1
|
Zhao L, Zhang X, Birmann BM, Danford CJ, Lai M, Simon TG, Chan AT, Giovannucci EL, Ngo L, Libermann TA, Zhang X. Pre-diagnostic plasma inflammatory proteins and risk of hepatocellular carcinoma in three population-based cohort studies from the United States and the United Kingdom. Int J Cancer 2024; 155:1593-1603. [PMID: 38861327 PMCID: PMC11537828 DOI: 10.1002/ijc.35054] [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: 11/10/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024]
Abstract
Previous studies suggest a role for inflammation in hepatocarcinogenesis. However, no study has comprehensively evaluated associations between circulating inflammatory proteins and risk of hepatocellular carcinoma (HCC) among the general population. We conducted a nested case-control study in the Nurses' Health Study (NHS) and the Health Professionals Follow-up Study (HPFS) with 56 pairs of incident HCC cases and controls. External validation was performed in the UK Biobank (34 HCC cases and 48,471 non-HCC controls). Inflammatory protein levels were measured in pre-diagnostic plasma using the Olink® Inflammation Panel. We used conditional logistic regression to calculate multivariable odds ratios (ORs) with 95% confidence intervals (CIs) for associations between a 1-standard deviation (SD) increase in biomarker levels and HCC risk, considering a statistically significant threshold of false discovery rate (FDR)-adjusted p < .05. In the NHS/HPFS, among 70 analyzed proteins with call rates >80%, 15 proteins had significant associations with HCC risk (pFDR < .05). Two proteins (stem cell factor, OR per SD = 0.31, 95% CI = 0.16-0.58; tumor necrosis factor superfamily member 12, OR per SD = 0.51, 95% CI = 0.31-0.85) were inversely associated whereas 13 proteins were positively associated with risk of HCC; positive ORs per SD ranged from 1.73 for interleukin (IL)-10 to 2.35 for C-C motif chemokine-19. A total of 11 proteins were further replicated in the UK Biobank. Seven of the eight selected positively associated proteins also showed positive associations with HCC risk by enzyme-linked immunosorbent assay, with ORs ranging from 1.56 for IL-10 to 2.72 for hepatocyte growth factor. More studies are warranted to further investigate the roles of these observed inflammatory proteins in HCC etiology, early detection, risk stratification, and disease treatment.
Collapse
Affiliation(s)
- Longgang Zhao
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Xinyuan Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brenda M. Birmann
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Michelle Lai
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Tracey G. Simon
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew T. Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Edward L. Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Long Ngo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Towia A. Libermann
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Yale University School of Nursing, Orange, Connecticut, USA
| |
Collapse
|
2
|
Chen Y, Zhong A. Causal effects of inflammatory cytokines on cardiovascular diseases: Insights from genetic evidence. Heliyon 2024; 10:e35447. [PMID: 39165962 PMCID: PMC11334864 DOI: 10.1016/j.heliyon.2024.e35447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Background The causal relationship between inflammatory cytokines and cardiovascular diseases (CVDs) has not been fully elucidated. Exploring this relationship between circulating inflammatory cytokines and CVDs is crucial for early clinical diagnosis and effective treatment. Methods and Results This study investigated the causal relationships between 41 inflammatory cytokines and six CVDs: heart failure (HF), myocardial infarction (MI), unstable angina pectoris (UAP), stable angina pectoris (SAP), valvular heart disease (VHD), and aortic aneurysm (AA), using the Mendelian Randomization (MR) method. The primary analysis employed the inverse-variance weighted (IVW) method within MR. Heterogeneity and pleiotropy were assessed through MR-Egger regression and the Q statistic. Strong evidence supported the effect of macrophage inflammatory protein-1β (MIP-1β) on MI (OR = 1.062, P < 0.001, FDR <0.001). Suggestive evidence showed that the Beta nerve growth factor increased the risk of MI (OR = 1.145, P = 0.025), but the stem cell factor (SCF) demonstrated a potential protective effect against MI (OR = 0.910, P = 0.04). SCF and hepatocyte growth factor (HGF) exhibited potential protective effects against SAP. No inflammatory cytokine was associated with UAP. Monocyte chemotactic protein-1 was linked to an increased risk of VHD (OR = 1.056, P = 0.049). Higher levels of interleukin-13 (IL-13), interferon gamma-induced protein 10 (IP-10), and growth-regulated oncogene-alpha were associated with increased susceptibility to HF. Elevated basic fibroblast growth factor (bFGF) levels exhibited protective effects against AA (OR = 0.751, P = 0.038). Reverse MR analyses revealed that AA significantly decreased circulating TNF-related apoptosis-inducing ligand (TRAIL) levels (OR = 0.907, P < 0.001, FDR = 0.01). MI significantly increased circulating IL-12-p70 levels (OR = 1.146, P < 0.001, FDR = 0.014). Suggestive evidence indicated the Causal effects of six CVDs on 17 circulating inflammatory cytokines. Conclusions This study clarified the causal relationships between specific inflammatory cytokines and six CVDs, providing novel insights and evidence into the genetic involvement of inflammatory cytokines in CVDs. These inflammatory cytokines may be potential biomarkers for early disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Yuxiu Chen
- Department of Emergency Medicine, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Aifang Zhong
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| |
Collapse
|
3
|
Chen Y, Gialeli C, Shen J, Dunér P, Walse B, Duelli A, Caing-Carlsson R, Blom AM, Zibert JR, Nilsson AH, Alenfall J, Liang C, Nilsson J. Identification of an osteopontin-derived peptide that binds neuropilin-1 and activates vascular repair responses and angiogenesis. Pharmacol Res 2024; 205:107259. [PMID: 38871237 DOI: 10.1016/j.phrs.2024.107259] [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: 12/28/2023] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
The osteopontin-derived peptide FOL-005 stimulates hair growth. Using ligand-receptor glyco-capture technology we identified neuropilin-1 (NRP-1), a known co-receptor for vascular endothelial growth factor (VEGF) receptors, as the most probable receptor for FOL-005 and the more stable analogue FOL-026. X-ray diffraction and microscale thermophoresis analysis revealed that FOL-026 shares binding site with VEGF in the NRP-1 b1-subdomain. Stimulation of human umbilical vein endothelial cells with FOL-026 resulted in phosphorylation of VEGFR-2, ERK1/2 and AKT, increased cell growth and migration, stimulation of endothelial tube formation and inhibition of apoptosis in vitro. FOL-026 also promoted angiogenesis in vivo as assessed by subcutaneous Matrigel plug and hind limb ischemia models. NRP-1 knock-down or treatment of NRP-1 antagonist EG00229 blocked the stimulatory effects of FOL-026 on endothelial cells. Exposure of human coronary artery smooth muscle cells to FOL-026 stimulated cell growth, migration, inhibited apoptosis, and induced VEGF gene expression and VEGFR-2/AKT phosphorylation by an NRP-1-dependent mechanism. RNA sequencing showed that FOL-026 activated pathways involved in tissue repair. These findings identify NRP-1 as the receptor for FOL-026 and show that its biological effects mimic that of growth factors binding to the VEGF receptor family. They also suggest that FOL-026 may have therapeutical potential in conditions that require vascular repair and/or enhanced angiogenesis.
Collapse
Affiliation(s)
- Yihong Chen
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, Shanghai, China; Department of Experimental Medical Science, Lund University, Sweden
| | | | - Junyan Shen
- Department of Experimental Medical Science, Lund University, Sweden; Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Pontus Dunér
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | | | | | | | - Anna M Blom
- Department of Translational Medicine, Lund University, Sweden
| | | | | | - Jan Alenfall
- Department of Clinical Sciences Malmö, Lund University, Sweden; Coegin Pharma AB, Lund, Sweden
| | - Chun Liang
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, Shanghai, China.
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Sweden.
| |
Collapse
|
4
|
Zelicha H, Kaplan A, Yaskolka Meir A, Rinott E, Tsaban G, Blüher M, Klöting N, Ceglarek U, Isermann B, Stumvoll M, Chassidim Y, Shelef I, Hu FB, Shai I. Altered proteome profiles related to visceral adiposity may mediate the favorable effect of green Mediterranean diet: the DIRECT-PLUS trial. Obesity (Silver Spring) 2024; 32:1245-1256. [PMID: 38757229 DOI: 10.1002/oby.24036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE The objective of this study was to explore the effects of a green Mediterranean (green-MED) diet, which is high in dietary polyphenols and green plant-based protein and low in red/processed meat, on cardiovascular disease and inflammation-related circulating proteins and their associations with cardiometabolic risk parameters. METHODS In the 18-month weight loss trial Dietary Intervention Randomized Controlled Trial Polyphenols Unprocessed Study (DIRECT-PLUS), 294 participants with abdominal obesity were randomized to basic healthy dietary guidelines, Mediterranean (MED), or green-MED diets. Both isocaloric MED diet groups consumed walnuts (28 g/day), and the green-MED diet group also consumed green tea (3-4 cups/day) and green shakes (Mankai plant shake, 500 mL/day) and avoided red/processed meat. Proteome panels were measured at three time points using Olink CVDII. RESULTS At baseline, a dominant protein cluster was significantly related to higher phenotypic cardiometabolic risk parameters, with the strongest associations attributed to magnetic resonance imaging-assessed visceral adiposity (false discovery rate of 5%). Overall, after 6 months of intervention, both the MED and green-MED diets induced improvements in cardiovascular disease and proinflammatory risk proteins (p < 0.05, vs. healthy dietary guidelines), with the green-MED diet leading to more pronounced beneficial changes, largely driven by dominant proinflammatory proteins (IL-1 receptor antagonist protein, IL-16, IL-18, thrombospondin-2, leptin, prostasin, galectin-9, and fibroblast growth factor 21; adjusted for age, sex, and weight loss; p < 0.05). After 18 months, proteomics cluster changes presented the strongest correlations with visceral adiposity reduction. CONCLUSIONS Proteomics clusters may enhance our understanding of the favorable effect of a green-MED diet that is enriched with polyphenols and low in red/processed meat on visceral adiposity and cardiometabolic risk.
Collapse
Affiliation(s)
- Hila Zelicha
- The Health and Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Alon Kaplan
- The Health and Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Anat Yaskolka Meir
- The Health and Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Ehud Rinott
- The Health and Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Gal Tsaban
- The Health and Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Berend Isermann
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | | | - Yoash Chassidim
- Department of Engineering, Sapir Academic College, Sapir, Israel
| | - Ilan Shelef
- Soroka University Medical Center, Be'er Sheva, Israel
| | - Frank B Hu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Harvard Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Iris Shai
- The Health and Nutrition Innovative International Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Liu J, Ying J, Hu T. Genetic effects of inflammatory cytokines on coronary artery disease and myocardial infarction and the mediating roles of lipid traits. Postgrad Med J 2024; 100:461-468. [PMID: 38409767 DOI: 10.1093/postmj/qgae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/27/2023] [Accepted: 01/19/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Chronic inflammation has been connected by epidemiological evidence to coronary artery disease (CAD) along with myocardial infarction (MI). Nevertheless, it is still unclear whether reverse causality or confounders account for these connections. Our objectives are to examine the causality between inflammatory cytokines and CAD/MI as well as the potential mediating influence of lipid characteristics. METHODS We acquired instrumental variables through genome-wide association studies meta-analyses of 41 inflammatory cytokines (8293 individuals). Genetic associations with CAD (122 733 cases and 424 528 controls), MI (~61 505 cases and 577 716 controls) and five candidate lipid mediators were obtained from the corresponding genome-wide association studies. A two-step, two-sample Mendelian randomization analysis was applied, followed with comprehensive sensitivity analyses. RESULTS Genetically determined growth regulated oncogene-α was causally linked to a decreased incidence of CAD [odds ratio (OR), 0.97; 95% confidence interval (CI), 0.95-0.99; P = .007] and MI (OR, 0.95; 95% CI, 0.92-0.98; P = .002). There is suggestive evidence indicating a causal impact of macrophage inflammatory protein-1β upon CAD (OR, 1.04; 95% CI, 1.01-1.07; P = .010) and MI (OR, 1.07; 95% CI, 1.02-1.11; P = .002). Furthermore, we discovered suggestive causal connections between tumor necrosis factor-related apoptosis-inducing ligand and CAD (OR, 0.97; 95% CI, 0.95-1.00; P = .020). Two-step Mendelian randomization analysis revealed that triglycerides partially mediate the effect of growth regulated oncogene-α on CAD (proportion-mediated: 13.28%) and MI (8.05%). CONCLUSIONS We provided novel genetic evidence supporting the causality of inflammatory cytokines on CAD/MI and elucidate the mediating effect of triglycerides in the causal pathways linking inflammatory cytokines and CAD/MI.
Collapse
Affiliation(s)
- Junsong Liu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, School of Medicine, Ningbo University, Ningbo, Zhejiang 315010, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Ningbo, Zhejiang 315010, China
| | - Jiajun Ying
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, School of Medicine, Ningbo University, Ningbo, Zhejiang 315010, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Ningbo, Zhejiang 315010, China
| | - Teng Hu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, School of Medicine, Ningbo University, Ningbo, Zhejiang 315010, China
- Cardiovascular Disease Clinical Medical Research Center of Ningbo, Ningbo, Zhejiang 315010, China
| |
Collapse
|
6
|
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
|
7
|
Li H, Wang Y, Sonestedt E, Borné Y. Associations of ultra-processed food consumption, circulating protein biomarkers, and risk of cardiovascular disease. BMC Med 2023; 21:415. [PMID: 37919714 PMCID: PMC10623817 DOI: 10.1186/s12916-023-03111-2] [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: 06/18/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND We aim to examine the association between ultra-processed foods (UPF) consumption and cardiovascular disease (CVD) risk and to identify plasma proteins associated with UPF. METHODS This prospective cohort study included 26,369 participants from the Swedish Malmö Diet and Cancer Study, established in 1991-1996. Dietary intake was assessed using a modified diet history method, and UPF consumption was estimated using the NOVA classification system. A total of 88 selected CVD-related proteins were measured among 4475 subjects. Incident CVD (coronary heart disease and ischemic stroke) was defined as a hospital admission or death through registers. Cox proportional hazards regression models were performed to analyze the associations of UPF intake with risks of CVD. Linear regression models were used to identify the plasma proteins associated with UPF intake. RESULTS During 24.6 years of median follow-up, 6236 participants developed CVD, of whom 3566 developed coronary heart disease and 3272 developed ischemic stroke. The adjusted hazard ratio (95% confidence interval) in the 4th versus 1st quartile of UPF was 1.18 (1.08, 1.29) for CVD, 1.20 (1.07, 1.35) for coronary heart disease, and 1.17 (1.03, 1.32) for ischemic stroke. Plasma proteins interleukin 18, tumor necrosis factor receptor 2, macrophage colony-stimulating factor 1, thrombomodulin, tumor necrosis factor receptor 1, hepatocyte growth factor, stem cell factor, resistin, C-C motif chemokine 3, and endothelial cell-specific molecule 1 were positively associated with UPF after correcting for multiple testing. CONCLUSIONS Our study showed that high UPF intake increased the risk of CVD and was associated with several protein biomarkers. Future studies are warranted to validate these findings and assess the potential pathways between UPF intake and CVD.
Collapse
Affiliation(s)
- Huiping Li
- School of Medicine, Northwest University, Taibai North Road, Beilin District, Xi'an, 710069, China.
- School of Public Health, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China.
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms Gata 35, 21428, Malmö, Sweden.
| | - Yaogang Wang
- School of Public Health, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Emily Sonestedt
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms Gata 35, 21428, Malmö, Sweden
| | - Yan Borné
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms Gata 35, 21428, Malmö, Sweden.
| |
Collapse
|
8
|
Wu Y, Pu X, Wu S, Zhang Y, Fu S, Tang H, Wang X, Xu M. PCIF1, the only methyltransferase of N6,2-O-dimethyladenosine. Cancer Cell Int 2023; 23:226. [PMID: 37779183 PMCID: PMC10544176 DOI: 10.1186/s12935-023-03066-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023] Open
Abstract
N6-methyladenosine(m6A), is the most abundant post-transcriptional modification of mRNA in biology. When the first nucleotide after the m7G cap is adenosine, it is methylated at the N6 position to form N6,2-O-dimethyladenosine (m6Am). m6Am is a reversible modification located at the first transcribed nucleotide, which is present in about 30% of cellular mRNAs, thus m6Am can have a significant impact on gene expression in the transcriptome. Phosphorylated CTD interaction factor 1(PCIF1), the unique and specific methyltransferase of m6Am, has been shown to affect mRNA stability, transcription, and translation. Several studies have shown that PCIF1 is clearly associated with tumor, viral, and endocrine diseases. Moreover, PCIF1 may be related to the tumor microenvironment, immune cell typing, and programmed cell death protein 1(PD-1) drug resistance. Here, we summarize the mechanism of PCIF1 involvement in mRNA modifications, and outline m6Am modifications and diseases in which PCIF1 is involved. We also summarized the role of PCIF1 in immune and immune checkpoint blockade(ICB) treatment, and predicted the possibility of PCIF1 as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yuting Wu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Xi Pu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Sihui Wu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Yiran Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Shengqiao Fu
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Haowen Tang
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Xu Wang
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
- Digestive Disease Research Institute of Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
| |
Collapse
|
9
|
Wei T, Zhu Z, Liu L, Liu B, Wu M, Zhang W, Cui Q, Liu F, Zhang R. Circulating levels of cytokines and risk of cardiovascular disease: a Mendelian randomization study. Front Immunol 2023; 14:1175421. [PMID: 37304261 PMCID: PMC10247976 DOI: 10.3389/fimmu.2023.1175421] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Background Epidemiological studies have linked various circulating cytokines to cardiovascular disease (CVD), which however remains uncertain whether these relationships represent causality or are due to bias. To address this question, we conducted a Mendelian randomization (MR) analysis to systematically investigate the causal effects of circulating cytokine levels on CVD development. Methods This study leveraged the summary statistic from respective genome-wide association study (GWAS) of 47 cytokines and four types of CVD. The cis-quantitative trait locus (cis-QTL) definition, derived from a GWAS meta-analysis comprising 31,112 participants of European descent, served as instruments for cytokines. A two-sample MR design was employed, followed by comprehensive sensitivity analyses to validate the robustness of results. Results The results of inverse-variance weighted method using cis-protein QTL (cis-pQTL) instruments, showed the causal effects of four cytokines (i.e., IL-1ra, MCSF, SeSelectin, SCF) on the risk of coronary artery disease (CAD). We also identified causal relationships between two cytokines (i.e., IL-2ra, IP-10) and heart failure (HF), as well as two cytokines (i.e., MCP-3, SeSelectin) and atrial fibrillation (AF), after controlling for false discovery rate (FDR). The use of cis-expression QTL (cis-eQTL) revealed additional causal associations between IL-1a, MIF and CAD, between IL-6, MIF, and HF, as well as between FGFBasic and AF. No significant sign was survived for stroke with FDR applied. Results were largely consistent across sensitivity analyses. Conclusion The present study provides supportive evidence that genetic predisposition to levels of certain cytokines causally affects the development of specific type of CVD. These findings have important implications for the creation of novel therapeutic strategies targeting these cytokines as a means of preventing and treating CVD.
Collapse
Affiliation(s)
- Tao Wei
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Zhanfang Zhu
- Department of General Internal Medicine, Xi’an Jiaotong University Hospital, Xi’an, China
| | - Lin Liu
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Bo Liu
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Min Wu
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Wei Zhang
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Qianwei Cui
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Fuqiang Liu
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Ronghuai Zhang
- Department of Cardiology, Shaanxi Provincial People’s Hospital, Xi’an, China
| |
Collapse
|
10
|
Fuselli A, de Los Milagros Bürgi M, Kratje R, Prieto C. Generation and functional evaluation of novel monoclonal antibodies targeting glycosylated human stem cell factor. Appl Microbiol Biotechnol 2022; 106:8121-8137. [PMID: 36401641 DOI: 10.1007/s00253-022-12282-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 11/21/2022]
Abstract
Human stem cell factor (hSCF) is an early-acting growth factor that promotes proliferation, differentiation, migration, and survival in several tissues. It plays a crucial role in hematopoiesis, gametogenesis, melanogenesis, intestinal motility, and in development and recovery of nervous and cardiovascular systems. Potential therapeutic applications comprise anemia treatment, mobilization of hematopoietic stem/progenitor cells to peripheral blood, and increasing gene transduction efficiency for gene therapy. Developing new tools to characterize recombinant hSCF in most native-like form as possible is crucial to understand the complexity of its in vivo functions and for improving its biotechnological applications. The soluble domain of hSCF was expressed in HEK293 cells. Highly purified rhSCF showed great molecular mass variability due to the presence of N- and O-linked carbohydrates, and it presented a 2.5-fold increase on proliferative activity compared to bacteria-derived hSCF. Three hybridoma clones producing monoclonal antibodies (mAbs) with high specificity for the glycoprotein were obtained. 1C4 and 2D3 mAbs were able to detect bacteria-derived and glycosylated rhSCF and demonstrated to be excellent candidates to develop a sandwich ELISA assay for rhSCF quantification, with detection limits of 0.18 and 0.07 ng/ml, respectively. Interestingly, 1A10 mAb only recognized glycosylated rhSCF, suggesting that sugar moieties might be involved in epitope recognition. 1A10 mAb showed the highest binding affinity, and it constituted the best candidate for immunodetection of the entire set rhSCF glycoforms in western blot assays, and for intracellular cytokine staining. Our work shows that combining glycosylated rhSCF expression with hybridoma technology is a powerful strategy to obtain specific suitable immunochemical assays and thus improve glycoprotein-producing bioprocesses. KEY POINTS: • Soluble glycosylated human SCF exerted improved proliferative activity on UT-7 cells. • Three mAbs with high specificity targeting glycosylated human SCF were obtained. • mAbs applications comprise sandwich ELISA, western blot, and immunofluorescence assays.
Collapse
Affiliation(s)
- Antonela Fuselli
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina
| | - María de Los Milagros Bürgi
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina
| | - Ricardo Kratje
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Cell Culture Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina
| | - Claudio Prieto
- UNL, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Biotechnological Development Laboratory, Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242 (S3000ZAA), Santa Fe, Argentina.
- Cellargen Biotech S.R.L., Antonia Godoy 6369 (S3000ZAA), Santa Fe, Argentina.
| |
Collapse
|
11
|
Rossignol P, Duarte K, Bresso E, A Å, Devignes MD, Eriksson N, Girerd N, Glerup R, Jardine AG, Holdaas H, Lamiral Z, Leroy C, Massy Z, März W, Krämer B, Wu PH, Schmieder R, Soveri I, Christensen JH, Svensson M, Zannad F, Fellström B. NT-proBNP and stem cell factor plasma concentrations are independently associated with cardiovascular outcomes in end-stage renal disease hemodialysis patients. EUROPEAN HEART JOURNAL OPEN 2022; 2:oeac069. [PMID: 36600882 PMCID: PMC9797490 DOI: 10.1093/ehjopen/oeac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/14/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Aims End-stage renal disease (ESRD) treated by chronic hemodialysis (HD) is associated with poor cardiovascular (CV) outcomes, with no available evidence-based therapeutics. A multiplexed proteomic approach may identify new pathophysiological pathways associated with CV outcomes, potentially actionable for precision medicine. Methods and results The AURORA trial was an international, multicentre, randomized, double-blind trial involving 2776 patients undergoing maintenance HD. Rosuvastatin vs. placebo had no significant effect on the composite primary endpoint of death from CV causes, nonfatal myocardial infarction or nonfatal stroke. We first compared CV risk-matched cases and controls (n = 410) to identify novel biomarkers using a multiplex proximity extension immunoassay (276 proteomic biomarkers assessed with OlinkTM). We replicated our findings in 200 unmatched cases and 200 controls. External validation was conducted from a multicentre real-life Danish cohort [Aarhus-Aalborg (AA), n = 331 patients] in which 92 OlinkTM biomarkers were assessed. In AURORA, only N-terminal pro-brain natriuretic peptide (NT-proBNP, positive association) and stem cell factor (SCF) (negative association) were found consistently associated with the trial's primary outcome across exploration and replication phases, independently from the baseline characteristics. Stem cell factor displayed a lower added predictive ability compared with NT-ProBNP. In the AA cohort, in multivariable analyses, BNP was found significantly associated with major CV events, while higher SCF was associated with less frequent CV deaths. Conclusions Our findings suggest that NT-proBNP and SCF may help identify ESRD patients with respectively high and low CV risk, beyond classical clinical predictors and also point at novel pathways for prevention and treatment.
Collapse
Affiliation(s)
- P Rossignol
- Corresponding author. Tel: +33383157322, Fax: +33383157324,
| | - K Duarte
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - E Bresso
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France,LORIA (CNRS, Inria NGE, Université de Lorraine), F-CRIN INI-CRCT, Vandœuvre-lès-Nancy, France
| | - Åsberg A
- Department of Transplantation Medicine Oslo University Hospital–Rikshospitalet, Oslo, Norway,Norway and Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - M D Devignes
- LORIA (CNRS, Inria NGE, Université de Lorraine), F-CRIN INI-CRCT, Vandœuvre-lès-Nancy, France
| | - N Eriksson
- UCR Uppsala Clinical Research Center, Uppsala Science Park, Uppsala, Sweden
| | - N Girerd
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - R Glerup
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark
| | - A G Jardine
- Renal Research Group, British Heart Foundation Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Z Lamiral
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - C Leroy
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - Z Massy
- CESP, Center for Research in Epidemiology and Population Health, University Paris-Saclay, University Paris-Sud, UVSQ, Villejuif, France,Division of Nephrology, Ambroise Paré University Hospital, APHP, Boulogne, Billancourt and FCRIN INI-CRCT, Paris, France
| | - W März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria,Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - B Krämer
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - P H Wu
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden,Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - R Schmieder
- Department of Nephrology and Hypertension, University Hospital Erlangen, Erlangen, Germany
| | - I Soveri
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - J H Christensen
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark
| | - M Svensson
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | | | | |
Collapse
|
12
|
Pandhi P, Ter Maaten JM, Anker SD, Ng LL, Metra M, Samani NJ, Lang CC, Dickstein K, de Boer RA, van Veldhuisen DJ, Voors AA, Sama IE. Pathophysiologic Processes and Novel Biomarkers Associated With Congestion in Heart Failure. JACC. HEART FAILURE 2022; 10:623-632. [PMID: 36049813 DOI: 10.1016/j.jchf.2022.05.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Congestion is the main driver behind symptoms of heart failure (HF), but pathophysiology related to congestion remains poorly understood. OBJECTIVES Using pathway and differential expression analyses, the authors aim to identify biological processes and biomarkers associated with congestion in HF. METHODS A congestion score (sum of jugular venous pressure, orthopnea, and peripheral edema) was calculated in 1,245 BIOSTAT-CHF patients with acute or worsening HF. Patients with a score ranking in the bottom or top categories of congestion were deemed noncongested (n = 408) and severely congested (n = 142), respectively. Plasma concentrations of 363 unique proteins (Olink Proteomics Multiplex CVD-II, CVD-III, Immune Response and Oncology II panels) were compared between noncongested and severely congested patients. Results were validated in an independent validation cohort of 1,342 HF patients (436 noncongested and 232 severely congested). RESULTS Differential protein expression analysis showed 107/363 up-regulated and 6/363 down-regulated proteins in patients with congestion compared with those without. FGF-23, FGF-21, CA-125, soluble ST2, GDF-15, FABP4, IL-6, and BNP were the strongest up-regulated proteins (fold change [FC] >1.30, false discovery rate [FDR], P < 0.05). KITLG, EGF, and PON3 were the strongest down-regulated proteins (FC <-1.30, FDR P < 0.05). Pathways most prominently involved in congestion were related to inflammation, endothelial activation, and response to mechanical stimulus. The validation cohort yielded similar findings. CONCLUSIONS Severe congestion in HF is mainly associated with inflammation, endothelial activation, and mechanical stress. Whether these pathways play a causal role in the onset or progression of congestion remains to be established. The identified biomarkers may become useful for diagnosing and monitoring congestion status.
Collapse
Affiliation(s)
- Paloma Pandhi
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jozine M Ter Maaten
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stefan D Anker
- Department of Cardiology, Charité Universitätsmedizin, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, German Centre for Cardiovascular Research, Charité Universitätsmedizin, Berlin, Germany
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom; NIHR Leicester Biomedical Research Centre, Leicester, United Kingdom
| | - Marco Metra
- Institute of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom; NIHR Leicester Biomedical Research Centre, Leicester, United Kingdom
| | - Chim C Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom; Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Kenneth Dickstein
- University of Bergen, Bergen, Norway; Stavanger University Hospital, Stavanger, Norway
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Iziah E Sama
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
13
|
Karason K, Girerd N, Andersson-Asssarsson J, Duarte K, Taube M, Svensson PA, Huby AC, Peltonen M, Carlsson LM, Zannad F. Heart failure in obesity: insights from proteomics in patients treated with or without weight-loss surgery. Int J Obes (Lond) 2022; 46:2088-2094. [PMID: 35945262 DOI: 10.1038/s41366-022-01194-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Obesity is associated with incident heart failure (HF), but the underlying mechanisms are unclear. METHODS We performed a nested case-control study within the Swedish-Obese-Subjects study, by identifying 411 cases who developed HF and matched them with respect to age, sex, weight-loss-surgery and length of follow-up with 410 controls who did not develop HF. In analyses corrected for multiple testing, we studied 182 plasma proteins known to be related to cardiovascular disease to investigate whether they could add to the understanding of the processes underlying obesity-related HF. RESULTS A total of 821 subjects were followed for 16 ± 6 years. Multivariable analysis adjusted for matching variables revealed that 32 proteins were significantly associated with HF. Twelve proteins were related to HF ≥ 80% of the time using a bootstrap resampling approach (false-discovery-rate [FDR] < 0.05): 11 were associated with increased HF-risk: TNFRSF10A*, ST6GAL1, PRCP, MMP12, TIMP1, CCL3, QPCT, ANG, C1QTNF1, SERPINA5 and GAL-9; and one was related to reduced HF-risk: LPL. An further 20 proteins were associated with onset of HF 50-80% of the time using bootstrap resampling (FDR < 0.05). A pathway analysis including all significant 32 proteins suggested that these biomarkers were related to inflammation, matrix remodeling, cardiometabolic hormones and hemostasis. Three proteins, C1QTNF1, FGF-21 and CST3, reflecting dyslipidemia and kidney disease, displayed a higher association with HF in patients who did not undergo weight-loss-surgery and maintained with obesity. CONCLUSION Pathways associated with HF in obesity include inflammation, matrix remodeling, cardiometabolic hormones and hemostasis; three protein biomarkers predicting HF appeared to be obesity-specific.
Collapse
Affiliation(s)
- Kristjan Karason
- Department of Cardiology and Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden. .,Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Nicolas Girerd
- Centre d'Investigation Clinique 1433 module Plurithématique, CHRU Nancy-Hopitaux de Brabois, Institut Lorrain du Coeur et des Vaisseaux Louis Mathieu, Vandoeuvre les Nancy, France
| | - Johanna Andersson-Asssarsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kevin Duarte
- Centre d'Investigation Clinique 1433 module Plurithématique, CHRU Nancy-Hopitaux de Brabois, Institut Lorrain du Coeur et des Vaisseaux Louis Mathieu, Vandoeuvre les Nancy, France
| | - Magdalena Taube
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per-Arne Svensson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Institute of Health and Care Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anne-Cecile Huby
- Centre d'Investigation Clinique 1433 module Plurithématique, CHRU Nancy-Hopitaux de Brabois, Institut Lorrain du Coeur et des Vaisseaux Louis Mathieu, Vandoeuvre les Nancy, France
| | - Markku Peltonen
- National Institute for Health and Welfare, Helsinki, Finland
| | - Lena M Carlsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Faiez Zannad
- Centre d'Investigation Clinique 1433 module Plurithématique, CHRU Nancy-Hopitaux de Brabois, Institut Lorrain du Coeur et des Vaisseaux Louis Mathieu, Vandoeuvre les Nancy, France
| |
Collapse
|
14
|
Stanne TM, Angerfors A, Andersson B, Brännmark C, Holmegaard L, Jern C. Longitudinal Study Reveals Long-Term Proinflammatory Proteomic Signature After Ischemic Stroke Across Subtypes. Stroke 2022; 53:2847-2858. [PMID: 35686557 PMCID: PMC9389938 DOI: 10.1161/strokeaha.121.038349] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inflammation contributes both to the pathogenesis of stroke and the response to brain injury. We aimed to identify proteins reflecting the acute-phase response and proteins more likely to reflect proinflammatory processes present before stroke by broadly profiling inflammation-related plasma proteins in a longitudinal ischemic stroke study.
Collapse
Affiliation(s)
- Tara M Stanne
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Sweden (T.M.S., A.A., C.B., C.J.)
| | - Annelie Angerfors
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Sweden (T.M.S., A.A., C.B., C.J.)
| | - Björn Andersson
- Bioinformatics Core Facility, University of Gothenburg, Sweden (B.A.)
| | - Cecilia Brännmark
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Sweden (T.M.S., A.A., C.B., C.J.)
| | - Lukas Holmegaard
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience, the Sahlgrenska Academy, University of Gothenburg, Sweden (L.H.).,Department of Neurology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden. (L.H.)
| | - Christina Jern
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Sweden (T.M.S., A.A., C.B., C.J.).,Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden. (C.J.)
| |
Collapse
|
15
|
Ponce-de-Leon M, Linseisen J, Peters A, Linkohr B, Heier M, Grallert H, Schöttker B, Trares K, Bhardwaj M, Gào X, Brenner H, Kamiński KA, Paniczko M, Kowalska I, Baumeister SE, Meisinger C. Novel associations between inflammation-related proteins and adiposity: A targeted proteomics approach across four population-based studies. Transl Res 2022; 242:93-104. [PMID: 34780968 DOI: 10.1016/j.trsl.2021.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 11/27/2022]
Abstract
Chronic low-grade inflammation has been proposed as a linking mechanism between obesity and the development of inflammation-related conditions such as insulin resistance and cardiovascular disease. Despite major advances in the last 2 decades, the complex relationship between inflammation and obesity remains poorly understood. Therefore, we aimed to identify novel inflammation-related proteins associated with adiposity. We investigated the association between BMI and waist circumference and 72 circulating inflammation-related proteins, measured using the Proximity Extension Assay (Olink Proteomics), in 3,308 participants of four independent European population-based studies (KORA-Fit, BVSII, ESTHER, and Bialystok PLUS). In addition, we used body fat mass measurements obtained by Dual-energy X-ray absorptiometry (DXA) in the Bialystok PLUS study to further validate our results and to explore the relationship between inflammation-related proteins and body fat distribution. We found 14 proteins associated with at least one measure of adiposity across all four studies, including four proteins for which the association is novel: DNER, SLAMF1, RANKL, and CSF-1. We confirmed previously reported associations with CCL19, CCL28, FGF-21, HGF, IL-10RB, IL-18, IL-18R1, IL-6, SCF, and VEGF-A. The majority of the identified inflammation-related proteins were associated with visceral fat as well as with the accumulation of adipose tissue in the abdomen and the trunk. In conclusion, our study provides new insights into the immune dysregulation observed in obesity that might help uncover pathophysiological mechanisms of disease development.
Collapse
Affiliation(s)
- Mariana Ponce-de-Leon
- Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany; Chair of Epidemiology, University of Augsburg, University Hospital Augsburg, Augsburg, Germany; Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Jakob Linseisen
- Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany; Chair of Epidemiology, University of Augsburg, University Hospital Augsburg, Augsburg, Germany; Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Birgit Linkohr
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany; KORA Study Centre, University Hospital Augsburg, Augsburg, Germany
| | - Harald Grallert
- Institute of Epidemiology, Helmholtz Zentrum Munich, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Kira Trares
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Megha Bhardwaj
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany
| | - Xīn Gào
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany
| | - Herman Brenner
- Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center, Heidelberg, Germany; Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Karol Adam Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Marlena Paniczko
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, Białystok, Poland
| | | | - Christa Meisinger
- Institute for Medical Information Processing, Biometry, and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany; Chair of Epidemiology, University of Augsburg, University Hospital Augsburg, Augsburg, Germany; Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
16
|
Wu PH, Glerup RI, Svensson MHS, Eriksson N, Christensen JH, de Laval P, Soveri I, Westerlund M, Linde T, Ljunggren Ö, Fellström B. Novel Biomarkers Detected by Proteomics Predict Death and Cardiovascular Events in Hemodialysis Patients. Biomedicines 2022; 10:biomedicines10040740. [PMID: 35453489 PMCID: PMC9026983 DOI: 10.3390/biomedicines10040740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/06/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
End-stage kidney disease increases mortality and the risk of cardiovascular (CV) disease. It is crucial to explore novel biomarkers to predict CV disease in the complex setting of patients receiving hemodialysis (HD). This study investigated the association between 92 targeted proteins with all-cause death, CV death, and composite vascular events (CVEs) in HD patients. From December 2010 to March 2011, 331 HD patients were included and followed prospectively for 5 years. Serum was analyzed for 92 CV-related proteins using Proseek Multiplex Cardiovascular I panel, a high-sensitivity assay based on proximity extension assay (PEA) technology. The association between biomarkers and all-cause death, CV death, and CVEs was evaluated using Cox-regression analyses. Of the PEA-based proteins, we identified 20 proteins associated with risk of all-cause death, 7 proteins associated with risk of CV death, and 17 proteins associated with risk of CVEs, independent of established risk factors. Interleukin-8 (IL-8), T-cell immunoglobulin and mucin domain 1 (TIM-1), and C-C motif chemokine 20 (CCL20) were associated with increased risk of all-cause death, CV death, and CVE in multivariable-adjusted models. Stem cell factor (SCF) and Galanin peptides (GAL) were associated with both decreased risk of all-cause death and CV death. In conclusion, IL-8, TIM-1, and CCL20 predicted death and CV outcomes in HD patients. Novel findings were that SCF and GAL were associated with a lower risk of all-cause death and CV death. The SCF warrants further study with regard to its possible biological effect in HD patients.
Collapse
Affiliation(s)
- Ping-Hsun Wu
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Rie Io Glerup
- Department of Nephrology, Aalborg University Hospital, 9000 Aalborg, Denmark; (R.I.G.); (J.H.C.)
| | - My Hanna Sofia Svensson
- Division of Medicine, Department of Nephrology, Akershus University Hospital, 1478 Oslo, Norway;
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, 75185 Uppsala, Sweden;
| | | | - Philip de Laval
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Inga Soveri
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Magnus Westerlund
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Torbjörn Linde
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Bengt Fellström
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Correspondence: ; Tel.: +46-18-6114348
| |
Collapse
|
17
|
Lopez-Perez D, Redruello-Romero A, Garcia-Rubio J, Arana C, Garcia-Escudero LA, Tamayo F, Salmeron J, Galvez J, Leon J, Carazo Á. In Obese Patients With Type 2 Diabetes, Mast Cells in Omental Adipose Tissue Decrease the Surface Expression of CD45, CD117, CD203c, and FcϵRI. Front Endocrinol (Lausanne) 2022; 13:818388. [PMID: 35370964 PMCID: PMC8965342 DOI: 10.3389/fendo.2022.818388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022] Open
Abstract
The paradigm of mast cells in type 2 diabetes is changing. Although they were first considered deleterious inflammatory cells, now they seem to be important players driving adipose tissue homeostasis. Here we have employed a flow cytometry-based approach for measuring the surface expression of 4 proteins (CD45, CD117, CD203c, and FcϵRI) on mast cells of omental (o-WAT) and subcutaneous white adipose tissue (s-WAT) in a cohort of 96 patients with morbid obesity. The cohort was split into three groups: non-T2D, pre-T2D, and T2D. Noteworthy, patients with T2D have a mild condition (HbA1c <7%). In o-WAT, mast cells of patients with T2D have a decrease in the surface expression of CD45 (p=0.0013), CD117 (p=0.0066), CD203c (p=0.0025), and FcϵRI (p=0.043). Besides, in s-WAT, the decrease was seen only in CD117 (p=0.046). These results indicate that T2D affects more to mast cells in o-WAT than in s-WAT. The decrease in these four proteins has serious effects on mast cell function. CD117 is critical for mast cell survival, while CD45 and FcϵRI are important for mast cell activation. Additionally, CD203c is only present on the cell surface after granule release. Taking together these observations, we suggest that mast cells in o-WAT of patients with T2D have a decreased survival, activation capacity, and secretory function.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Anaïs Redruello-Romero
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | | | - Carlos Arana
- Endocrinology and nutrition department, Virgen de la Luz University Hospital, Cuenca, Spain
| | - Luis A. Garcia-Escudero
- Department of Statistics and Operative Research, Faculty of Sciences, University of Valladolid, Valladolid, Spain
| | | | - Javier Salmeron
- Gastroenterology Unit, San Cecilio University Hospital, Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica En Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, Granada, Spain
- *Correspondence: Julio Galvez, ; Ángel Carazo,
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Granada, Spain
| | - Ángel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Granada, Spain
- *Correspondence: Julio Galvez, ; Ángel Carazo,
| |
Collapse
|
18
|
Schmidt IM, Sarvode Mothi S, Wilson PC, Palsson R, Srivastava A, Onul IF, Kibbelaar ZA, Zhuo M, Amodu A, Stillman IE, Rennke HG, Humphreys BD, Waikar SS. Circulating Plasma Biomarkers in Biopsy-Confirmed Kidney Disease. Clin J Am Soc Nephrol 2022; 17:27-37. [PMID: 34759008 PMCID: PMC8763150 DOI: 10.2215/cjn.09380721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/02/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Biomarkers for noninvasive assessment of histopathology and prognosis are needed in patients with kidney disease. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Using a proteomics assay, we measured a multimarker panel of 225 circulating plasma proteins in a prospective cohort study of 549 individuals with biopsy-confirmed kidney diseases and semiquantitative assessment of histopathology. We tested the associations of each biomarker with histopathologic lesions and the risks of kidney disease progression (defined as ≥40% decline in eGFR or initiation of KRT) and death. RESULTS After multivariable adjustment and correction for multiple testing, 46 different proteins were associated with histopathologic lesions. The top-performing markers positively associated with acute tubular injury and interstitial fibrosis/tubular atrophy were kidney injury molecule-1 (KIM-1) and V-set and Ig domain-containing protein 2 (VSIG2), respectively. Thirty proteins were significantly associated with kidney disease progression, and 35 were significantly associated with death. The top-performing markers for kidney disease progression were placental growth factor (hazard ratio per doubling, 5.4; 95% confidence interval, 3.4 to 8.7) and BMP and activin membrane-bound inhibitor (hazard ratio, 3.0; 95% confidence interval, 2.1 to 4.2); the top-performing markers for death were TNF-related apoptosis-inducing ligand receptor-2 (hazard ratio, 2.9; 95% confidence interval, 2.0 to 4.0) and CUB domain-containing protein-1 (hazard ratio, 2.4; 95% confidence interval, 1.8 to 3.3). CONCLUSION We identified several plasma protein biomarkers associated with kidney disease histopathology and adverse clinical outcomes in individuals with a diverse set of kidney diseases. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2021_12_28_CJN09380721.mp3.
Collapse
Affiliation(s)
- Insa M. Schmidt
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Suraj Sarvode Mothi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Parker C. Wilson
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Ragnar Palsson
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Anand Srivastava
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ingrid F. Onul
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Zoe A. Kibbelaar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Min Zhuo
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Afolarin Amodu
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Isaac E. Stillman
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Helmut G. Rennke
- Department of Pathology, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University, St. Louis, Missouri
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
19
|
Koshiol J, Argirion I, Liu Z, Kim Lam T, O'Brien TR, Yu K, McGlynn KA, Petrick JL, Pinto L, Chen CJ, Hildesheim A, Pfeiffer RM, Lee MH, Yang HI. Immunologic markers and risk of hepatocellular carcinoma in hepatitis B virus- and hepatitis C virus-infected individuals. Aliment Pharmacol Ther 2021; 54:833-842. [PMID: 34286851 DOI: 10.1111/apt.16524] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/09/2021] [Accepted: 06/25/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Clinical and experimental studies suggest immunologic proteins contribute to hepatocellular carcinoma (HCC) development. AIM To evaluate circulating immunologic markers and HCC risk. METHODS From a Taiwanese cohort of chronically hepatitis B virus (HBV)-infected individuals followed over time (REVEAL-HBV), we sampled 175 who developed HCC, 117 cirrhosis only, and 165 non-cirrhotic controls. From a similar Taiwanese cohort of chronically hepatitis C virus (HCV)-infected individuals (REVEAL-HCV), we included 94 individuals who developed HCC, 68 cirrhosis only and 100 non-cirrhotic controls. We compared pre-diagnostic plasma levels of 102 markers in HCC cases to non-cirrhotic and cirrhotic controls using polytomous logistic regression. A priori markers included insulin-like growth factor binding protein-3 (IGFBP-3), intercellular adhesion molecule 1 (ICAM-1) and interleukin 6 (IL-6). P-values for other markers were corrected for multiple testing (false discovery rate = 10%). RESULTS In both REVEAL-HBV and REVEAL-HCV, increasing levels of ICAM-1 were associated with increased risk of HCC compared to non-cirrhotic controls (P-trend 0.02 and 0.001, respectively). In both REVEAL-HBV and REVEAL-HCV, two novel markers [C-X-C motif chemokine 11 (CXCL11) and hepatocyte growth factor (HGF)] were positively associated [strongest odds ratioquartile 4 versus 1 (OR) 4.55 for HGF in HCV], while two [complement factor H related 5 (CFHR5) and stem cell factor (SCF)] were negatively associated (strongest ORQ4vQ1 0.14 for SCF in HCV) with development of HCC compared to non-cirrhotic controls. CONCLUSIONS We confirmed the association for ICAM-1 and identified 4 additional proteins associated with HBV- and HCV-related HCC. These findings highlight the importance of immunologic processes in HBV- and HCV-related HCC.
Collapse
Affiliation(s)
- Jill Koshiol
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ilona Argirion
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Zhiwei Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Tram Kim Lam
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Thomas R O'Brien
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kelly Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Katherine A McGlynn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jessica L Petrick
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.,School of Medicine, Slone Epidemiology Center, Boston University, Boston, MA, USA
| | - Ligia Pinto
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos, Biomedical Research, Inc, Frederick, MD, USA
| | - Chien-Jen Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Epidemiology and Preventative Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mei-Hsuan Lee
- National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hwai-I Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
20
|
Pan J, Borné Y, Orho-Melander M, Nilsson J, Melander O, Engström G. The associations between red cell distribution width and plasma proteins in a general population. Clin Proteomics 2021; 18:12. [PMID: 33781199 PMCID: PMC8008679 DOI: 10.1186/s12014-021-09319-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/12/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND High red cell distribution width (RDW) has been increasingly recognized as a risk factor for cardiovascular diseases (CVDs), but the underlying mechanisms remain unknown. Our aim was to explore the associations between RDW and plasma proteins implicated in the pathogenesis of CVD using a targeted proteomics panel. METHODS RDW and 88 plasma proteins were measured in a population-based cohort study (n = 4726), Malmö Diet and Cancer-Cardiovascular Cohort (MDC-CC). A random 2/3 of the cohort was used as discovery sample and remaining 1/3 was used for replication. Multiple linear regression was used to assess the associations between RDW and plasma proteins, with adjustments for age, sex, and other potential confounders. Proteins with Bonferroni-corrected significant associations with RDW in the discovery sub-cohort were validated in the replication cohort. RESULTS Thirteen of 88 plasma proteins had significant associations with RDW in the discovery sample, after multivariate adjustments. Eleven of them were also significant in the replication sample, including SIR2-like protein 2 (SIRT2), stem cell factor (SCF, inversely), melusin (ITGB1BP2), growth differentiation factor-15 (GDF-15), matrix metalloproteinase-7 (MMP-7), hepatocyte growth factor (HGF), chitinase-3-like protein 1 (CHI3L1), interleukin-8 (IL-8), CD40 ligand (CD40-L), urokinase plasminogen activator surface receptor (U-PAR) and matrix metalloproteinase-3 (MMP-3). CONCLUSIONS Several proteins from this targeted proteomics panel were associated with RDW in this cohort. These proteins could potentially be linked to the increased cardiovascular risk in individuals with high RDW.
Collapse
Affiliation(s)
- Jingxue Pan
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden.
| | - Yan Borné
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| |
Collapse
|
21
|
Drake I, Hindy G, Almgren P, Engström G, Nilsson J, Melander O, Orho-Melander M. Methodological considerations for identifying multiple plasma proteins associated with all-cause mortality in a population-based prospective cohort. Sci Rep 2021; 11:6734. [PMID: 33762603 PMCID: PMC7990913 DOI: 10.1038/s41598-021-85991-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 03/08/2021] [Indexed: 11/09/2022] Open
Abstract
Novel methods to characterize the plasma proteome has made it possible to examine a wide range of proteins in large longitudinal cohort studies, but the complexity of the human proteome makes it difficult to identify robust protein-disease associations. Nevertheless, identification of individuals at high risk of early mortality is a central issue in clinical decision making and novel biomarkers may be useful to improve risk stratification. With adjustment for established risk factors, we examined the associations between 138 plasma proteins measured using two proximity extension assays and long-term risk of all-cause mortality in 3,918 participants of the population-based Malmö Diet and Cancer Study. To examine the reproducibility of protein-mortality associations we used a two-step random-split approach to simulate a discovery and replication cohort and conducted analyses using four different methods: Cox regression, stepwise Cox regression, Lasso-Cox regression, and random survival forest (RSF). In the total study population, we identified eight proteins that associated with all-cause mortality after adjustment for established risk factors and with Bonferroni correction for multiple testing. In the two-step analyses, the number of proteins selected for model inclusion in both random samples ranged from 6 to 21 depending on the method used. However, only three proteins were consistently included in both samples across all four methods (growth/differentiation factor-15 (GDF-15), N-terminal pro-B-type natriuretic peptide, and epididymal secretory protein E4). Using the total study population, the C-statistic for a model including established risk factors was 0.7222 and increased to 0.7284 with inclusion of the most predictive protein (GDF-15; P < 0.0001). All multiple protein models showed additional improvement in the C-statistic compared to the single protein model (all P < 0.0001). We identified several plasma proteins associated with increased risk of all-cause mortality independently of established risk factors. Further investigation into the putatively causal role of these proteins for longevity is needed. In addition, the examined methods for identifying multiple proteins showed tendencies for overfitting by including several putatively false positive findings. Thus, the reproducibility of findings using such approaches may be limited.
Collapse
Affiliation(s)
- Isabel Drake
- Diabetes and Cardiovascular Disease-Genetic Epidemiology, Department of Clinical Sciences in Malmö, Lund University, Clinical Research Centre House 60 Floor 13, Jan Waldenströms gata 35, 205 02, Malmö, Sweden.
| | - George Hindy
- Diabetes and Cardiovascular Disease-Genetic Epidemiology, Department of Clinical Sciences in Malmö, Lund University, Clinical Research Centre House 60 Floor 13, Jan Waldenströms gata 35, 205 02, Malmö, Sweden.,Department of Population Medicine, College of Medicine Qatar University, Doha, Qatar
| | - Peter Almgren
- Diabetes and Cardiovascular Disease-Genetic Epidemiology, Department of Clinical Sciences in Malmö, Lund University, Clinical Research Centre House 60 Floor 13, Jan Waldenströms gata 35, 205 02, Malmö, Sweden.,Hypertension and Cardiovascular Disease, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Gunnar Engström
- Cardiovascular Epidemiology, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Jan Nilsson
- Experimental Cardiovascular Research, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Olle Melander
- Hypertension and Cardiovascular Disease, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Marju Orho-Melander
- Diabetes and Cardiovascular Disease-Genetic Epidemiology, Department of Clinical Sciences in Malmö, Lund University, Clinical Research Centre House 60 Floor 13, Jan Waldenströms gata 35, 205 02, Malmö, Sweden
| |
Collapse
|
22
|
Hahn J, Leatherwood C, Malspeis S, Liu X, Lu B, Roberts AL, Sparks JA, Karlson EW, Feldman CH, Munroe ME, James JA, Kubzansky LD, Costenbader KH. Associations between daily alcohol consumption and systemic lupus erythematosus-related cytokines and chemokines among US female nurses without SLE. Lupus 2020; 29:976-982. [PMID: 32507075 PMCID: PMC7497791 DOI: 10.1177/0961203320929427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES Moderate alcohol consumption has been associated with decreased systemic lupus erythematosus risk, but the biologic basis for this association is unknown. We aimed to determine whether moderate alcohol consumption was associated with lower concentrations of systemic lupus erythematosus-associated chemokines/cytokines in an ongoing cohort of female nurses without systemic lupus erythematosus, and whether the association was modified by the presence of systemic lupus erythematosus-related autoantibodies. METHODS About 25% of participants from the Nurses' Health Study (n = 121,700 women) and Nurses' Health Study 2 (n = 116,429) donated a blood sample; of these, 1177 women were without systemic lupus erythematosus at time of donation. Cumulative average and current (within 4 years) intakes of beer, wine or liquor were assessed from pre-blood draw questionnaires. Chemokine/cytokine concentrations (stem cell factor, B-lymphocyte stimulator, interferon-inducible protein-10, interferon-alpha, interleukin-10) and antibodies against dsDNA and extractable nuclear antigens were obtained using enzyme-linked immunosorbent assays. Antinuclear antibodies were detected by indirect immunofluorescence on HEp-2 cells. RESULTS At blood draw, the women's mean age was 56 years and 22% were antinuclear antibody positive; 36% were African-American. About half (46%) reported consuming 0-5 g/day of alcohol. Stem cell factor levels were 0.5% lower (p < 0.0001) for every gram per day increase in cumulative average alcohol consumption. Women who consumed >5 g/day had mean stem cell factor levels 7% lower (p = 0.002) than non-drinkers. Other cytokines were not significantly associated with alcohol intake. Autoantibody status did not modify observed associations. CONCLUSION In this study of female nurses, moderate alcohol consumption was associated with lower stem cell factor levels, suggesting a plausible mechanism through which alcohol may lower systemic lupus erythematosus risk might be by decreasing circulating stem cell factor.
Collapse
Affiliation(s)
- Jill Hahn
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | | | - Susan Malspeis
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Xinyi Liu
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Bing Lu
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Andrea L. Roberts
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Jeffrey A. Sparks
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Elizabeth W. Karlson
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Candace H. Feldman
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Melissa E. Munroe
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Judith A. James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Laura D. Kubzansky
- Department of Social and Behavioral Sciences, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Karen H. Costenbader
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
23
|
Ahmed S, Ahmed A, Säleby J, Bouzina H, Lundgren J, Rådegran G. Elevated plasma tyrosine kinases VEGF-D and HER4 in heart failure patients decrease after heart transplantation in association with improved haemodynamics. Heart Vessels 2020; 35:786-799. [PMID: 31960146 PMCID: PMC7198637 DOI: 10.1007/s00380-019-01548-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/13/2019] [Indexed: 12/14/2022]
Abstract
Receptor tyrosine kinases (RTKs) are implicated in cardiovascular growth and remodelling. We aimed to identify the plasma levels of RTKs and related proteins and their association with haemodynamic alterations in heart failure (HF) and related pulmonary hypertension (PH) following heart transplantation (HT). Using proximity extension assay, 28 RTKs and related proteins were analysed in plasma from 20 healthy controls and 26 HF patients before and 1-year after HT. In end-stage HF, out of 28 RTKs, plasma vascular endothelial growth factor-D (VEGF-D) and human epidermal growth factor-4 (HER4) were elevated compared to controls (p < 0.001), but decreased (p < 0.0001) and normalised after HT. Following HT, plasma changes (Δ) of VEGF-D correlated with Δmean pulmonary artery pressure (rs = 0.65, p = 0.00049), Δpulmonary artery wedge pressure (rs = 0.72, p < 0.0001), Δpulmonary arterial compliance (PAC) (rs = - 0.52, p = 0.0083) and Δpulmonary vascular resistance (PVR) (rs = 0.58, p = 0.0032). ΔHER4 correlated with Δmean right atrial pressure (rs = 0.51, p = 0.012), ΔNT-proBNP (rs = 0.48, p = 0.016) and Δcardiac index (rs = - 0.56, p = 0.0044). In HF patients following HT, normalisation of VEGF-D reflected reversal of passive pulmonary congestion and restored PAC and PVR; whereas the normalisation of HER4 reflected decreased volume overload and improved cardiac function. The precise function of these proteins, their potential clinical use and pathophysiological relation in HF and related PH remain to be elucidated.
Collapse
Affiliation(s)
- Salaheldin Ahmed
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden.
- The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Getingevägen 4, EA15, 22185, Lund, Sweden.
| | - Abdulla Ahmed
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Getingevägen 4, EA15, 22185, Lund, Sweden
| | - Joanna Säleby
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Getingevägen 4, EA15, 22185, Lund, Sweden
| | - Habib Bouzina
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Getingevägen 4, EA15, 22185, Lund, Sweden
| | - Jakob Lundgren
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Getingevägen 4, EA15, 22185, Lund, Sweden
| | - Göran Rådegran
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Haemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Getingevägen 4, EA15, 22185, Lund, Sweden
| |
Collapse
|
24
|
Plasma protein biomarkers and their association with mutually exclusive cardiovascular phenotypes: the FIBRO-TARGETS case-control analyses. Clin Res Cardiol 2019; 109:22-33. [PMID: 31062082 DOI: 10.1007/s00392-019-01480-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/12/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hypertension, obesity and diabetes are major and potentially modifiable "risk factors" for cardiovascular diseases. Identification of biomarkers specific to these risk factors may help understanding the underlying pathophysiological pathways, and developing individual treatment. METHODS The FIBRO-TARGETS (targeting cardiac fibrosis for heart failure treatment) consortium has merged data from 12 patient cohorts in 1 common database of > 12,000 patients. Three mutually exclusive main phenotypic groups were identified ("cases"): (1) "hypertensive"; (2) "obese"; and (3) "diabetic"; age-sex matched in a 1:2 proportion with "healthy controls" without any of these phenotypes. Proteomic associations were studied using a biostatistical method based on LASSO and confronted with machine-learning and complex network approaches. RESULTS The case:control distribution by each cardiovascular phenotype was hypertension (50:100), obesity (50:98), and diabetes (36:72). Of the 86 studied proteins, 4 were found to be independently associated with hypertension: GDF-15, LEP, SORT-1 and FABP-2; 3 with obesity: CEACAM-8, LEP and PRELP; and 4 with diabetes: GDF-15, REN, CXCL-1 and SCF. GDF-15 (hypertension + diabetes) and LEP (hypertension + obesity) are shared by 2 different phenotypes. A machine-learning approach confirmed GDF-15, LEP and SORT-1 as discriminant biomarkers for the hypertension group, and LEP plus PRELP for the obesity group. Complex network analyses provided insight on the mechanisms underlying these disease phenotypes where fibrosis may play a central role. CONCLUSION Patients with "mutually exclusive" phenotypes display distinct bioprofiles that might underpin different biological pathways, potentially leading to fibrosis. Plasma protein biomarkers and their association with mutually exclusive cardiovascular phenotypes: the FIBRO-TARGETS case-control analyses. Patients with "mutually exclusive" phenotypes (blue: obesity, hypertension and diabetes) display distinct protein bioprofiles (green: decreased expression; red: increased expression) that might underpin different biological pathways (orange arrow), potentially leading to fibrosis.
Collapse
|
25
|
Bouzina H, Rådegran G. Low plasma stem cell factor combined with high transforming growth factor-α identifies high-risk patients in pulmonary arterial hypertension. ERJ Open Res 2018; 4:00035-2018. [PMID: 30443557 PMCID: PMC6230818 DOI: 10.1183/23120541.00035-2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/17/2018] [Indexed: 12/29/2022] Open
Abstract
In pulmonary arterial hypertension (PAH), severe vasoconstriction and remodelling of small pulmonary arteries result in high mortality. Receptor tyrosine kinases and their ligands, such as transforming growth factor (TGF)-α, modulate proliferation in PAH. Although the receptor tyrosine kinase c-Kit has been shown to be overexpressed in PAH, the expression and role of its ligand stem cell factor (SCF) remain unknown. However, low plasma SCF levels are known to be linked to higher cardiovascular mortality risk. Using proximity extension assays, we measured SCF and TGF-α in venous plasma from treatment-naïve PAH patients and healthy controls. Patients were stratified into risk classes based on PAH guidelines. Plasma SCF was decreased (p=0.013) and TGF-α was increased (p<0.0001) in PAH patients compared to controls. SCF correlated to pulmonary vascular resistance (r=−0.66, p<0.0001), cardiac index (r=0.66, p<0.0001), venous oxygen saturation (r=0.47, p<0.0008), mean right atrial pressure (r=−0.44, p<0.002) and N-terminal pro-brain natriuretic protein (r=−0.39, p<0.006). SCF was lower in “high-risk” compared to “intermediate-risk” (p=0.0015) or “low-risk” (p=0.0009) PAH patients. SCF and TGF-α levels combined (SCF/TGF-α) resulted in 85.7% sensitivity and 81.5% specificity for detecting high-risk patients (p<0.0001). Finally, REVEAL (Registry to Evaluate Early and Long-Term Pulmonary Arterial Hypertension Disease Management) risk scores in PAH patients correlated to SCF/TGF-α levels (r=−0.50, p=0.0003). In conclusion, low plasma SCF combined with high TGF-α identifies high-risk PAH patients at baseline. Lower circulating SCF levels, which are associated with worse haemodynamics, may be related to the c-Kit accumulation previously observed in PAH. Plasma stem cell factor and transforming growth factor-α are related to pulmonary arterial hypertension severity and risk assessmenthttp://ow.ly/s5O330lDjN9
Collapse
Affiliation(s)
- Habib Bouzina
- Lund University, Dept of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund, Sweden.,The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Göran Rådegran
- Lund University, Dept of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund, Sweden.,The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
26
|
Hyperglycemia does not affect tissue repair responses in shear stress-induced atherosclerotic plaques in ApoE-/- mice. Sci Rep 2018; 8:7530. [PMID: 29760458 PMCID: PMC5951920 DOI: 10.1038/s41598-018-25942-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022] Open
Abstract
The mechanisms responsible for macrovascular complications in diabetes remain to be fully understood. Recent studies have identified impaired vascular repair as a possible cause of plaque vulnerability in diabetes. This notion is supported by observations of a reduced content of fibrous proteins and smooth muscle cell mitogens in carotid endarterectomy from diabetic patients along with findings of decreased circulating levels of endothelial progenitor cells. In the present study we used a diabetic mouse model to characterize how hyperglycemia affects arterial repair responses. We induced atherosclerotic plaque formation in ApoE-deficient (ApoE−/−) and heterozygous glucokinase knockout ApoE-deficient mice (ApoE−/− GK+/−) mice with a shear stress-modifying cast. There were no differences in cholesterol or triglyceride levels between the ApoE−/− and ApoE−/− GK+/− mice. Hyperglycemia did not affect the size of the formed atherosclerotic plaques, and no effects were seen on activation of cell proliferation, smooth muscle cell content or on the expression and localization of collagen, elastin and several other extracellular matrix proteins. The present study demonstrates that hyperglycemia per se has no significant effects on tissue repair processes in injured mouse carotid arteries, suggesting that other mechanisms are involved in diabetic plaque vulnerability.
Collapse
|