, Norway
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Chim C Lang
- School of Medicine Centre for Cardiovascular and Lung Biology, Division of Medical Sciences, University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Stefan D Anker
- Department of Cardiology (CVK); and Berlin Institute of Health Center for Regenerative Therapies (BCRT); German Centre for Cardiovascular Research (DZHK) partner site Berlin; Charité Universitätsmedizin Berlin, Germany
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, and NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Marco Metra
- Institute of Cardiology, ASST Spedali Civili di Brescia and Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Piotr Ponikowski
- Department of Heart Diseases, Wrocław Medical University, Wroclaw, Poland; Center for Heart Diseases, University Hospital in Wrocław, Wroclaw, Poland
| | - Sylwia M Figarska
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
15
|
Diagnostic biomarkers of dilated cardiomyopathy. Immunobiology 2021; 226:152153. [PMID: 34784575 DOI: 10.1016/j.imbio.2021.152153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a condition involving dilation of cardiac chambers, which results in contraction impairment. Besides invasive and non-invasive diagnostic procedures, cardiac biomarkers are of great importance in both diagnosis and prognosis of the disease. These biomarkers are categorized into three groups based on their site; cardiomyocyte biomarkers, microenvironmental biomarkers and macroenvironmental biomarkers. AIMS In this review, an overview of characteristics, epidemiology, etiology and clinical manifestations of DCM is provided. In addition, the most important biomarkers, of all three categories, and their diagnostic and prognostic values are discussed. CONCLUSION Considering the association of DCM with conditions such as infections and autoimmunity, which are prevalent among the population, introducing efficient diagnostic tools is of high value for the early detection of DCM to prevent its severe complications. The three discussed classes of biomarkers are potential candidates for the detection of DCM. However, further studies are necessary in this regard.
Collapse
|
16
|
Mörth C, Sabaa AA, Freyhult E, Christersson C, Hashemi J, Hashemi N, Kamali-Moghaddam M, Molin D, Höglund M, Eriksson A, Enblad G. Plasma proteome profiling of cardiotoxicity in patients with diffuse large B-cell lymphoma. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2021; 7:6. [PMID: 33536059 PMCID: PMC7856776 DOI: 10.1186/s40959-021-00092-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardiovascular toxicity is a notorious complication of doxorubicin (DXR) therapy for diffuse large B-cell lymphoma (DLBCL). Although surveillance of well-known biological markers for cardiovascular disease (CVD) as NTproBNP and Troponins may be helpful, there are no established markers to monitor for evolving CVD during treatment. New possibilities have arisen with the emergence of newer techniques allowing for analysis of plasma proteins that can be associated with cardiovascular disease. Proximity Extension Assay is one of them. OBJECTIVES We aimed to illustrate the incidence of CVD in DLBCL patients treated with DXR and to establish whether there are plasma proteins associated with pre-existing or emerging CVD. METHODS In 95 patients, 182 different proteins from OLINK panels, NTproBNP, Troponin I and CRP were assessed prior to, during and after treatment. For comparison, samples from controls were analyzed. RESULTS In the DLBCL cohort, 33.3% had pre-treatment CVD compared to 5.0% in the controls and 23.2% developed new CVD. Of the 32.6% who died during follow up, CVD was the cause in 4 patients. Spondin-1 (SPON-1) correlated to pre-treatment CVD (1.22 fold change, 95% CI 1.10-1.35, p = 0.00025, q = 0.045). Interleukin-1 receptor type 1 (IL-1RT1) was associated to emerging CVD (1.24 fold change, 95% CI 1.10-1.39, p = 0.00044, q = 0.082). CONCLUSION We observed a higher prevalence of CVD in DLBCL patients compared to controls prior to DXR therapy. Two proteins, SPON-1 and IL-1RT1, were related to pre-existing and emerging CVD in DXR treated patients. If confirmed in larger cohorts, IL-1RT1 may emerge as a reliable biomarker for unfolding CVD in DLBCL.
Collapse
Affiliation(s)
- Charlott Mörth
- Department of Immunology, Genetics & Pathology, Uppsala University, Rudbecklaboratoriet, 75185, Uppsala, Sweden.
- Centre for Clinical Research Sörmland, Uppsala University, Uppsala, Sweden.
| | - Amal Abu Sabaa
- Department of Immunology, Genetics & Pathology, Uppsala University, Rudbecklaboratoriet, 75185, Uppsala, Sweden
- Center for Research and Development, Uppsala University/Region Gävleborg, Uppsala, Sweden
| | - Eva Freyhult
- Department of Medical Sciences, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Jamileh Hashemi
- Department of Immunology, Genetics & Pathology, Uppsala University, Rudbecklaboratoriet, 75185, Uppsala, Sweden
| | - Nashmil Hashemi
- Department of Cardiology, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics & Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Daniel Molin
- Department of Immunology, Genetics & Pathology, Uppsala University, Rudbecklaboratoriet, 75185, Uppsala, Sweden
| | - Martin Höglund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Anna Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics & Pathology, Uppsala University, Rudbecklaboratoriet, 75185, Uppsala, Sweden
| |
Collapse
|
17
|
Osteopontin: The Molecular Bridge between Fat and Cardiac-Renal Disorders. Int J Mol Sci 2020; 21:ijms21155568. [PMID: 32759639 PMCID: PMC7432729 DOI: 10.3390/ijms21155568] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
Osteopontin (OPN) is a multifaceted matricellular protein, with well-recognized roles in both the physiological and pathological processes in the body. OPN is expressed in the main organs and cell types, in which it induces different biological actions. During physiological conditioning, OPN acts as both an intracellular protein and soluble excreted cytokine, regulating tissue remodeling and immune-infiltrate in adipose tissue the heart and the kidney. In contrast, the increased expression of OPN has been correlated with the severity of the cardiovascular and renal outcomes associated with obesity. Indeed, OPN expression is at the “cross roads” of visceral fat extension, cardiovascular diseases (CVDs) and renal disorders, in which OPN orchestrates the molecular interactions, leading to chronic low-grade inflammation. The common factor associated with OPN overexpression in adipose, cardiac and renal tissues seems attributable to the concomitant increase in visceral fat size and the increase in infiltrated OPN+ macrophages. This review underlines the current knowledge on the molecular interactions between obesity and the cardiac–renal disorders ruled by OPN.
Collapse
|
18
|
Fernández Cisnal A, García-Blas S, Valero E, Miñana G, Núñez J, Sanchis Forés J. Trefoil factor-3 and galectin-4 as new candidates for prognostic biomarkers in ST-segment elevation myocardial infarction. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2020; 73:418-420. [PMID: 31761572 DOI: 10.1016/j.rec.2019.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/15/2019] [Indexed: 06/10/2023]
Affiliation(s)
- Agustín Fernández Cisnal
- Hospital Clínic i Universitari de València - Unidad de Hemodinámica y Cardiología Intervencionista, Servicio de Cardiología, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain; Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - Sergio García-Blas
- Hospital Clínic i Universitari de València - Unidad de Hemodinámica y Cardiología Intervencionista, Servicio de Cardiología, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain; Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ernesto Valero
- Hospital Clínic i Universitari de València - Unidad de Hemodinámica y Cardiología Intervencionista, Servicio de Cardiología, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain; Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Gema Miñana
- Hospital Clínic i Universitari de València - Unidad de Hemodinámica y Cardiología Intervencionista, Servicio de Cardiología, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain; Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Julio Núñez
- Hospital Clínic i Universitari de València - Unidad de Hemodinámica y Cardiología Intervencionista, Servicio de Cardiología, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain; Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Juan Sanchis Forés
- Hospital Clínic i Universitari de València - Unidad de Hemodinámica y Cardiología Intervencionista, Servicio de Cardiología, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain; Universidad de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
19
|
Fernández Cisnal A, García-Blas S, Valero E, Miñana G, Núñez J, Sanchis Forés J. Factor trefoil-3 y galectina-4 como nuevos candidatos para biomarcadores pronósticos en infarto de miocardio con elevación del segmento ST. Rev Esp Cardiol (Engl Ed) 2020. [DOI: 10.1016/j.recesp.2019.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Klimczak-Tomaniak D, Bouwens E, Schuurman AS, Akkerhuis KM, Constantinescu A, Brugts J, Westenbrink BD, van Ramshorst J, Germans T, Pączek L, Umans V, Boersma E, Kardys I. Temporal patterns of macrophage- and neutrophil-related markers are associated with clinical outcome in heart failure patients. ESC Heart Fail 2020; 7:1190-1200. [PMID: 32196993 PMCID: PMC7261550 DOI: 10.1002/ehf2.12678] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/17/2020] [Accepted: 02/22/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS Evidence on the association of macrophage- and neutrophil-related blood biomarkers with clinical outcome in heart failure patients is limited, and, with the exception of C-reactive protein, no data exist on their temporal evolution. We aimed to investigate whether temporal patterns of these biomarkers are related to clinical outcome in patients with stable chronic heart failure (CHF). METHODS AND RESULTS In 263 patients with CHF, we performed serial plasma measurements of scavenger receptor cysteine-rich type 1 protein M130 (CD163), tartrate-resistant acid phosphatase type 5 (TRAP), granulins (GRN), spondin-1 (SPON1), peptidoglycan recognition protein 1 (PGLYRP1), and tissue factor pathway inhibitor (TFPI). The Cardiovascular Panel III (Olink Proteomics AB, Uppsala, Sweden) was used. During 2.2 years of follow-up, we collected 1984 samples before the occurrence of the composite primary endpoint (PE) or censoring. For efficiency, we selected 567 samples for the measurements (all baseline samples, the last two samples preceding the PE, and the last sample before censoring in event-free patients). The relationship between repeatedly measured biomarker levels and the PE was evaluated by joint models. Mean (±standard deviation) age was 67 ± 13 years; 189 (72%) were men; left ventricular ejection fraction (%) was 32 ± 11. During follow-up, 70 (27%) patients experienced the PE. Serially measured biomarkers predicted the PE in a multivariable model adjusted for baseline clinical characteristics [hazard ratio (95% confidence interval) per 1-standard deviation change in biomarker]: CD163 [2.07(1.47-2.98), P < 0.001], TRAP [0.62 (0.43-0.90), P = 0.009], GRN [2.46 (1.64-3.84), P < 0.001], SPON1 [3.94 (2.50-6.50), P < 0.001], and PGLYRP1 [1.62 (1.14-2.31), P = 0.006]. CONCLUSIONS Changes in plasma levels of CD163, TRAP, GRN, SPON1, and PGLYRP1 precede adverse cardiovascular events in patients with CHF.
Collapse
Affiliation(s)
- Dominika Klimczak-Tomaniak
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland.,Division of Heart Failure and Cardiac Rehabilitation, Medical University of Warsaw, Warsaw, Poland
| | - Elke Bouwens
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anne-Sophie Schuurman
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alina Constantinescu
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jasper Brugts
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - B Daan Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan van Ramshorst
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Leszek Pączek
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Victor Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Comprehensive analyses of the genome, transcriptome, proteome and metabolome are instrumental in identifying biomarkers of disease, to gain insight into mechanisms underlying the development of cardiovascular disease, and show promise for better stratifying patients according to disease subtypes. This review highlights recent 'omics' studies, including integration of multiple 'omics' that have advanced mechanistic understanding and diagnosis in humans and animal models. RECENT FINDINGS Transcriptome-based discovery continues to be a primary method to obtain data for hypothesis generation and the understanding of disease pathogenesis has been enhanced by single cell-based methods capable of revealing heterogeneity in cellular responses. Advances in proteome coverage and quantitation of individual protein species, together with enhanced methods for detecting posttranslational modifications, have improved discovery of protein-based biomarkers. SUMMARY High-throughput assays capable of quantitating the vast majority of any particular type of biomolecule within a tissue sample, isolated cells or plasma are now available. In order to make best use of the large amount of data that can be generated on given molecule types, as well as their interrelationships in disease, continued development of pattern-recognition algorithms ('machine learning') will be required and the subclassification of disease that is made possible by such algorithms will be likely to inform clinical practice, and vice versa.
Collapse
|
22
|
Stienen S, Ferreira JP, Kobayashi M, Preud'homme G, Dobre D, Machu JL, Duarte K, Bresso E, Devignes MD, López N, Girerd N, Aakhus S, Ambrosio G, Brunner-La Rocca HP, Fontes-Carvalho R, Fraser AG, van Heerebeek L, Heymans S, de Keulenaer G, Marino P, McDonald K, Mebazaa A, Papp Z, Raddino R, Tschöpe C, Paulus WJ, Zannad F, Rossignol P. Enhanced clinical phenotyping by mechanistic bioprofiling in heart failure with preserved ejection fraction: insights from the MEDIA-DHF study (The Metabolic Road to Diastolic Heart Failure). Biomarkers 2020; 25:201-211. [PMID: 32063068 DOI: 10.1080/1354750x.2020.1727015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome for which clear evidence of effective therapies is lacking. Understanding which factors determine this heterogeneity may be helped by better phenotyping. An unsupervised statistical approach applied to a large set of biomarkers may identify distinct HFpEF phenotypes.Methods: Relevant proteomic biomarkers were analyzed in 392 HFpEF patients included in Metabolic Road to Diastolic HF (MEDIA-DHF). We performed an unsupervised cluster analysis to define distinct phenotypes. Cluster characteristics were explored with logistic regression. The association between clusters and 1-year cardiovascular (CV) death and/or CV hospitalization was studied using Cox regression.Results: Based on 415 biomarkers, we identified 2 distinct clusters. Clinical variables associated with cluster 2 were diabetes, impaired renal function, loop diuretics and/or betablockers. In addition, 17 biomarkers were higher expressed in cluster 2 vs. 1. Patients in cluster 2 vs. those in 1 experienced higher rates of CV death/CV hospitalization (adj. HR 1.93, 95% CI 1.12-3.32, p = 0.017). Complex-network analyses linked these biomarkers to immune system activation, signal transduction cascades, cell interactions and metabolism.Conclusion: Unsupervised machine-learning algorithms applied to a wide range of biomarkers identified 2 HFpEF clusters with different CV phenotypes and outcomes. The identified pathways may provide a basis for future research.Clinical significanceMore insight is obtained in the mechanisms related to poor outcome in HFpEF patients since it was demonstrated that biomarkers associated with the high-risk cluster were related to the immune system, signal transduction cascades, cell interactions and metabolismBiomarkers (and pathways) identified in this study may help select high-risk HFpEF patients which could be helpful for the inclusion/exclusion of patients in future trials.Our findings may be the basis of investigating therapies specifically targeting these pathways and the potential use of corresponding markers potentially identifying patients with distinct mechanistic bioprofiles most likely to respond to the selected mechanistically targeted therapies.
Collapse
Affiliation(s)
- Susan Stienen
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - João Pedro Ferreira
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France.,Department of Physiology and Cardiothoracic Surgery, Cardiovascular Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Masatake Kobayashi
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Gregoire Preud'homme
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Daniela Dobre
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France.,Clinical research and Investigation Unit, Psychotherapeutic Center of Nancy, Laxou, France
| | - Jean-Loup Machu
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Kevin Duarte
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Emmanuel Bresso
- Equipe CAPSID, LORIA (CNRS, Inria NGE, Université de Lorraine), Vandoeuvre-lès-Nancy, France
| | | | - Natalia López
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Nicolas Girerd
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Svend Aakhus
- Department of Cardiology and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.,ISB, Norwegian University of Science and Technology, Trondheim, Norway
| | - Giuseppe Ambrosio
- Division of Cardiology, University of Perugia School of Medicine, Perugia, Italy
| | | | - Ricardo Fontes-Carvalho
- Department of Surgery and Physiology, Cardiovascular Research Unit (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
| | - Alan G Fraser
- Wales Heart Research Institute, Cardiff University, Cardiff, UK
| | - Loek van Heerebeek
- Department of Cardiology, Onze Lieve Vrouwe Gasthuis, Amsterdam, the Netherlands
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, Leuven, Belgium.,William Harvey Research Institute, Barts Heart Centre, Queen Mary University of London, London, UK
| | - Gilles de Keulenaer
- Laboratory of Physiopharmacology, Antwerp University, and ZNA Hartcentrum, Antwerp, Belgium
| | - Paolo Marino
- Clinical Cardiology, Università del Piemonte Orientale, Department of Translational Medicine, Azienda Ospedaliero Universitaria "Maggiore della Carità", Novara, Italy
| | - Kenneth McDonald
- School of Medicine and Medical Sciences, St Michael's Hospital Dun Laoghaire Co. Dublin, Dublin, Ireland
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Critical Care Medicine, Saint Louis and Lariboisière University Hospitals and INSERM UMR-S 942, Paris, France
| | - Zoltàn Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Riccardo Raddino
- Department of Cardiology, Spedali Civili di Brescia, Brescia, Italy
| | - Carsten Tschöpe
- Department of Cardiology, Campus Virchow-Klinikum, C, Harite Universitaetsmedizin Berlin, Berlin Institute of Health - Center for Regenerative Therapies (BIH-BCRT), and the German Center for Cardiovascular Research (DZHK; Berlin partner site), Berlin, Germany
| | - Walter J Paulus
- Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Faiez Zannad
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Patrick Rossignol
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| |
Collapse
|
23
|
Colmorten KB, Nexoe AB, Sorensen GL. The Dual Role of Surfactant Protein-D in Vascular Inflammation and Development of Cardiovascular Disease. Front Immunol 2019; 10:2264. [PMID: 31616435 PMCID: PMC6763600 DOI: 10.3389/fimmu.2019.02264] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/09/2019] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular disease (CVD) is responsible for 31% of all global deaths. Atherosclerosis is the major cause of cardiovascular disease and is a chronic inflammatory disorder in the arteries. Atherosclerosis is characterized by the accumulation of cholesterol, extracellular matrix, and immune cells in the vascular wall. Recently, the collectin surfactant protein-D (SP-D), an important regulator of the pulmonary immune response, was found to be expressed in the vasculature. Several in vitro studies have examined the role of SP-D in the vascular inflammation leading to atherosclerosis. These studies show that SP-D plays a dual role in the development of atherosclerosis. In general, SP-D shows anti-inflammatory properties, and dampens local inflammation in the vessel, as well as systemic inflammation. However, SP-D can also exert a pro-inflammatory role, as it stimulates C-C chemokine receptor 2 inflammatory blood monocytes to secrete tumor necrosis-factor α and increases secretion of interferon-γ from natural killer cells. In vivo studies examining the role of SP-D in the development of atherosclerosis agree that SP-D plays a proatherogenic role, with SP-D knockout mice having smaller atherosclerotic plaque areas, which might be caused by a decreased systemic inflammation. Clinical studies examining the association between SP-D and cardiovascular disease have reported a positive association between circulatory SP-D level, carotid intima-media thickness, and coronary artery calcification. Other studies have found that circulatory SP-D is correlated with increased risk of both total and cardiovascular disease mortality. Both in vitro, in vivo, and clinical studies examining the relationship between SP-D and CVDs will be discussed in this review.
Collapse
Affiliation(s)
- Kimmie B Colmorten
- Department of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Anders Bathum Nexoe
- Department of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Grith L Sorensen
- Department of Molecular Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|