1
|
Lee SE, Joo JH, Hwang HS, Chen SF, Evans D, Lee KY, Kim KH, Hyun J, Kim MS, Jung SH, Kim JJ, Lee JS, Torkamani A. Spatial transcriptional landscape of human heart failure. Eur Heart J 2025:ehaf272. [PMID: 40335066 DOI: 10.1093/eurheartj/ehaf272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/30/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND AND AIMS Heart failure (HF) remains a significant clinical challenge due to its diverse aetiologies and complex pathophysiology. The molecular alterations specific to distinct cell types and histological patterns during HF progression are still poorly characterized. This study aimed to explore cell-type- and histology-specific gene expression profiles in cardiomyopathies. METHODS Ninety tissue cores from 44 participants, encompassing various forms of cardiomyopathy and control samples with diverse histological features, were analysed using the GeoMx Whole Human Transcriptome Atlas. Data on cell types, clinical information, and histological features were integrated to examine gene expression profiles in cardiomyopathy. RESULTS The study characterized the cellular composition of ventricular myocardium and validated the GeoMx platform's efficiency in compartmentalizing specific cell types, demonstrating high accuracy for cardiomyocytes but limitations for endothelial cells and fibroblasts. Differentially expressed genes, including UCHL1 from cardiomyocytes, were associated with degeneration, while CCL14, ACKR1, and PLVAP from endothelial cells were linked to fibrosis. Multiplex immunohistochemistry and integrative analysis of prior sc/snRNA-seq data identified a PLVAP, ACKR1, and CCL14-positive pro-inflammatory endothelial cell subtype linked to fibrosis in HF. Downregulation of ribosomal proteins in cardiomyocytes was associated with myocyte disarray in hypertrophic cardiomyopathy. Additionally, pronounced inflammatory responses were observed in end-stage HF. Combined histological and clinical analysis identified CRIP3, PFKFB2, and TAX1BP3 as novel contributors to HF pathogenesis. CONCLUSIONS These findings highlight the critical role of cell-enriched and histology-specific transcriptome mapping in understanding the complex pathophysiological landscape of failing hearts, offering molecular insights and potential therapeutic targets for future interventions.
Collapse
Affiliation(s)
- Sang Eun Lee
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
| | - Jeong Ho Joo
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Hee Sang Hwang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shang-Fu Chen
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
| | - Douglas Evans
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
| | - Kyoung Yul Lee
- Pathology Center, Seegene Medical Foundation, Seoul, Korea
| | - Kyung-Hee Kim
- Division of Cardiology, Cardiovascular Center, Incheon Sejong Hospital, Incheon, Korea
| | - Junho Hyun
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min-Seok Kim
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Ho Jung
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Joong Kim
- Department of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Seok Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Ali Torkamani
- Scripps Research Translational Institute, 3344 North Torrey Pines Court, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| |
Collapse
|
2
|
Chong A, Joshua J, Raheb S, Pires A, Colpitts M, Caswell JL, Fonfara S. Evaluation of potential novel biomarkers for feline hypertrophic cardiomyopathy. Res Vet Sci 2024; 180:105430. [PMID: 39395261 DOI: 10.1016/j.rvsc.2024.105430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common cardiomyopathy in cats. The diagnosis can be difficult, requiring advanced echocardiographic skills. Additionally, circulating biomarkers (N-terminal pro-B type natriuretic peptide and cardiac troponin I) have several limitations when used for HCM screening. In previous work, we identified interleukin 18 (IL-18), insulin-like growth factor binding protein 2 (IGFBP-2), brain-type glycogen phosphorylase B (PYGB), and WNT Family Member 5 A (WNT5A) as myocardial genes that show significant differential expression between cats with HCM and healthy cats. The products of these genes are released into the circulation, and we hypothesized that IL-18, IGFBP-2, PYGB, and WNT5A serum RNA and protein concentrations differ between healthy cats, cats with subclinical HCM, and those with HCM and congestive heart failure (HCM + CHF). Reverse transcriptase quantitative polymerase chain reaction (RTqPCR) and enzyme-linked immunosorbent assay (ELISA) were applied to evaluate gene and protein expression, respectively, in the serum of eight healthy controls, eight cats with subclinical HCM, and six cats with HCM + CHF. Serum IGFBP-2 RNA concentrations were significantly different among groups and were highest in cats with subclinical HCM. Compared to healthy controls, serum IL-18 and WNT5A gene expression were significantly higher in cats with HCM + CHF, and WNT5A was higher in cats with subclinical HCM. No differences were observed for PYGB. These results indicate that further investigation via large scale clinical studies for IGFBP-2, WNT5A, and IL-18 may be valuable in diagnosing and staging feline HCM.
Collapse
Affiliation(s)
- Andrew Chong
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Jessica Joshua
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada; Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road E., Guelph, ON N1G 2W1, Canada
| | - Shari Raheb
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Ananda Pires
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Michelle Colpitts
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Jeff L Caswell
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road E., Guelph, ON N1G 2W1, Canada
| | - Sonja Fonfara
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
3
|
Adasheva DA, Serebryanaya DV. IGF Signaling in the Heart in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1402-1428. [PMID: 39245453 DOI: 10.1134/s0006297924080042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 06/22/2024] [Indexed: 09/10/2024]
Abstract
One of the most vital processes of the body is the cardiovascular system's proper operation. Physiological processes in the heart are regulated by the balance of cardioprotective and pathological mechanisms. The insulin-like growth factor system (IGF system, IGF signaling pathway) plays a pivotal role in regulating growth and development of various cells and tissues. In myocardium, the IGF system provides cardioprotective effects as well as participates in pathological processes. This review summarizes recent data on the role of IGF signaling in cardioprotection and pathogenesis of various cardiovascular diseases, as well as analyzes severity of these effects in various scenarios.
Collapse
Affiliation(s)
- Daria A Adasheva
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Daria V Serebryanaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
4
|
Kobayashi M, Ferreira JP, Duarte K, Bresso E, Huttin O, Bozec E, Brunner La Rocca HP, Delles C, Clark AL, Edelmann F, González A, Heymans S, Pellicori P, Petutschnigg J, Verdonschot JAJ, Rossignol P, Cleland JGF, Zannad F, Girerd N. Proteomic profiles of left atrial volume and its influence on response to spironolactone: Findings from the HOMAGE trial and STANISLAS cohort. Eur J Heart Fail 2024; 26:1231-1241. [PMID: 38528728 DOI: 10.1002/ejhf.3202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 03/27/2024] Open
Abstract
AIMS High left ventricular filling pressure increases left atrial volume and causes myocardial fibrosis, which may decrease with spironolactone. We studied clinical and proteomic characteristics associated with left atrial volume indexed by body surface area (LAVi), and whether LAVi influences the response to spironolactone on biomarker expression and clinical variables. METHODS AND RESULTS In the HOMAGE trial, where people at risk of heart failure were randomized to spironolactone or control, we analysed 421 participants with available LAVi and 276 proteomic measurements (Olink) at baseline, month 1 and 9 (mean age 73 ± 6 years; women 26%; LAVi 32 ± 9 ml/m2). Circulating proteins associated with LAVi were also assessed in asymptomatic individuals from a population-based cohort (STANISLAS; n = 1640; mean age 49 ± 14 years; women 51%; LAVi 23 ± 7 ml/m2). In both studies, greater LAVi was significantly associated with greater left ventricular masses and volumes. In HOMAGE, after adjustment and correction for multiple testing, greater LAVi was associated with higher concentrations of matrix metallopeptidase-2 (MMP-2), insulin-like growth factor binding protein-2 (IGFBP-2) and N-terminal pro-B-type natriuretic peptide (NT-proBNP) (false discovery rates [FDR] <0.05). These associations were externally replicated in STANISLAS (all FDR <0.05). Among these biomarkers, spironolactone decreased concentrations of MMP-2 and NT-proBNP, regardless of baseline LAVi (pinteraction > 0.10). Spironolactone also significantly reduced LAVi, improved left ventricular ejection fraction, lowered E/e', blood pressure and serum procollagen type I C-terminal propeptide (PICP) concentration, a collagen synthesis marker, regardless of baseline LAVi (pinteraction > 0.10). CONCLUSION In individuals without heart failure, LAVi was associated with MMP-2, IGFBP-2 and NT-proBNP. Spironolactone reduced these biomarker concentrations as well as LAVi and PICP, irrespective of left atrial size.
Collapse
Affiliation(s)
- Masatake Kobayashi
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- Department of Cardiology, Tokyo Medical University Hospital, Tokyo, Japan
| | - João Pedro Ferreira
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- Cardiovascular Research and Development Center, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Kevin Duarte
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Emmanuel Bresso
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Olivier Huttin
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Erwan Bozec
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | | | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew L Clark
- Department of Cardiology, University of Hull, Castle Hill Hospital, Yorkshire, UK
| | - Frank Edelmann
- Department of Internal Medicine and Cardiology Campus Virchow Klinikum, Charité University Medicine Berlin and German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Arantxa González
- CIMA Universidad de Navarra, Department of Pathology, Anatomy and Physiology Universidad de Navarra and IdiSNA, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Stephane Heymans
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pierpaolo Pellicori
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Johannes Petutschnigg
- Department of Internal Medicine and/Cardiology, Campus Virchow Klinikum, Charité University Medicine Berlin, and German Heart Center Berlin, and Berlin Institute of Health (BIH), and German Centre for Cardiovascular research (DZHK), Berlin, Germany
| | - Job A J Verdonschot
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Patrick Rossignol
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
- Medical Specialties and Nephrology Dialysis Departments, Monaco Princess Grace Hospital and Monaco Private Hemodialysis Centre, Monaco, Monaco
| | - John G F Cleland
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Faiez Zannad
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, INSERM, Centre d'Investigations Cliniques Plurithématique 1433, Inserm U1116, CHRU de Nancy and F-CRIN INI-CRCT, Nancy, France
| |
Collapse
|
5
|
Keil L, Berisha F, Ritter S, Skibowski J, Subramanian H, Nikolaev VO, Kubisch C, Woitschach R, Fabritz L, Twerenbold R, Blankenberg S, Weidemann S, Zeller T, Kirchhof P, Reichart D, Magnussen C. Multimodal characterization of dilated cardiomyopathy: Geno- And Phenotyping of PrImary Cardiomyopathy (GrAPHIC). ESC Heart Fail 2024; 11:541-549. [PMID: 37964758 PMCID: PMC10804161 DOI: 10.1002/ehf2.14544] [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: 04/11/2023] [Revised: 08/23/2023] [Accepted: 09/15/2023] [Indexed: 11/16/2023] Open
Abstract
AIMS Cardiomyopathies (CMPs) are a heterogeneous group of diseases that are defined by structural and functional abnormalities of the cardiac muscle. Dilated cardiomyopathy (DCM), the most common CMP, is defined by left ventricular dilation and impaired contractility and represents a common cause of heart failure. Different phenotypes result from various underlying genetic and acquired causes with variable effects on disease development and progression, prognosis, and response to medical treatment. Current treatment algorithms do not consider these different aetiologies, due to lack of insights into treatable drivers of cardiac failure in patients with DCM. Our study aims to precisely phenotype and genotype the various subtypes of DCM and hereby lay the foundation for individualized therapy. METHODS AND RESULTS The Geno- And Phenotyping of PrImary Cardiomyopathy (GrAPHIC) is a currently ongoing prospective observational monocentric cohort study that recruits patients with DCM after exclusion of other causes such as coronary artery disease, valvular dysfunction, myocarditis, exposure to toxins, and peripartum CMP. Patients are enrolled at our heart failure outpatient clinic or during hospitalization at the University Hospital Hamburg. Clinical parameters, multimodal imaging and functional assessment, cardiac biopsies, and blood samples are obtained to enable an integrated genomic, functional, and biomarker analysis. CONCLUSIONS The GrAPHIC will contribute to a better understanding of the heterogeneous nature of primary CMPs focusing on DCM and provide improved prognostic approaches and more individualized therapies.
Collapse
Affiliation(s)
- Laura Keil
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Filip Berisha
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Stella Ritter
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Johanna Skibowski
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Hariharan Subramanian
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Viacheslav O. Nikolaev
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- Institute of Experimental Cardiovascular ResearchUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christian Kubisch
- Institute of Human GeneticsUniversity Hospital Hamburg‐EppendorfHamburgGermany
| | - Rixa Woitschach
- Institute of Human GeneticsUniversity Hospital Hamburg‐EppendorfHamburgGermany
| | - Larissa Fabritz
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- University Centre of Cardiovascular Science, UKE HamburgHamburgGermany
| | - Raphael Twerenbold
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- University Centre of Cardiovascular Science, UKE HamburgHamburgGermany
| | - Stefan Blankenberg
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
| | - Sören Weidemann
- Department of PathologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Tanja Zeller
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
- University Centre of Cardiovascular Science, UKE HamburgHamburgGermany
| | - Paulus Kirchhof
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
| | - Daniel Reichart
- Department of Medicine IUniversity Hospital, LMU MunichMunichGermany
| | - Christina Magnussen
- Department of CardiologyUniversity Heart and Vascular Center Hamburg, University Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/LuebeckHamburgGermany
| |
Collapse
|
6
|
Woo J, Lu D, Lewandowski A, Xu H, Serrano P, Healey M, Yates DP, Beste MT, Libby P, Ridker PM, Steensma DP. Effects of IL-1β inhibition on anemia and clonal hematopoiesis in the randomized CANTOS trial. Blood Adv 2023; 7:7471-7484. [PMID: 37934948 PMCID: PMC10758744 DOI: 10.1182/bloodadvances.2023011578] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/11/2023] [Accepted: 10/29/2023] [Indexed: 11/09/2023] Open
Abstract
Canakinumab, a monoclonal antibody targeting proinflammatory cytokine interleukin-1β (IL-1β), improved hemoglobin levels while preventing recurrent cardiovascular events in the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). This cardiovascular (CV) preventive effect was greater in patients with TET2 mutations associated with clonal hematopoiesis (CH). The current proteogenomic analysis aimed to understand the clinical response to canakinumab and underlying proteomic profiles in the context of CH and anemia. The analysis included 4595 patients from the CANTOS study who received either canakinumab or placebo and evaluated multiplexed proteomics (4785 proteins) using SomaScan and targeted deep sequencing for CH mutations. Incident anemia was more common in the presence of CH mutations but reduced by canakinumab treatment. Canakinumab treatment was significantly associated with higher hemoglobin increment in patients with concurrent CH mutations and anemia than patients with CH mutations without anemia or without CH mutations. Compared with those without CH mutations, the presence of CH mutations was associated with proteomic signatures of inflammation and defense response to infection, as well as markers of high-risk CV disease which was further enhanced by the presence of anemia. Canakinumab suppressed hepcidin, proinflammatory cytokines, myeloid activation, and complement pathways, and reversed pathologically deregulated pathways to a greater extent in patients with CH mutations and anemia. These molecular findings provide evidence of the clinical use of IL-1β blockade and support further study of canakinumab for patients with concurrent anemia and CH mutations. This study was registered at www.clinicaltrials.gov as #NCT01327846.
Collapse
Affiliation(s)
- Janghee Woo
- Novartis Institutes for BioMedical Research, Cambridge, MA
| | - Darlene Lu
- Novartis Institutes for BioMedical Research, Cambridge, MA
| | | | - Huilei Xu
- Novartis Institutes for BioMedical Research, Cambridge, MA
| | - Pablo Serrano
- Novartis Institutes for BioMedical Research, Cambridge, MA
| | | | | | | | - Peter Libby
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Paul M. Ridker
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Boston, MA
| | | |
Collapse
|
7
|
Bello C, Filipovic MG, Huber M, Flannery S, Kobel B, Fischer R, Kessler BM, Räber L, Stueber F, Luedi MM. Discovery of plasma proteome markers associated with clinical outcome and immunological stress after cardiac surgery. Front Cardiovasc Med 2023; 10:1287724. [PMID: 38379859 PMCID: PMC10876477 DOI: 10.3389/fcvm.2023.1287724] [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: 09/02/2023] [Accepted: 11/20/2023] [Indexed: 02/22/2024] Open
Abstract
Background Molecular mechanisms underlying perioperative acute phase reactions in cardiac surgery are largely unknown. We aimed to characterise perioperative alterations of the acute phase plasma proteome in a cohort of adult patients undergoing on-pump cardiac surgery using high-throughput mass spectrometry and to identify candidate proteins potentially relevant to postoperative clinical outcome through a novel, multi-step approach. Methods This study is an analysis of the Bern Perioperative Biobank, a prospective cohort of adults who underwent cardiac surgery with the use of cardiopulmonary bypass (CPB) at Bern University Hospital between January and December 2019. Blood samples were taken before induction of anaesthesia and on postoperative day one. Proteomic analyses were performed by mass spectrometry. Through a multi-step, exploratory approach, hit-proteins were first identified according to their perioperative prevalence and dynamics. The set of hit-proteins were associated with predefined clinical outcome measures (all-cause one-year mortality, length of hospital stay, postoperative myocardial infarction and stroke until hospital discharge). Results 192 patients [75.5% male, median age 67.0 (IQR 60.0-73.0)] undergoing cardiac surgery with the use of CPB were included in this analysis. In total, we identified and quantified 402 proteins across all samples, whereof 30/402 (7%) proteins were identified as hit-proteins. Three hit-proteins-LDHB, VCAM1 and IGFBP2-demonstrated the strongest associations with clinical outcomes. After adjustment both for age, sex, BMI and for multiple comparisons, the scaled preoperative levels of IGFBP2 were associated with 1-year all-cause mortality (OR 10.63; 95% CI: 2.93-64.00; p = 0.046). Additionally, scaled preoperative levels of LDHB (OR 5.58; 95% CI: 2.58-8.57; p = 0.009) and VCAM1 (OR 2.32; 95% CI: 0.88-3.77; p = 0.05) were found to be associated with length of hospital stay. Conclusions We identified a subset of promising candidate plasma proteins relevant to outcome after on-pump cardiac surgery. IGFBP2 showed a strong association with clinical outcome measures and a significant association of preoperative levels with 1-year all-cause mortality. Other proteins strongly associated with outcome were LDHB and VCAM1, reflecting the dynamics in the acute phase response, inflammation and myocardial injury. We recommend further investigation of these proteins as potential outcome markers after cardiac surgery. Clinical Trial Registration ClinicalTrials.gov; NCT04767685, data are available via ProteomeXchange with identifier PXD046496.
Collapse
Affiliation(s)
- Corina Bello
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Mark G. Filipovic
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Markus Huber
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sarah Flannery
- Nuffield Department of Medicine, Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Beatrice Kobel
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Benedikt M. Kessler
- Nuffield Department of Medicine, Target Discovery Institute, Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Lorenz Räber
- Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Frank Stueber
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Markus M. Luedi
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Hou Q, Sun Z, Zhao L, Liu Y, Zhang J, Huang J, Luo Y, Xiao Y, Hu Z, Shen A. Role of serum cytokines in the prediction of heart failure in patients with coronary artery disease. ESC Heart Fail 2023; 10:3102-3113. [PMID: 37608687 PMCID: PMC10567644 DOI: 10.1002/ehf2.14491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/12/2023] [Accepted: 07/16/2023] [Indexed: 08/24/2023] Open
Abstract
AIMS Coronary artery disease (CAD) is the most common cause of heart failure (HF). This study aimed to identify cytokine biomarkers for predicting HF in patients with CAD. METHODS AND RESULTS Twelve patients with CAD without HF (CAD-non HF), 12 patients with CAD complicated with HF (CAD-HF), and 12 healthy controls were enrolled for Human Cytokine Antibody Array, which were used as the training dataset. Then, differentially expressed cytokines among the different groups were identified, and crucial characteristic proteins related to CAD-HF were screened using a combination of the least absolute shrinkage and selection operator, recursive feature elimination, and random forest methods. A support vector machine (SVM) diagnostic model was constructed based on crucial characteristic proteins, followed by receiver operating characteristic curve analysis. Finally, two validation datasets, GSE20681 and GSE59867, were downloaded to verify the diagnostic performance of the SVM model and expression of crucial proteins, as well as enzyme-linked immunosorbent assay was also used to verify the levels of crucial proteins in blood samples. In total, 12 differentially expressed proteins were overlapped in the three comparison groups, and then four optimal characteristic proteins were identified, including VEGFR2, FLRG, IL-23, and FGF-21. After that, the area under the receiver operating characteristic curve of the constructed SVM classification model for the training dataset was 0.944. The accuracy of the SVM classification model was validated using the GSE20681 and GSE59867 datasets, with area under the receiver operating characteristic curve values of 0.773 and 0.745, respectively. The expression trends of the four crucial proteins in the training dataset were consistent with those in the validation dataset and those determined by enzyme-linked immunosorbent assay. CONCLUSIONS The combination of VEGFR2, FLRG, IL-23, and FGF-21 can be used as a candidate biomarker for the prediction and prevention of HF in patients with CAD.
Collapse
Affiliation(s)
- Qingzhen Hou
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhuhua Sun
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Liqin Zhao
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Ye Liu
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Junfang Zhang
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Jing Huang
- Department of Laboratory Medicine, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Yifeng Luo
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Yan Xiao
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhaoting Hu
- Department of Health Management Center, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| | - Anna Shen
- Department of Cardiology, The Third Affiliated HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
9
|
Thijssen CGE, Dekker S, Bons LR, Geenen LW, Gökalp AL, Takkenberg JJM, Mokhles MM, Bekkers JA, Boersma E, Bouwens E, van Kimmenade RRJ, Roos-Hesselink JW. Novel biomarkers associated with thoracic aortic disease. Int J Cardiol 2023; 378:115-122. [PMID: 36796491 DOI: 10.1016/j.ijcard.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 01/18/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Biomarkers might help to improve diagnosis, surveillance and risk stratification of thoracic aortic disease (TAD). We explored the association between a broad spectrum of cardiovascular biomarkers with clinical characteristics and thoracic aortic diameter in TAD patients. METHODS Venous blood-samples were obtained in 158 clinically stable TAD patients visiting our outpatient clinic (2017-2020). TAD was defined as a thoracic aortic diameter ≥ 40 mm, or genetic confirmation (hereditary TAD). The cardiovascular panel III of the Olink multiplex platform was used for batch analysis of 92 proteins. A comparison was made between biomarker levels in patients with and without previous aortic dissection and/or surgery, and with and without hereditary TAD. Linear regression analyses were applied to identify (relative, normalized) biomarker concentrations associated with the absolute thoracic aortic diameter (ADmax), and thoracic aortic diameter indexed for body surface area (IDmax). RESULTS Median age of study patients was 61.0 (IQR 50.3-68.8) years, 37.3% females. Mean ADmax and IDmax were 43.3 ± 5.4 mm and 21.3 ± 3.3 mm/m2. After multivariable adjustment, Matrix Metalloproteinase-3 (MMP-3) and Insulin-like growth factor binding protein 2 (IGFBP-2) showed a significant positive association with ADmax and IDmax, respectively. Patients with previous aortic surgery/dissection had higher N-terminal-pro hormone BNP (NTproBNP) (median 3.67 [IQR 3.01-3.99] vs 2.84 [2.32-3.26], p ≤0.001). Patients with hereditary TAD had higher Trem-like transcript protein 2 (TLT-2) (median 4.64 [IQR 4.45-4.84]) than those with non-heriditary TAD (4.40 [4.17-4.64]; p = 0.00042). CONCLUSIONS Among a broad range of biomarkers, MMP-3 and IGFBP-2 were associated with disease severity in TAD patients. The pathophysiological pathways uncovered by these biomarkers, and their potential clinical use warrants further research.
Collapse
Affiliation(s)
- Carlijn G E Thijssen
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Silvy Dekker
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands
| | - Lidia R Bons
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands
| | - Laurie W Geenen
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands
| | - Arjen L Gökalp
- Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, the Netherlands
| | | | - Mostafa M Mokhles
- Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, the Netherlands; Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jos A Bekkers
- Department of Cardiothoracic Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands
| | - Elke Bouwens
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands; Department of Anesthesiology, Erasmus MC, Rotterdam, the Netherlands
| | - Roland R J van Kimmenade
- Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | |
Collapse
|
10
|
Batra R, Uni R, Akchurin OM, Alvarez-Mulett S, Gómez-Escobar LG, Patino E, Hoffman KL, Simmons W, Whalen W, Chetnik K, Buyukozkan M, Benedetti E, Suhre K, Schenck E, Cho SJ, Choi AMK, Schmidt F, Choi ME, Krumsiek J. Urine-based multi-omic comparative analysis of COVID-19 and bacterial sepsis-induced ARDS. Mol Med 2023; 29:13. [PMID: 36703108 PMCID: PMC9879238 DOI: 10.1186/s10020-023-00609-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/11/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS), a life-threatening condition during critical illness, is a common complication of COVID-19. It can originate from various disease etiologies, including severe infections, major injury, or inhalation of irritants. ARDS poses substantial clinical challenges due to a lack of etiology-specific therapies, multisystem involvement, and heterogeneous, poor patient outcomes. A molecular comparison of ARDS groups holds the potential to reveal common and distinct mechanisms underlying ARDS pathogenesis. METHODS We performed a comparative analysis of urine-based metabolomics and proteomics profiles from COVID-19 ARDS patients (n = 42) and bacterial sepsis-induced ARDS patients (n = 17). To this end, we used two different approaches, first we compared the molecular omics profiles between ARDS groups, and second, we correlated clinical manifestations within each group with the omics profiles. RESULTS The comparison of the two ARDS etiologies identified 150 metabolites and 70 proteins that were differentially abundant between the two groups. Based on these findings, we interrogated the interplay of cell adhesion/extracellular matrix molecules, inflammation, and mitochondrial dysfunction in ARDS pathogenesis through a multi-omic network approach. Moreover, we identified a proteomic signature associated with mortality in COVID-19 ARDS patients, which contained several proteins that had previously been implicated in clinical manifestations frequently linked with ARDS pathogenesis. CONCLUSION In summary, our results provide evidence for significant molecular differences in ARDS patients from different etiologies and a potential synergy of extracellular matrix molecules, inflammation, and mitochondrial dysfunction in ARDS pathogenesis. The proteomic mortality signature should be further investigated in future studies to develop prediction models for COVID-19 patient outcomes.
Collapse
Affiliation(s)
- Richa Batra
- grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Rie Uni
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY USA
| | - Oleh M. Akchurin
- grid.5386.8000000041936877XDivision of Pediatric Nephrology, Department of Pediatrics, Weill Cornell Medicine, New York, NY USA ,grid.413734.60000 0000 8499 1112New York-Presbyterian Hospital, New York, NY USA
| | - Sergio Alvarez-Mulett
- grid.5386.8000000041936877XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Luis G. Gómez-Escobar
- grid.5386.8000000041936877XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Edwin Patino
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY USA
| | - Katherine L. Hoffman
- grid.5386.8000000041936877XDivision of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY USA
| | - Will Simmons
- grid.5386.8000000041936877XDivision of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY USA
| | - William Whalen
- grid.5386.8000000041936877XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Kelsey Chetnik
- grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Mustafa Buyukozkan
- grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Elisa Benedetti
- grid.5386.8000000041936877XDepartment of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Karsten Suhre
- grid.418818.c0000 0001 0516 2170Bioinformatics Core, Weill Cornell Medicine –Qatar, Qatar Foundation, Doha, Qatar
| | - Edward Schenck
- grid.5386.8000000041936877XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Soo Jung Cho
- grid.5386.8000000041936877XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Augustine M. K. Choi
- grid.5386.8000000041936877XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine -Qatar, Qatar Foundation, Doha, Qatar.
| | - Mary E. Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
11
|
Batra R, Uni R, Akchurin OM, Alvarez-Mulett S, Gómez-Escobar LG, Patino E, Hoffman KL, Simmons W, Chetnik K, Buyukozkan M, Benedetti E, Suhre K, Schenck E, Cho SJ, Choi AMK, Schmidt F, Choi ME, Krumsiek J. Urine-based multi-omic comparative analysis of COVID-19 and bacterial sepsis-induced ARDS. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.08.10.22277939. [PMID: 35982662 PMCID: PMC9387152 DOI: 10.1101/2022.08.10.22277939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Acute respiratory distress syndrome (ARDS), a life-threatening condition during critical illness, is a common complication of COVID-19. It can originate from various disease etiologies, including severe infections, major injury, or inhalation of irritants. ARDS poses substantial clinical challenges due to a lack of etiology-specific therapies, multisystem involvement, and heterogeneous, poor patient outcomes. A molecular comparison of ARDS groups holds the potential to reveal common and distinct mechanisms underlying ARDS pathogenesis. In this study, we performed a comparative analysis of urine-based metabolomics and proteomics profiles from COVID-19 ARDS patients (n = 42) and bacterial sepsis-induced ARDS patients (n = 17). The comparison of these ARDS etiologies identified 150 metabolites and 70 proteins that were differentially abundant between the two groups. Based on these findings, we interrogated the interplay of cell adhesion/extracellular matrix molecules, inflammation, and mitochondrial dysfunction in ARDS pathogenesis through a multi-omic network approach. Moreover, we identified a proteomic signature associated with mortality in COVID-19 ARDS patients, which contained several proteins that had previously been implicated in clinical manifestations frequently linked with ARDS pathogenesis. In summary, our results provide evidence for significant molecular differences in ARDS patients from different etiologies and a potential synergy of extracellular matrix molecules, inflammation, and mitochondrial dysfunction in ARDS pathogenesis. The proteomic mortality signature should be further investigated in future studies to develop prediction models for COVID-19 patient outcomes.
Collapse
Affiliation(s)
- Richa Batra
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Rie Uni
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Oleh M Akchurin
- Department of Pediatrics, Division of Pediatric Nephrology, Weill Cornell Medicine, New York, NY, USA
- New York-Presbyterian Hospital, New York, NY, USA
| | - Sergio Alvarez-Mulett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Luis G Gómez-Escobar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Edwin Patino
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Katherine L Hoffman
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, NY, USA
| | - Will Simmons
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, NY, USA
| | - Kelsey Chetnik
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mustafa Buyukozkan
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Elisa Benedetti
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine - Qatar, Qatar Foundation, Doha, Qatar
| | - Edward Schenck
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Soo Jung Cho
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Augustine M K Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine - Qatar, Qatar Foundation, Doha, Qatar
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
12
|
Topf A, Mirna M, Paar V, Motloch LJ, Bacher N, Franz M, Hoppe UC, Kretzschmar D, Lichtenauer M. Differential Diagnosis between Takotsubo Syndrome and Acute Coronary Syndrome—A Prospective Analysis of Novel Cardiovascular Biomarkers for a More Selective Triage. J Clin Med 2022; 11:jcm11112974. [PMID: 35683362 PMCID: PMC9180967 DOI: 10.3390/jcm11112974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/11/2022] Open
Abstract
Introduction: Takotsubo syndrome (TTS) is clinically indistinguishable from an ACS. Despite the implementation of clinical scoring systems and novel biomarkers, coronary angiography currently remains necessary for differential diagnosis. Methods: 93 patients with chest pain and the suspicion of TTS were enrolled in two study centers. Fetuin-A, IGFBP-2, Galectin-3, and TNF α were determined in serum samples, collected within 24 h after the onset of symptoms. Serum levels of biomarkers were analyzed for the differential diagnostic value between TTS and ACS. Results: Compared to TTS, patients with ACS had significantly lower serum levels of Fetuin-A and IGFBP-2. The cut-off value of Fetuin-A for the identification of TTS compared to ACS was 55.74 μg/mL (sensitivity: 100.0%, specificity: 82.6%, PPV: 63.2%, NPV: 100.0%). An optimal cut-off value for IGFBP-2 for the differential diagnosis between TTS and ACS was determined as 171.77 ng/mL (sensitivity: 76.0%, specificity: 82.6%, PPV: 76.4%, NPV 72.7%). Conclusion: Fetuin-A and IGFBP-2 might facilitate the triage between TTS and ACS and could be therefore of great benefit for the guidance of treatment.
Collapse
Affiliation(s)
- Albert Topf
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
- Correspondence: ; Tel.: +43-(0)-57-57418; Fax: +43-(0)-57255-4111
| | - Moritz Mirna
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Vera Paar
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Lukas J. Motloch
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Nina Bacher
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Marcus Franz
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, 07743 Jena, Germany; (M.F.); (D.K.)
| | - Uta C. Hoppe
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Daniel Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, 07743 Jena, Germany; (M.F.); (D.K.)
| | - Michael Lichtenauer
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| |
Collapse
|
13
|
McNeill JN, Lee DH, Hwang SJ, Courchesne P, Yao C, Huan T, Joehanes R, O’Connor GT, Ho JE, Levy D. Association of 71 cardiovascular disease-related plasma proteins with pulmonary function in the community. PLoS One 2022; 17:e0266523. [PMID: 35390066 PMCID: PMC8989231 DOI: 10.1371/journal.pone.0266523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 03/22/2022] [Indexed: 11/19/2022] Open
Abstract
RATIONALE It has been speculated that shared mechanisms underlie respiratory and cardiovascular diseases (CVD) including systemic inflammation or mutual risk factors. In this context, we sought to examine the associations of CVD-related plasma proteins with lung function as measured by spirometry in a large community-based cohort of adults. METHODS The study included 5777 Framingham Heart Study participants who had spirometry and measurement of 71 CVD-related plasma proteins. The association of plasma proteins with lung function was assessed cross-sectionally and longitudinally using models accounting for familial correlations. Linear mixed models were used for the following measurements: FEV1%predicted, FVC%predicted, and FEV1/FVC ratio with secondary analyses examining obstructive and restrictive physiology at baseline and their new onset during follow up. MEASUREMENTS AND MAIN RESULTS Among the 71 CVD-related plasma proteins, 13 proteins were associated in cross-sectional analyses with FEV1%predicted, 17 proteins were associated with FVC%predicted, and 1 protein was associated with FEV1/FVC. The proteins with the greatest inverse relations to FEV1%predicted and FVC%predicted included leptin, adrenomedullin, and plasminogen activator inhibitor-1; in contrast there were three proteins with positive relations to FEV1%predicted and FVC%predicted including insulin growth factor binding protein 2, tetranectin, and soluble receptor for advanced glycation end products. In longitudinal analyses, three proteins were associated with longitudinal change in FEV1 (ΔFEV1) and four with ΔFVC; no proteins were associated with ΔFEV1/FVC. CONCLUSION Our findings highlight CVD-related plasma proteins that are associated with lung function including markers of inflammation, adiposity, and fibrosis, representing proteins that may contribute both to respiratory and CVD risk.
Collapse
Affiliation(s)
- Jenna N. McNeill
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Dong Heon Lee
- The Framingham Heart Study, Framingham, Massachusetts, and the Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Shih-Jen Hwang
- The Framingham Heart Study, Framingham, Massachusetts, and the Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Paul Courchesne
- The Framingham Heart Study, Framingham, Massachusetts, and the Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Chen Yao
- The Framingham Heart Study, Framingham, Massachusetts, and the Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tianxiao Huan
- The Framingham Heart Study, Framingham, Massachusetts, and the Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Roby Joehanes
- The Framingham Heart Study, Framingham, Massachusetts, and the Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - George T. O’Connor
- Pulmonary Center, Boston University, Boston, Massachusetts, United States of America
| | - Jennifer E. Ho
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Daniel Levy
- The Framingham Heart Study, Framingham, Massachusetts, and the Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
14
|
Identification and Comparison of Potential Biomarkers by Proteomic Analysis in Traditional Chinese Medicine-Based Heart Failure Syndromes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6338508. [PMID: 35087594 PMCID: PMC8789435 DOI: 10.1155/2022/6338508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/27/2021] [Indexed: 11/17/2022]
Abstract
Heart failure (HF) is an epidemic disease affecting a large population worldwide. Traditional Chinese medicine (TCM) is playing an increasingly important role in the clinical treatment of HF. According to the TCM theory, HF could be classified into Yang deficiency and Qi-yin deficiency; however, there are few objective and biological lines of evidence for differentiation of TCM HF syndromes to date. In this study, data-independent acquisition (DIA) mass spectrometry was applied to comparatively analyze the protein expression in serum samples obtained from 12 Yang deficiency patients, 12 Qi-yin deficiency patients, and 12 healthy volunteers. Compared to the healthy controls, a total of 121 differentially expressed proteins (DEPs) (77 upregulated and 44 downregulated proteins) were identified in Yang deficiency samples, while 59 DEPs (49 upregulated and 10 downregulated proteins) were detected in Qi-yin deficiency samples. Enrichment analyses of these DEPs based on the GO and KEGG databases revealed functional clusters associated with the immune system, signal transduction, and infectious disease. Several previously reported HF biomarker proteins were found to be the hub proteins in a protein-protein interaction network analysis. Three novel hub DEPs were identified as potential biomarkers for differentiation between different TCM syndromes of HF. The results provide biological insight into the differences of different TCM HF syndromes and an opportunity for specific biomarker identification for different TCM HF syndromes.
Collapse
|
15
|
Tantawy M, Chekka LM, Huang Y, Garrett TJ, Singh S, Shah CP, Cornell RF, Baz RC, Fradley MG, Waheed N, DeRemer DL, Yuan L, Langaee T, March K, Pepine CJ, Moreb JS, Gong Y. Lactate Dehydrogenase B and Pyruvate Oxidation Pathway Associated With Carfilzomib-Related Cardiotoxicity in Multiple Myeloma Patients: Result of a Multi-Omics Integrative Analysis. Front Cardiovasc Med 2021; 8:645122. [PMID: 33996940 PMCID: PMC8116486 DOI: 10.3389/fcvm.2021.645122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/02/2021] [Indexed: 01/20/2023] Open
Abstract
Multiple myeloma (MM) is the second most frequent hematologic cancer in the United States. Carfilzomib (CFZ), an irreversible proteasome inhibitor being used to treat relapsed and refractory MM, has been associated with cardiotoxicity, including heart failure. We hypothesized that a multi-omics approach integrating data from different omics would provide insights into the mechanisms of CFZ-related cardiovascular adverse events (CVAEs). Plasma samples were collected from 13 MM patients treated with CFZ (including 7 with CVAEs and 6 with no CVAEs) at the University of Florida Health Cancer Center. These samples were evaluated in global metabolomic profiling, global proteomic profiling, and microRNA (miRNA) profiling. Integrative pathway analysis was performed to identify genes and pathways differentially expressed between patients with and without CVAEs. The proteomics analysis identified the up-regulation of lactate dehydrogenase B (LDHB) [fold change (FC) = 8.2, p = 0.01] in patients who experienced CVAEs. The metabolomics analysis identified lower plasma abundance of pyruvate (FC = 0.16, p = 0.0004) and higher abundance of lactate (FC = 2.4, p = 0.0001) in patients with CVAEs. Differential expression analysis of miRNAs profiling identified mir-146b to be up-regulatein (FC = 14, p = 0.046) in patients with CVAE. Pathway analysis suggested that the pyruvate fermentation to lactate pathway is associated with CFZ-CVAEs. In this pilot multi-omics integrative analysis, we observed the down-regulation of pyruvate and up-regulation of LDHB among patients who experienced CVAEs, suggesting the importance of the pyruvate oxidation pathway associated with mitochondrial dysfunction. Validation and further investigation in a larger independent cohort are warranted to better understand the mechanisms of CFZ-CVAEs.
Collapse
Affiliation(s)
- Marwa Tantawy
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Lakshmi Manasa Chekka
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Yimei Huang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Sonal Singh
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Chintan P Shah
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Robert F Cornell
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Preston Research Building, Nashville, TN, United States
| | - Rachid C Baz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Nida Waheed
- Department of Internal Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | | | - Lihui Yuan
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Taimour Langaee
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Keith March
- Division of Cardiovascular Medicine, Department of Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, United States
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, United States
| | - Jan S Moreb
- Novant Health Forsyth Medical Center, Hematology, Transplantation, and Cellular Therapy Division, Winston-Salem, NC, United States
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States.,UF Health Cancer Center, Gainesville, FL, United States.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, United States
| |
Collapse
|
16
|
Pediatric pulmonary hypertension: insulin-like growth factor-binding protein 2 is a novel marker associated with disease severity and survival. Pediatr Res 2020; 88:850-856. [PMID: 32927467 PMCID: PMC7704926 DOI: 10.1038/s41390-020-01113-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/07/2020] [Accepted: 07/26/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Insulin-like growth factors (IGFs), and their binding proteins (IGFBPs), play a significant role in cardiovascular function and may influence the pathobiology of PAH. We determined the diagnostic and prognostic value of IGF1 and IGFBP2 in pediatric PAH. METHODS Serum was analyzed by ELISA for IGF1 and IGFBP2 in pediatric PAH subjects from the NHLBI PAH Biobank (PAHB, n = 175) and a cohort of asthmatic subjects (n = 46, age 0-21 years) as a chronic pediatric pulmonary disease control. Biomarkers were analyzed with demographic and clinical variables for PAH severity. RESULTS Serum IGF1 was significantly lower in PAH compared to controls, while IGFBP2 was elevated in PAH subjects compared to controls. In the PAHB, IGF1 was negatively associated with mPAP and PVR, while IGFBP2 was positively associated with PVR and negatively associated with cardiac output and 6-min walk distance. Higher IGFBP2 levels were associated with use of prostacyclin therapy. IGFBP2 was associated with death, transplant, or palliative shunt with a Cox proportional hazard ratio of 8.8 (p < 0.001) but not IGF1 (p = 0.13). CONCLUSIONS Circulating IGFBP2 is a novel marker for pediatric PAH, which is associated with worse functional status, and survival. IGF axis dysregulation may be an important mechanistic target in pediatric pulmonary arterial hypertension. IMPACT Pediatric pulmonary hypertension is a severe disease, with poorly understood pathobiology. There are few studies looking at the pathobiology of pulmonary hypertension only in children. The IGF axis is dysregulated in pediatric pulmonary arterial hypertension. IGF axis dysregulation, with increased IGFBP2, is associated with worse clinical outcomes in pediatric pulmonary artery hypertension. IGF axis dysregulation gives new insight into the disease process and may be a mechanistic or therapeutic target.
Collapse
|
17
|
Girerd N, Bresso E, Devignes MD, Rossignol P. Insulin-like growth factor binding protein 2: A prognostic biomarker for heart failure hardly redundant with natriuretic peptides. Int J Cardiol 2020; 300:252-254. [PMID: 31761405 DOI: 10.1016/j.ijcard.2019.11.100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 11/26/2022]
Affiliation(s)
- Nicolas Girerd
- Université de Lorraine, Centre d'Investigations Cliniques Plurithématique 1433, INSERM 1116, CHRU de Nancy, France; F-CRIN INI-CRCT Cardiovascular and Renal Clinical Trialists Network, France.
| | - Emmanuel Bresso
- Université de Lorraine, LORIA, UMR 7503, Vandoeuvre-lès-Nancy, France
| | | | - Patrick Rossignol
- Université de Lorraine, Centre d'Investigations Cliniques Plurithématique 1433, INSERM 1116, CHRU de Nancy, France; F-CRIN INI-CRCT Cardiovascular and Renal Clinical Trialists Network, France
| |
Collapse
|
18
|
Watts EL, Perez‐Cornago A, Appleby PN, Albanes D, Ardanaz E, Black A, Bueno‐de‐Mesquita HB, Chan JM, Chen C, Chubb SP, Cook MB, Deschasaux M, Donovan JL, English DR, Flicker L, Freedman ND, Galan P, Giles GG, Giovannucci EL, Gunter MJ, Habel LA, Häggström C, Haiman C, Hamdy FC, Hercberg S, Holly JM, Huang J, Huang W, Johansson M, Kaaks R, Kubo T, Lane JA, Layne TM, Le Marchand L, Martin RM, Metter EJ, Mikami K, Milne RL, Morris HA, Mucci LA, Neal DE, Neuhouser ML, Oliver SE, Overvad K, Ozasa K, Pala V, Pernar CH, Pollak M, Rowlands M, Schaefer CA, Schenk JM, Stattin P, Tamakoshi A, Thysell E, Touvier M, Trichopoulou A, Tsilidis KK, Van Den Eeden SK, Weinstein SJ, Wilkens L, Yeap BB, Key TJ, Allen NE, Travis RC. The associations of anthropometric, behavioural and sociodemographic factors with circulating concentrations of IGF-I, IGF-II, IGFBP-1, IGFBP-2 and IGFBP-3 in a pooled analysis of 16,024 men from 22 studies. Int J Cancer 2019; 145:3244-3256. [PMID: 30873591 PMCID: PMC6745281 DOI: 10.1002/ijc.32276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/28/2019] [Accepted: 02/04/2019] [Indexed: 12/24/2022]
Abstract
Insulin-like growth factors (IGFs) and insulin-like growth factor binding proteins (IGFBPs) have been implicated in the aetiology of several cancers. To better understand whether anthropometric, behavioural and sociodemographic factors may play a role in cancer risk via IGF signalling, we examined the cross-sectional associations of these exposures with circulating concentrations of IGFs (IGF-I and IGF-II) and IGFBPs (IGFBP-1, IGFBP-2 and IGFBP-3). The Endogenous Hormones, Nutritional Biomarkers and Prostate Cancer Collaborative Group dataset includes individual participant data from 16,024 male controls (i.e. without prostate cancer) aged 22-89 years from 22 prospective studies. Geometric means of protein concentrations were estimated using analysis of variance, adjusted for relevant covariates. Older age was associated with higher concentrations of IGFBP-1 and IGFBP-2 and lower concentrations of IGF-I, IGF-II and IGFBP-3. Higher body mass index was associated with lower concentrations of IGFBP-1 and IGFBP-2. Taller height was associated with higher concentrations of IGF-I and IGFBP-3 and lower concentrations of IGFBP-1. Smokers had higher concentrations of IGFBP-1 and IGFBP-2 and lower concentrations of IGFBP-3 than nonsmokers. Higher alcohol consumption was associated with higher concentrations of IGF-II and lower concentrations of IGF-I and IGFBP-2. African Americans had lower concentrations of IGF-II, IGFBP-1, IGFBP-2 and IGFBP-3 and Hispanics had lower IGF-I, IGF-II and IGFBP-3 than non-Hispanic whites. These findings indicate that a range of anthropometric, behavioural and sociodemographic factors are associated with circulating concentrations of IGFs and IGFBPs in men, which will lead to a greater understanding of the mechanisms through which these factors influence cancer risk.
Collapse
Affiliation(s)
- Eleanor L. Watts
- Cancer Epidemiology UnitNuffield Department of Population Health, University of OxfordOxfordUnited Kingdom
| | - Aurora Perez‐Cornago
- Cancer Epidemiology UnitNuffield Department of Population Health, University of OxfordOxfordUnited Kingdom
| | - Paul N. Appleby
- Cancer Epidemiology UnitNuffield Department of Population Health, University of OxfordOxfordUnited Kingdom
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, Department of Health and Human ServicesNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Eva Ardanaz
- Navarra Public Health InstitutePamplonaSpain
| | - Amanda Black
- Division of Cancer Epidemiology and Genetics, Department of Health and Human ServicesNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - H. Bas Bueno‐de‐Mesquita
- Department for Determinants of Chronic DiseasesNational Institute for Public Health and the Environment (RIVM)BilthovenThe Netherlands
- Department of Gastroenterology and HepatologyUniversity Medical CentreUtrechtThe Netherlands
- Department of Epidemiology and BiostatisticsImperial College LondonLondonUnited Kingdom
- Department of Social & Preventive MedicineUniversity of MalayaKuala LumpurMalaysia
| | - June M. Chan
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCA
- Department UrologyUniversity of California‐San FranciscoSan FranciscoCA
| | - Chu Chen
- Public Health Sciences Division, Program in EpidemiologyFred Hutchinson Cancer Research CenterSeattleWA
| | - S.A. Paul Chubb
- PathWest Laboratory MedicineFiona Stanley HospitalPerthWAAustralia
- Medical SchoolUniversity of Western AustraliaPerthWAAustralia
| | - Michael B. Cook
- Division of Cancer Epidemiology and Genetics, Department of Health and Human ServicesNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Mélanie Deschasaux
- Sorbonne Paris Cité Epidemiology and Statistics Research Center (CRESS)Nutritional Epidemiology Research Team (EREN), Inserm U1153/Inra U1125/Cnam/Paris 13 UniversityParisFrance
| | - Jenny L. Donovan
- Department of Population Health SciencesBristol Medical School, University of BristolBristolUnited Kingdom
| | - Dallas R. English
- Cancer Epidemiology and Intelligence DivisionCancer Council VictoriaMelbourneVICAustralia
- Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global Health, The University of MelbourneMelbourneVICAustralia
| | - Leon Flicker
- Medical SchoolUniversity of Western AustraliaPerthWAAustralia
- WA Centre for Health & Ageing, Centre for Medical ResearchHarry Perkins Institute of Medical ResearchPerthWAAustralia
- Department of Geriatric MedicineRoyal Perth HospitalPerthWAAustralia
| | - Neal D. Freedman
- Division of Cancer Epidemiology and Genetics, Department of Health and Human ServicesNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Pilar Galan
- Sorbonne Paris Cité Epidemiology and Statistics Research Center (CRESS)Nutritional Epidemiology Research Team (EREN), Inserm U1153/Inra U1125/Cnam/Paris 13 UniversityParisFrance
| | - Graham G. Giles
- Cancer Epidemiology and Intelligence DivisionCancer Council VictoriaMelbourneVICAustralia
- Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global Health, The University of MelbourneMelbourneVICAustralia
| | - Edward L. Giovannucci
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMA
- Channing Division of Network MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
- Department of NutritionHarvard T.H. Chan School of Public HealthBostonMA
| | - Marc J. Gunter
- Section of Nutrition and MetabolismInternational Agency for Research on CancerLyonFrance
| | - Laurel A. Habel
- Division of ResearchKaiser Permanente Northern CaliforniaOaklandCA
| | | | | | - Freddie C. Hamdy
- Nuffield Department of SurgeryUniversity of OxfordOxfordUnited Kingdom
| | - Serge Hercberg
- Sorbonne Paris Cité Epidemiology and Statistics Research Center (CRESS)Nutritional Epidemiology Research Team (EREN), Inserm U1153/Inra U1125/Cnam/Paris 13 UniversityParisFrance
| | - Jeff M. Holly
- IGFs & Metabolic Endocrinology Group, Translational Health SciencesBristol Medical School, Faculty of Health Sciences, University of BristolBristolUnited Kingdom
| | - Jiaqi Huang
- Division of Cancer Epidemiology and Genetics, Department of Health and Human ServicesNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Wen‐Yi Huang
- Division of Cancer Epidemiology and Genetics, Department of Health and Human ServicesNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Mattias Johansson
- Genetic Epidemiology GroupInternational Agency for Research on CancerLyonFrance
| | - Rudolf Kaaks
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Tatsuhiko Kubo
- Department of Environmental EpidemiologyUniversity of Occupational and Environmental HealthKitakyushuJapan
| | - J. Athene Lane
- Department of Population Health SciencesBristol Medical School, University of BristolBristolUnited Kingdom
- National Institute for Health Research Bristol Biomedical Research Unit in NutritionBristolUnited Kingdom
| | | | | | - Richard M. Martin
- Department of Population Health SciencesBristol Medical School, University of BristolBristolUnited Kingdom
- National Institute for Health Research Bristol Biomedical Research Unit in NutritionBristolUnited Kingdom
- Medical Research Council/University of Bristol Integrative Epidemiology Unit, University of BristolBristolUnited Kingdom
| | - E. Jeffrey Metter
- Department of NeurologyUniversity of Tennessee Health Science CenterMemphisTN
| | | | - Roger L. Milne
- Cancer Epidemiology and Intelligence DivisionCancer Council VictoriaMelbourneVICAustralia
- Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global Health, The University of MelbourneMelbourneVICAustralia
| | | | - Lorelei A. Mucci
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMA
- Channing Division of Network MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
| | - David E. Neal
- Nuffield Department of SurgeryUniversity of OxfordOxfordUnited Kingdom
| | - Marian L. Neuhouser
- Cancer Prevention Program, Public Health Sciences DivisionFred Hutchinson Cancer Research CenterSeattleWA
| | - Steven E. Oliver
- Department of Health SciencesUniversity of York and the Hull York Medical SchoolYorkUK
| | - Kim Overvad
- Department of Public HealthSection for Epidemiology, Aarhus UniversityAarhusDenmark
| | - Kotaro Ozasa
- Radiation Effects Research FoundationHiroshimaJapan
| | - Valeria Pala
- Epidemiology and Prevention UnitFondazione IRCCS Istituto Nazionale dei Tumori di MilanoMilanItaly
| | - Claire H. Pernar
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMA
| | - Michael Pollak
- Department of Medicine and OncologyMcGill UniversityMontrealQCCanada
- Segal Cancer CentreJewish General HospitalMontrealQCCanada
| | - Mari‐Anne Rowlands
- Department of Population Health SciencesBristol Medical School, University of BristolBristolUnited Kingdom
| | | | - Jeannette M. Schenk
- Cancer Prevention Program, Public Health Sciences DivisionFred Hutchinson Cancer Research CenterSeattleWA
| | - Pär Stattin
- Department of Surgical SciencesUppsala UniversityUppsalaSweden
| | | | - Elin Thysell
- Department of Medical Biosciences and PathologyUmea UniversityUmeaSweden
| | - Mathilde Touvier
- Sorbonne Paris Cité Epidemiology and Statistics Research Center (CRESS)Nutritional Epidemiology Research Team (EREN), Inserm U1153/Inra U1125/Cnam/Paris 13 UniversityParisFrance
| | | | - Konstantinos K. Tsilidis
- Department of Epidemiology and BiostatisticsImperial College LondonLondonUnited Kingdom
- Department of Hygiene and Epidemiology, School of MedicineUniversity of IoanninaIoanninaGreece
| | | | - Stephanie J. Weinstein
- Division of Cancer Epidemiology and Genetics, Department of Health and Human ServicesNational Cancer Institute, National Institutes of HealthBethesdaMD
| | | | - Bu B. Yeap
- Medical SchoolUniversity of Western AustraliaPerthWAAustralia
- Department of Endocrinology and DiabetesFiona Stanley HospitalPerthWAAustralia
| | - Timothy J. Key
- Cancer Epidemiology UnitNuffield Department of Population Health, University of OxfordOxfordUnited Kingdom
| | - Naomi E. Allen
- Clinical Trial Service Unit and Epidemiological Studies UnitNuffield Department of Population Health, University of OxfordOxfordUnited Kingdom
| | - Ruth C. Travis
- Cancer Epidemiology UnitNuffield Department of Population Health, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
19
|
Barutaut M, Fournier P, Peacock WF, Evaristi MF, Caubère C, Turkieh A, Desmoulin F, Eurlings LWM, van Wijk S, Rocca HPBL, Butler J, Koukoui F, Dambrin C, Mazeres S, Le Page S, Delmas C, Galinier M, Jung C, Smih F, Rouet P. Insulin-like Growth Factor Binding Protein 2 predicts mortality risk in heart failure. Int J Cardiol 2019; 300:245-251. [PMID: 31806281 DOI: 10.1016/j.ijcard.2019.09.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 08/15/2019] [Accepted: 09/13/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Insulin-like Growth Factor Binding Protein 2 (IGFBP2) showed greater heart failure (HF) diagnostic accuracy than the "grey zone" B-type natriuretic peptides, and may have prognostic utility as well. OBJECTIVES To determine if IGFBP2 provides independent information on cardiovascular mortality in HF. METHODS A retrospective study of 870 HF patients from 3 independent international cohorts. Presentation IGFBP2 plasma levels were measured by ELISA, and patients were followed from 1 year (Maastricht, Netherlands) to 6 years (Atlanta, GA, USA and Toulouse, France). Multivariate analysis, Net Reclassification Improvement (NRI) and Integrated Discrimination Improvement (IDI) were performed in the 3 cohorts. The primary outcome was cardiovascular mortality. RESULTS In multivariate Cox proportional hazards analysis, the highest quartile of IGFBP2 was associated with mortality in the Maastricht cohort (adjusted hazard ratio 1.69 (95% CI, 1.18-2.41), p = 0.004) and in the combined Atlanta and Toulouse cohorts (adjusted hazard ratio 2.04 (95%CI, 1.3-3.3), p = 0.003). Adding IGFBP2 to a clinical model allowed a reclassification of adverse outcome risk in the Maastricht cohort (NRI = 18.7% p = 0.03; IDI = 3.9% p = 0.02) and with the Atlanta/Toulouse patients (NRI of 40.4% p = 0.01, 31,2% p = 0.04, 31.5% p = 0,02 and IDI of 2,9% p = 0,0005, 3.1% p = 0,0005 and 4,2%, p = 0.0005, for a follow-up of 1, 2 and 3 years, respectively). CONCLUSION In 3 international cohorts, IGFBP2 level is a strong prognostic factor for cardiovascular mortality in HF, adding information to natriuretic monitoring and usual clinical markers, that should be further prospectively evaluated for patients' optimized care.
Collapse
Affiliation(s)
- Manon Barutaut
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France
| | - Pauline Fournier
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France; University Hospital of Toulouse, Cardiology Department, F-31432 Toulouse, France
| | - William F Peacock
- Emergency Medicine at the Baylor College of Medicine in Houston, TX, USA
| | - Maria Francesca Evaristi
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France
| | - Céline Caubère
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France
| | - Annie Turkieh
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France
| | - Franck Desmoulin
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France
| | - Luc W M Eurlings
- Department of Cardiology, VieCuri Medical Center, Venlo, the Netherlands
| | - Sandra van Wijk
- Department of Cardiology, Maastricht University Medical Center, CARIM, Maastricht, the Netherlands
| | | | - Javed Butler
- Department of Medicine, Stony Brook University, NY, USA
| | - François Koukoui
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France
| | - Camille Dambrin
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France; University Hospital of Toulouse, Cardiology Department, F-31432 Toulouse, France
| | - Serge Mazeres
- Institute of Pharmacology and Structural Biology (IPBS), Toulouse, France
| | - Servane Le Page
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France
| | - Clement Delmas
- University Hospital of Toulouse, Cardiology Department, F-31432 Toulouse, France
| | - Michel Galinier
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France; University Hospital of Toulouse, Cardiology Department, F-31432 Toulouse, France
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Germany
| | - Fatima Smih
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France; Spartacus-Biomed, Clermont Le Fort, France
| | - Philippe Rouet
- LA Maison de la MItochondrie (LAMMI), Obesity and heart failure: molecular and clinical investigations, UMR CNRS 5288, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse cedex 4, France; INI-CRCT F-CRIN, GREAT Networks, France.
| |
Collapse
|
20
|
Zhang Z, Nkuipou‐Kenfack E, Staessen JA. Urinary Peptidomic Biomarker for Personalized Prevention and Treatment of Diastolic Left Ventricular Dysfunction. Proteomics Clin Appl 2019; 13:e1800174. [PMID: 30632674 PMCID: PMC6519355 DOI: 10.1002/prca.201800174] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/24/2018] [Indexed: 12/11/2022]
Abstract
Diastolic heart failure (DHF) is characterized by slow left ventricular (LV) relaxation, increased LV stiffness, interstitial deposition of collagen, and a modified extracellular matrix proteins. Among Europeans, the frequency of asymptomatic diastolic LV dysfunction (DD) is 25%. This constitutes a large pool of people at high risk of DHF. The goal of this review was to describe the discovery and the initial validation of new multidimensional urinary peptidomic biomarkers (UPB) indicative of DD, mainly consisting of collagen fragments, and to describe a roadmap for their introduction into clinical practice. The availability of new drugs creates a window of opportunity for mounting a randomized clinical trial consolidating the clinical applicability of UPB to screen for DD. If successfully completed, such trial will benefit ≈25% of all people older than 50 years and open a large market for a UPB diagnostic tool and the drug tested. Moreover, sequenced peptides making up UPB will generate novel insights in the pathophysiology of DD and facilitate personalized treatment of patients with DHF for whom prevention came too late. If proven cost-effective, the clinical application of UPB will contribute to the sustainability of health care in aging population in epidemiologic transition.
Collapse
Affiliation(s)
- Zhen‐Yu Zhang
- Studies Coordinating CentreResearch Unit Hypertension and Cardiovascular EpidemiologyKU Leuven Department of Cardiovascular SciencesUniversity of LeuvenLeuvenBelgium
| | | | - Jan A. Staessen
- Studies Coordinating CentreResearch Unit Hypertension and Cardiovascular EpidemiologyKU Leuven Department of Cardiovascular SciencesUniversity of LeuvenLeuvenBelgium
| |
Collapse
|
21
|
|
22
|
Olszanecka A, Dragan A, Kawecka-Jaszcz K, Fedak D, Czarnecka D. Relationships of insulin-like growth factor-1, its binding proteins, and cardiometabolic risk in hypertensive perimenopausal women. Metabolism 2017; 69:96-106. [PMID: 28285656 DOI: 10.1016/j.metabol.2017.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 01/04/2023]
Abstract
PURPOSE During the transition from premenopause to postmenopause, many women experience weight gain and central fat deposition; therefore, we hypothesized that circulating growth factors can play a role in the pathogenesis of hypertension, metabolic syndrome, and subclinical organ damage in perimenopausal women. BASIC PROCEDURES The study included 192 women aged 40 to 60years; 152 had newly diagnosed essential hypertension that had never been treated, and 40 were normotensive age-matched controls. For all subjects, 24-h ambulatory blood pressure monitoring (ABPM), echocardiographic examination with assessment of left ventricular mass (LVM) and systolic and diastolic functions (GE Vivid 7.0, General Electric Vingmed Ultrasound, Horten, Norway), carotid ultrasound with measurement of intima-media thickness, and carotid-femoral pulse wave velocity (PWV) measurement (SphygmoCor, AtCor Medical, Sydney, Australia) were performed. Serum levels of insulin-like growth factor 1 (IGF-1), insulin-like growth factor-binding protein 2 (IGFBP-2), and insulin-like growth factor-binding protein 3 (IGFBP-3) were measured using an immunochemical assay. MAIN FINDINGS Hypertensive women had significantly lower IGFBP-2 levels than did normotensive controls (162.9±83.7 vs. 273.1±103.0μg/L, p<0.001); the groups did not differ regarding IGF and IGFBP-3 concentrations. After adjusting the covariates, multivariate analysis showed that IGFBP2 was significantly negatively correlated with 24-h systolic blood pressure (β=-0.31, p=0.02). The adjusted odds ratio for hypertension per standard deviation decrease in IGFBP-2 was 3.43 (95% confidence interval [CI] 1.65-7.13). IGFBP-2 showed a negative correlation with the number of metabolic syndrome components. Independent of body composition, IGFBP-2 was significantly related to left ventricular relative wall thickness and the ratio of mitral inflow velocities as parameter of diastolic function. PRINCIPAL CONCLUSIONS In perimenopausal women, decreased IGFBP-2 levels may play a role in blood pressure regulation and the development of subclinical left ventricular diastolic dysfunction. Whether IGFBP-2 is a marker or a mediator of cardiovascular disease in this population merits further investigation.
Collapse
Affiliation(s)
- Agnieszka Olszanecka
- 1st Department of Cardiology, Interventional Electrocardiology, and Hypertension, Jagiellonian University Medical College, Krakow, Poland.
| | - Aneta Dragan
- 1st Department of Cardiology, Interventional Electrocardiology, and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Kalina Kawecka-Jaszcz
- 1st Department of Cardiology, Interventional Electrocardiology, and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Danuta Fedak
- Chair of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Danuta Czarnecka
- 1st Department of Cardiology, Interventional Electrocardiology, and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|