1
|
Patterson BK, Yogendra R, Francisco EB, Guevara-Coto J, Long E, Pise A, Osgood E, Bream J, Kreimer M, Jeffers D, Beaty C, Vander Heide R, Mora-Rodríguez RA. Detection of S1 spike protein in CD16+ monocytes up to 245 days in SARS-CoV-2-negative post-COVID-19 vaccine syndrome (PCVS) individuals. Hum Vaccin Immunother 2025; 21:2494934. [PMID: 40358138 PMCID: PMC12077440 DOI: 10.1080/21645515.2025.2494934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Despite over 13 billion SARS-CoV-2 vaccine doses administered globally, persistent post-vaccination symptoms, termed post-COVID-19 vaccine syndrome (PCVS), resemble post-acute sequelae of COVID-19 (PASC). Symptoms like cardiac, vascular, and neurological issues often emerge shortly after vaccination and persist for months to years, mirroring PASC. We previously showed the S1 subunit of the SARS-CoV-2 spike protein persists in CD16+ monocytes after infection, potentially driving PASC. Approved vaccines (Pfizer, Moderna, Janssen, AstraZeneca) deliver synthetic S1 to elicit immunity, suggesting a shared mechanism. We hypothesized that vaccine-derived S1 persistence in CD16+ monocytes sustains inflammation akin to PASC, contributing to PCVS. We studied 50 individuals with PCVS symptoms lasting over 30 days post-vaccination and 26 asymptomatic controls, using (1) machine learning-based immune profiling to compare cytokine signatures with PASC, (2) flow cytometry to detect S1 in CD16+ monocytes, and (3) LC-MS to confirm S1 across vaccine types. We correlated S1 persistence with symptom duration and inflammation. Prior infection was excluded via clinical history, anti-nucleocapsid antibody tests, and T-detect assays, though definitive tests are lacking. Preliminary findings suggest S1 persistence in CD16+ monocytes and an associated inflammatory profile may contribute to PCVS. Further studies are needed to confirm causality and prevalence.
Collapse
Affiliation(s)
| | - Ram Yogendra
- Department of Anesthesiology, Lawrence General Hospital, Lawrence, MA, USA
| | | | - Jose Guevara-Coto
- Lab of Tumor Chemosensitivity, CIET/DC Lab, Faculty of Microbiology, Universidad de Costa Rica, San Jose, Costa Rica
| | - Emily Long
- Research and Development Department, IncellDx Inc, Hayward, CA, USA
| | - Amruta Pise
- Research and Development Department, IncellDx Inc, Hayward, CA, USA
| | - Eric Osgood
- Department of Medicine, St. Francis Medical Center, Trenton, NJ, USA
| | - John Bream
- Department of Emergency Medicine, Novant Health Kernersville Medical Center, Kernersville, NC, USA
| | - Mark Kreimer
- Department of Emergency Medicine, New York Presbyterian Hospital, Brooklyn, NY, USA
| | - Devon Jeffers
- Department of Anesthesiology, Stamford Hospital, Stamford, CT, USA
| | | | | | - Rodrigo A. Mora-Rodríguez
- Lab of Tumor Chemosensitivity, CIET/DC Lab, Faculty of Microbiology, Universidad de Costa Rica, San Jose, Costa Rica
| |
Collapse
|
2
|
Zhu Q, Chen Z, Wang D, Jiao X, Luan Y, Wang M, Luo R, Wang Y, Fu G, Wang Y, Zhang W. Microenvironment-responsive coating for vascular stents to regulate coagulation-inflammation interaction and promote vascular recovery. Bioact Mater 2025; 48:443-457. [PMID: 40093305 PMCID: PMC11909720 DOI: 10.1016/j.bioactmat.2025.02.031] [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: 10/27/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Early coagulation-inflammation interaction and late in-stent restenosis undermine the efficacy of vascular stents after implantation. Targeting the interplay between inflammation and coagulation, and smooth muscle cell (SMC) proliferation, we presented a microenvironment-responsive coating designed to regulate tissue responses and vascular regeneration throughout the remodeling process. Coagulation was inhibited by incorporating anticoagulant tirofiban into the coating. MMP9-responsive nanoparticles embedded in the coating released salvianolic acid A to modulate inflammatory cell behavior and inhibit SMC dysfunction. By effectively interfering with clotting and inflammation, the coating suppressed platelet-fibrin interaction and formation of platelet-monocyte aggregates, thereby mitigating adverse effects on reendothelialization. Its ability to influence SMC proliferation and migration resulted in reduced intimal hyperplasia. Coated stents were shown to significantly regulate tissue regeneration, improve the vascular environment and even reduced the lipid content in the narrowed atherosclerotic vessels in vivo. This direct approach enhanced the vascular tissue regeneration after stent implantation, and offered promising insights for optimizing vascular stent design.
Collapse
Affiliation(s)
- Qiongjun Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Zhezhe Chen
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Dan'an Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Xiaolu Jiao
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Min Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials and College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Yanan Wang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, 310016, China
| |
Collapse
|
3
|
Lee SK, Xiong T, Qian AS, Yoo JA, Sokeechand BSH, Fuller MT, Gross PL, Austin RC, Igdoura SA, Trigatti BL. Scavenger receptor class B type I knockout mice develop extensive diet-induced coronary artery atherosclerosis in an age-dependent manner. PLoS One 2025; 20:e0318118. [PMID: 40403091 PMCID: PMC12097598 DOI: 10.1371/journal.pone.0318118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/10/2025] [Indexed: 05/24/2025] Open
Abstract
OBJECTIVE Homozygous knockout of scavenger receptor class B type I (SR-B1) in mice with atherogenic mutations (such as knockout of the apolipoprotein E or low density lipoprotein receptor genes) results in spontaneous or diet-induced coronary heart disease characterized by atherosclerosis development in the aortic sinus and coronary arteries, platelet accumulation in coronary artery plaques, myocardial fibrosis, and early death. However, the extent of coronary artery atherothrombosis and myocardial fibrosis in mice lacking SR-B1 alone (homozygous SR-B1 knockout mice) has not been examined. Although age is a major risk factor for coronary artery disease, few studies directly examine the effects of age on susceptibility to atherosclerosis or coronary artery atherothrombosis and myocardial fibrosis in mice. Therefore, we set out to examine the effects of age on diet-induced atherosclerosis in female homozygous SR-B1 knockout mice. APPROACH AND RESULTS SR-B1 knockout mice exhibited little-to-no aortic sinus or coronary artery atherosclerosis at 52 weeks of age, when fed a normal diet. However when fed a high-fat, high-cholesterol, cholate-containing (HFCC) diet for 12 weeks from either 14 weeks of age (26-week-old at analysis) or 40 weeks of age (52-week-old at analysis), they developed similar degrees of atherosclerosis in their aortic sinuses. Interestingly, the older aged SR-B1 knockout mice exhibited increased coronary artery atherosclerosis, increased vascular cell adhesion molecule 1 levels and platelet accumulation in coronary arteries, and increased myocardial fibrosis and plasma levels of cardiac troponin I compared to the younger aged mice. Older-aged HFCC diet-fed SR-B1 knockout mice also exhibited reduced survival to humane endpoint. Moreover, older-aged HFCC diet-fed SR-B1 knockout mice exhibited a greater inflammatory state with increased levels of circulating interleukin-6, tumour necrosis factor alpha, and neutrophils, despite plasma lipid levels being unchanged. Consistent with the increased circulating neutrophils, older-aged HFCC diet-fed SR-B1 knockout mice exhibited increased accumulation of the neutrophil marker myeloperoxidase and increased neutrophil extracellular traps in atherosclerotic plaques in the aortic sinus and increased abundance of atherosclerotic coronary arteries containing neutrophil extracellular traps. CONCLUSIONS HFCC diet-fed homozygous SR-B1 knockout mice develop occlusive coronary artery atherothrombosis and myocardial fibrosis in an age-dependent manner, and exhibit an increased inflammatory state with older age. Therefore, aged SR-B1 knockout mice may prove to be an attractive mouse model to analyze age-dependent mechanisms associated with coronary artery disease development, which may facilitate the discovery of more effective therapeutics to treat cardiovascular disease.
Collapse
Affiliation(s)
- Samuel K. Lee
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Ting Xiong
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Alexander S. Qian
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Jeong-Ah Yoo
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - B. Sumayyah H. Sokeechand
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Mark T. Fuller
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Peter L. Gross
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Richard C. Austin
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe’s Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Suleiman A. Igdoura
- Department of Biology and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
4
|
Harm T, Lahu S, Mayer K, Rath D, Geisler T, Müller KAL, Janisch M, Adler K, Münch G, Massberg S, Kastrati A, Gawaz MP. Plasma chemokines indicate enhanced bleeding in patients with chronic coronary syndrome undergoing percutaneous coronary stenting. Clin Res Cardiol 2025:10.1007/s00392-025-02675-8. [PMID: 40397144 DOI: 10.1007/s00392-025-02675-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/21/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Patients with coronary artery disease (CAD) are at increased risk of developing ischemic events and contemporary antiplatelet therapy often leads to bleeding events following percutaneous coronary intervention (PCI). Glycoprotein VI (GPVI) is the key receptor of collagen-dependent thrombus formation and crucial for platelet homeostasis. METHODS We analysed the influence of GPVI inhibition with revacept in a randomized double-blinded trial enrolling 334 patients with CAD undergoing elective PCI. Ex vivo platelet function analyses were assessed alongside plasma chemokine concentrations. We then elucidate changes of GPVI-dependent chemokine concentrations in patients with bleeding events during the 30-day clinical follow-up. RESULTS Changes in platelet function occur in patients with revacept treatment and are associated with a characteristic alteration of circulating chemokine concentrations. Further, patients with adverse bleeding events share a distinct fingerprint of chemokines that is associated with modulation of in vitro platelet functions. In addition, assessment of GPVI-associated changes in chemokine signalling and platelet functions demonstrated an increased diagnostic value in patients with CAD and might improve early risk discrimination for bleeding events. CONCLUSION The composition of platelet-derived chemokines correlated with platelet functions following antiplatelet treatment. Thus, assessment of chemokines may offer the perspective to identify patients at increased risk for bleeding events. Likewise, modulation of platelet chemokines in patients with revacept treatment contributes to the efficacy of antiplatelet treatment and might attenuate pathophysiological cascades leading to haemorrhagic diathesis in patients with CAD.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Shqipdona Lahu
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany
| | - Katharina Mayer
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Marion Janisch
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany
| | | | | | - Steffen Massberg
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Cardiology, Munich University Clinic, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Adnan Kastrati
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
| |
Collapse
|
5
|
Tian SY, Yu MJ, Mei K, Xu B, Xiao LC, Wen HB, Shang FR. Elevated MPV: A Key Indicator of Acute Anterior Circulation Stroke Prognosis in Mechanical Thrombectomy. Ther Clin Risk Manag 2025; 21:715-725. [PMID: 40417170 PMCID: PMC12102740 DOI: 10.2147/tcrm.s522253] [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: 02/21/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025] Open
Abstract
Objective The aim of this study is to investigate the relationship between admission platelet indices and 90-day clinical outcomes in patients with acute ischemic stroke (AIS) undergoing mechanical thrombectomy (MT). Methods A retrospective analysis was conducted on 247 AIS patients with anterior circulation large vessel occlusion (LVO) treated with MT between July 2021 and April 2024. Platelet indices (PIs) were measured at admission. Participants were stratified into two groups based on 90-day modified Rankin Scale (mRS) outcomes. Multivariate regression analysis and receiver operating characteristic (ROC) curves were employed to evaluate relationships between admission platelet indices, clinical parameters, and functional outcomes. Results Among 247 enrolled patients, those with unfavorable outcomes (mRS 3-6) exhibited significantly higher platelet distribution width (PDW) and Mean Platelet Volume (MPV) levels compared to the favorable outcome group (mRS 0-2). Elevated MPV remained an independent predictor of unfavorable outcomes after multivariate adjustment (OR=2.747, 95% CI: 1.791-4.216, P<0.001). ROC analysis identified a MPV threshold >10.75 fL for predicting unfavorable prognosis, demonstrating 55.4% sensitivity and 81.2% specificity. Conclusion PDW is associated with unfavorable 90-day outcomes in patients with acute anterior circulation LVO following MT, while elevated MPV may serve as a prognostic indicator for unfavorable functional outcomes in patients with acute anterior circulation LVO following MT.
Collapse
Affiliation(s)
- Shi-Yuan Tian
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Min-Jie Yu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Kefu Mei
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Bing Xu
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Lian-Chen Xiao
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Hong-Bin Wen
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Fu-Rong Shang
- Department of Neurology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| |
Collapse
|
6
|
Wang Y, Dou W, Qian X, Chen H, Zhang Y, Yang L, Wu Y, Xu X. Advancements in the study of short-chain fatty acids and their therapeutic effects on atherosclerosis. Life Sci 2025; 369:123528. [PMID: 40049368 DOI: 10.1016/j.lfs.2025.123528] [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: 12/01/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis (AS) remains a leading cause of cardiovascular disease and mortality globally. This chronic condition is characterized by inflammation, lipid accumulation, and the deposition of cellular components within arterial walls. Emerging evidence has highlighted the multifaceted therapeutic potential of short-chain fatty acids (SCFAs) in mitigating AS progression. SCFAs have demonstrated anti-inflammatory properties and the ability to regulate immune responses, metabolic pathways, vascular integrity, and intestinal barrier function in animal models of AS. Consequently, SCFAs have garnered significant attention as a promising approach for the prevention and treatment of AS. However, further clinical trials and studies are necessary to fully elucidate the underlying mechanisms and effects of SCFAs. Additionally, different types of SCFAs may exert distinct impacts, necessitating more in-depth investigation into their specific roles and mechanisms. This review provides an overview of the diverse cellular mechanisms contributing to AS formation, as well as a discussion of the significance of SCFAs in AS pathogenesis and their multifaceted therapeutic potential. Nonetheless, additional research is warranted to comprehensively understand and harness the potential of various SCFAs in the context of AS.
Collapse
Affiliation(s)
- Yongsen Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China; Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China; Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Wei Dou
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xin Qian
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Hao Chen
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Yi Zhang
- Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Liu Yang
- Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xiongfei Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
7
|
Luo H, Li G, Chen Y, Shen Y, Shen W. Association of platelet-to-high-density lipoprotein cholesterol ratio and its cumulative exposure with cardiovascular disease risk: a prospective cohort study in Chinese population. Front Cardiovasc Med 2025; 12:1580359. [PMID: 40416811 PMCID: PMC12098545 DOI: 10.3389/fcvm.2025.1580359] [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: 02/20/2025] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Abstract
Objective This study aimed to investigate the association of platelet-to-high-density lipoprotein cholesterol ratio (PHR) and its cumulative exposure with cardiovascular disease (CVD) risk. Methods The investigation utilized data from the China Health and Retirement Longitudinal Study (CHARLS). Platelet-to-high-density lipoprotein cholesterol ratio was calculated as platelet count (×10⁹/L)/high-density lipoprotein cholesterol (mmol/L), and a cumulative platelet-to-high-density lipoprotein cholesterol ratio (Cumulative PHR) was derived for longitudinal assessment. Multivariable logistic regression models were used to evaluate the association between PHR, cumulative PHR, and CVD risk across three models with increasing adjustments for confounders. Restricted cubic splines (RCS) regressions were utilized to examine if there were non-linear relationships. Subgroup analyses were conducted to enhance the reliability of the study findings. Furthermore, predictive performance was assessed using concordance index (C-index), net reclassification improvement (NRI), and integrated discrimination improvement (IDI). Results A total of 7,063 participants aged 45 and older were included, of whom 1,433 (20.29%) experienced a cardiovascular disease. Participants with CVD had higher PHR (167.93 vs. 156.84, P < 0.001) and Log PHR (5.12 vs. 5.06, P < 0.001) values compared to non-CVD participants. Multivariable logistic regression revealed that Log PHR was independently associated with CVD risk [Odds ratio (OR) per-unit: 1.30, 95% confidence interval (CI): 1.13-1.49, P < 0.001; OR per- standard deviation (SD): 1.13, 95% CI: 1.06-1.21, P < 0.001]. Log cumulative PHR showed similar associations (OR per-unit: 1.34, 95% CI: 1.05-1.71, P = 0.02). Participants in the highest quartile of Log PHR had a nearly 1.32-fold higher risk of CVD compared to the lowest quartile (OR: 1.32, 95% CI: 1.10-1.57, P = 0.002). Addition of Log PHR and Log cumulative PHR slightly improved predictive performance metrics of baseline model. Conclusion Both Log PHR and Log cumulative PHR are independently associated with increased CVD risk and slightly improved the predictive performance of the baseline risk model. Future research should focus on its clinical implementation and integration into existing risk assessment frameworks.
Collapse
Affiliation(s)
- Honglian Luo
- Department of Neurology, Wuhan Fourth Hospital (Wuhan Puai Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Gang Li
- Department of Neurology, Wuhan Fourth Hospital (Wuhan Puai Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Chen
- Department of Neurology, Wuhan Fourth Hospital (Wuhan Puai Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yun Shen
- Division of Population and Public Health Science, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Wei Shen
- Department of Neurology, Wuhan Fourth Hospital (Wuhan Puai Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
8
|
Luo Y, Liu J, Qu P, Han S, Li X, Wang Y, Su X, Zeng J, Li J, Deng S, Liang Q, Hou L, Cheng P. The crosstalk of breast cancer and ischemic heart disease. Cell Death Discov 2025; 11:185. [PMID: 40251177 PMCID: PMC12008236 DOI: 10.1038/s41420-025-02428-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
In recent years, the continuous optimization of anti-tumor therapy has greatly improved the cancer-specific survival rate for patients with breast cancer (BC). The prevention and treatment of breast cancer-related heart diseases have become a new breakthrough in improving the long-term survival for BC patient. The cardiac damages caused by BC treatment are increasingly prominent among BC patients, of which ischemic heart disease (IHD) is the most prominent. Besides, the systemic inflammatory response activated by tumor microenvironment c an induce and exacerbate IHD and increase the risk of myocardial infarction (MI). Conversely, IHD can also exert detrimental effects on tumors. MI not only increases the risk of BC, but also induces specialized immune cell to BC and accelerates the progression of BC. Meanwhile, the treatment of IHD can also promote BC metastasis and transition to more aggressive phenotypes. Although BC and IHD are diseases of two independent systems, their crosstalk increases the difficulty of anti-cancer treatment and IHD management, which reduces the survival for both diseases. Therefore, this review mainly explores the mutual influence and underlying mechanisms between BC and IHD, aiming to provide insights for improving the long-term survival for patients with BC or IHD.
Collapse
Affiliation(s)
- Yunbo Luo
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Jun Liu
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Peng Qu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
| | - Shiqi Han
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Xue Li
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
| | - Yali Wang
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Xiaohan Su
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Jiao Zeng
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Jinsui Li
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Shishan Deng
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Qi Liang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China.
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China.
| | - Lingmi Hou
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China.
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, 610072, P.R. China.
| |
Collapse
|
9
|
Sun W, Chen Z, Luo Y. Association Between Systemic Immune-Inflammation Index and Outcomes of Acute Myocardial Infarction: A Systemic Review and Meta-Analysis. Surg Infect (Larchmt) 2025; 26:183-194. [PMID: 39699344 DOI: 10.1089/sur.2024.172] [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] [Indexed: 12/20/2024] Open
Abstract
Objective: To assess the link between systemic immune-inflammation index (SII) and risk of major adverse cardiovascular events (MACE), contrast-induced nephropathy (CIN), and overall mortality in patients with acute myocardial infarction (AMI). Patients and Methods: Electronic search of PubMed, EMBASE, Web of Science, and Scopus databases was done for observational studies with the data on the association of SII and outcomes, such as MACE, and CIN in adult (≥18 y) patients with AMI. A random-effects model was used, and the pooled effect sizes were expressed as relative risk (RR) with corresponding 95% confidence intervals (CI). Subgroup analysis was conducted on the basis of the type of AMI (ST elevation myocardial infarction and non-ST elevation myocardial infarction), sample size (≥500 and <500), and study design. GRADE assessment was used to evaluate the certainty of the evidence. Results: The analysis included 23 studies. Most studies were conducted in China (n = 13), followed by Turkey (n = 10). Majority of the studies (n = 20) had a retrospective cohort design. Patients with high SII had increased risk of MACE (RR 2.95, 95% CI: 1.25, 6.99; n = 5, I2 = 97.5%), overall mortality (RR 2.59, 95% CI: 1.64, 4.07; n = 6, I2 = 58.0%), and CIN (RR 4.58, 95% CI: 3.44, 6.10; n = 4, I2 = 0.0%), compared with patients with lower SII. Egger's test detected publication bias for MACE (p = 0.047) and overall mortality (p = 0.012) but not for CIN. These associations remained valid in subgroup analysis. Conclusion: Findings suggest that higher SII in patients with AMI is associated with increased risks of MACE, CIN, and overall mortality. This underscores SII's potential as a prognostic marker in AMI.
Collapse
Affiliation(s)
- Wen Sun
- EICU, Changxing People's Hospital of Zhejiang, Huzhou City, China
| | - Zheye Chen
- Department of Emergency, Changxing People's Hospital of Zhejiang, Huzhou City, China
| | - Yi Luo
- EICU, Changxing People's Hospital of Zhejiang, Huzhou City, China
| |
Collapse
|
10
|
Sheptulina AF, Liusina EO, Zlobovskaya OA, Kiselev AR, Drapkina OM. Possible Role of Platelets in the Development and Progression of Non-Alcoholic Fatty Liver Disease. FRONT BIOSCI-LANDMRK 2025; 30:26748. [PMID: 40152376 DOI: 10.31083/fbl26748] [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: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 03/29/2025]
Abstract
To date, an increasing body of evidence supports the potential role of activated platelets in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). This is likely due to their ability to secrete biologically active substances that regulate liver regeneration processes, ensure hemostasis, and participate in the immune response. Additionally, several studies have demonstrated the efficacy of antiplatelet agents in reducing inflammation, the severity of liver fibrosis, and the progression of fibrosis in non-alcoholic steatohepatitis (NASH). Since NAFLD is not an independent indication for antiplatelet therapy, the primary evidence regarding their efficacy in NAFLD has been derived from studies using animal models of NAFLD or in patients with concomitant cardiovascular diseases. This narrative review will discuss the main functions of platelets, their unique interactions with liver cells, and the outcomes of these interactions, as well as the results of studies evaluating the efficacy and safety of antiplatelet therapy in patients with NAFLD.
Collapse
Affiliation(s)
- Anna F Sheptulina
- Department of Fundamental and Applied Aspects of Obesity, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
- Department of Therapy and Preventive Medicine, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
| | - Ekaterina O Liusina
- Department of Fundamental and Applied Aspects of Obesity, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
| | - Olga A Zlobovskaya
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 123182 Moscow, Russia
| | - Anton R Kiselev
- Coordinating Center for Fundamental Research, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
| | - Oxana M Drapkina
- Department of Fundamental and Applied Aspects of Obesity, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
- Department of Therapy and Preventive Medicine, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
| |
Collapse
|
11
|
Coenen DM, Alfar HR, Whiteheart SW. Platelet endocytosis and α-granule cargo packaging are essential for normal skin wound healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.01.636051. [PMID: 39975047 PMCID: PMC11838500 DOI: 10.1101/2025.02.01.636051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The high prevalence of chronic wounds, i.e., 2.5-3% of the US population, causes a large social and financial burden. Physiological wound healing is a multi-step process that involves different cell types and growth factors. Platelet-rich plasma or platelet-derived factors have been used to accelerate wound repair, but their use has been controversial with mixed results. Thus, a detailed functional understanding of platelet functions in wound healing beyond hemostasis is needed. This study investigated the importance of platelet α-granule cargo packaging and endocytosis in a dorsal full-thickness excisional skin wound model using mice with defects in α-granule cargo packaging (Nbeal2 -/- mice) and endocytosis (platelet-specific Arf6 -/- and VAMP2/3 Δ mice). We found that proper kinetic and morphological healing of dorsal skin wounds in mice requires both de novo as well as endocytosed platelet α-granule cargo. Histological and morphometric analyses of cross-sectional wound sections illustrated that mice with defects in α-granule cargo packaging or platelet endocytosis had delayed (epi)dermal regeneration in both earlier and advanced healing. This was reflected by reductions in wound collagen and muscle/keratin content, delayed scab formation and/or resolution, re-epithelialization, and cell migration and proliferation. Molecular profiling analysis of wound extracts showed that the impact of platelet function extends beyond hemostasis to the inflammation, proliferation, and tissue remodeling phases via altered expression of several bioactive molecules, including IL-1β, VEGF, MMP-9, and TIMP-1. These findings provide a basis for advances in clinical wound care through a better understanding of key mechanistic processes and cellular interactions in (patho)physiological wound healing. Key points De novo and endocytosed platelet α-granule cargo support physiological skin wound healing Platelet function in wound healing extends to the inflammation, proliferation, and tissue remodeling phases.
Collapse
|
12
|
Kennedy NN, Xia Y, Barrett T, Luttrell-Williams E, Berland T, Cayne N, Garg K, Jacobowitz G, Lamparello PJ, Maldonado TS, Newman J, Sadek M, Smilowitz NR, Rockman C, Berger JS. Dynamic perioperative platelet activity and cardiovascular events in peripheral artery disease. J Vasc Surg 2025; 81:432-440.e3. [PMID: 39362415 DOI: 10.1016/j.jvs.2024.09.028] [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: 05/03/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024]
Abstract
OBJECTIVE Patients with peripheral artery disease (PAD) undergo lower extremity revascularization (LER) for symptomatic relief or limb salvage. Despite LER, patients remain at increased risk of platelet-mediated complications, such as major adverse cardiac and limb events (MACLEs). Platelet activity is associated with cardiovascular events, yet little is known about the dynamic nature of platelet activity over time. We, therefore, investigated the change in platelet activity over time and its association with long-term cardiovascular risk. METHODS Patients with PAD undergoing LER were enrolled into the multicenter, prospective Platelet Activity and Cardiovascular Events study. Platelet aggregation was assessed by light transmission aggregometry to submaximal epinephrine (0.4 μmol/L) immediately before LER, and on postoperative day 1 or 2 (POD1 or POD2) and 30 (POD30). A hyperreactive platelet phenotype was defined as >60% aggregation. Patients were followed longitudinally for MACLEs, defined as the composite of death, myocardial infarction, stroke, major lower extremity amputation, or acute limb ischemia leading to reintervention. RESULTS Among 287 patients undergoing LER, the mean age was 70 ± 11 years, 33% were female, 61% were White, and 89% were on baseline antiplatelet therapy. Platelet aggregation to submaximal epinephrine induced a bimodal response; 15.5%, 16.8%, and 16.4% of patients demonstrated a hyperreactive platelet phenotype at baseline, POD1, and POD30, respectively. Platelet aggregation increased by 18.5% (P = .001) from baseline to POD1, which subsequently returned to baseline at POD30. After a median follow-up of 19 months, MACLEs occurred in 165 patients (57%). After adjustment for demographics, clinical risk factors, procedure type, and antiplatelet therapy, platelet hyperreactivity at POD1 was associated with a significant hazard of long-term MACLE (adjusted hazard ratio, 4.61; 95% confidence interval, 2.08-10.20; P < .001). CONCLUSIONS Among patients with severe PAD, platelet activity increases after LER. Platelet hyperreactivity to submaximal epinephrine on POD1 is associated with long-term MACLE. Platelet activity after LER may represent a modifiable biomarker associated with excess cardiovascular risk.
Collapse
Affiliation(s)
- Natalie N Kennedy
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Yuhe Xia
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Tessa Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Elliot Luttrell-Williams
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Todd Berland
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Neal Cayne
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Karan Garg
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Glenn Jacobowitz
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Patrick J Lamparello
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Thomas S Maldonado
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Jonathan Newman
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Mikel Sadek
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Nathaniel R Smilowitz
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY; Cardiology Section, Department of Medicine, VA New York Harbor Healthcare System, New York, NY
| | - Caron Rockman
- Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY
| | - Jeffrey S Berger
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, NY; Division of Vascular and Endovascular Surgery, Department of Surgery, NYU Langone Health and Manhattan Veterans Affairs Hospital, New York, NY.
| |
Collapse
|
13
|
Huang C, Wei Z, Zheng N, Yan J, Zhang J, Ye X, Zhao W. The interaction between dysfunction of vasculature and tauopathy in Alzheimer's disease and related dementias. Alzheimers Dement 2025; 21:e14618. [PMID: 39998958 PMCID: PMC11854360 DOI: 10.1002/alz.14618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/01/2025] [Accepted: 01/12/2025] [Indexed: 02/27/2025]
Abstract
Tauopathy is one of the pathological features of Alzheimer's disease and related dementias (ADRD). At present, there have been many studies on the formation, deposition, and intercellular transmission of tau in neurons and immune cells. The vasculature is an important component of the central nervous system. This review discusses the interaction between vasculature and tau in detail from three aspects. (1) The vascular risk factors (VRFs) discussed in this review include diabetes mellitus (DM), abnormal blood pressure (BP), and hypercholesterolemia. (2) In ADRD pathology, the hyperphosphorylation and deposition of tau interact with disrupted vasculature, such as different cells (endothelial cells, smooth muscular cells, and pericytes), the blood-brain barrier (BBB), and the cerebral lymphatic system. (3) The functions of vasculature are regulated by various signaling transductions. Endothelial nitric oxide synthase/nitric oxide, calcium signaling, Rho/Rho-associated coiled-coil containing Kinase, and receptors for advanced glycation end products are discussed in this review. Our findings indicate that the prevention and treatment of vascular health may be a potential target for ADRD combination therapy. HIGHLIGHTS: Persistent VRFs increase early disruption of vascular mechanisms and are strongly associated with tau pathology in ADRD. Cell dysfunction in the vasculature causes BBB leakage and drainage incapacity of the cerebral lymphatic system, which interacts with tau pathology. Signaling molecules in the vasculature regulate vasodilation and contraction, angiogenesis, and CBF. Abnormal signaling transduction is related to tau hyperphosphorylation and deposition.
Collapse
Affiliation(s)
- Chuyao Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Zhenwen Wei
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Ningxiang Zheng
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jingsi Yan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jiayu Zhang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xinyi Ye
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Wei Zhao
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| |
Collapse
|
14
|
Gürdal A, Serin E, Sarilar M, Sümerkan MÇ, Kalender E, Değirmenci H, Şahin S, Keskin K. Prognostic value of systemic immune-inflammation index in older patients with acute coronary syndrome. Coron Artery Dis 2025:00019501-990000000-00335. [PMID: 39817705 DOI: 10.1097/mca.0000000000001496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
OBJECTIVES Contemporary studies assessing the importance of the systemic immune-inflammation index (SII) in older patients presenting with acute coronary syndrome (ACS) are scarce. This study investigated the impact and prognostic value of the SII regarding long-term mortality in older patients with ACS. METHODS The study included 401 older patients aged 75 years and above admitted with ACS between May 2015 and December 2022. Predictors of mortality were determined using multivariate Cox regression analysis. Survival curves were generated using the Kaplan-Meier method. RESULTS The patients' median age was 81 (77-85) years, and 197 (49.1%) were male. The median follow-up was 23 months (Q1-Q3 : 4-43, maximum: 102). All short- and long-term deaths, including in-hospital deaths, were significantly high in patients with high SII (P = 0.001). Inflammatory variables, including C-reactive protein, SII, the neutrophil-to-lymphocyte ratio, monocyte-to-lymphocyte ratio, and platelet-to-lymphocyte ratio, were positively correlated with the SYNTAX score (for SII; R = 0.492, P = 0.001). Multivariate Cox regression analysis revealed that age [hazard ratio (HR): 1.082, 95% confidence interval (CI): 1.051-1.114, P = 0.001], estimated glomerular filtration rate (HR: 0.988, 95% CI: 0.982-0.994, P = 0.001), SII (HR: 1.004, 95% CI: 1.001-1.006, P = 0.001), and left ventricular ejection fraction (HR: 0.959, 95% CI: 0.947-0.97, P = 0.001) were independent predictors of mortality in older patients with ACS. Kaplan-Meier analysis also showed that patients with high SII had a significantly higher mortality rate (P = 0.001). CONCLUSION A high SII is an independent predictor of long-term mortality in older patients with ACS.
Collapse
Affiliation(s)
- Ahmet Gürdal
- Department of Cardiology, University of Health Sciences, Şişli Hamidiye Etfal Education and Research Hospital, Istanbul, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Li X, Wang J, Zhang M, Li Y, Li X, Zhang J, Zhang L, Zhang Y, Qiu Z. Phenotypic age mediates the associations between platelet-to-lymphocyte ratio and all-cause and cause-specific mortality: A prospective cohort study. Heliyon 2025; 11:e41506. [PMID: 39831170 PMCID: PMC11742625 DOI: 10.1016/j.heliyon.2024.e41506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/03/2025] Open
Abstract
OBJECTIVES The platelet-to-lymphocyte ratio (PLR) is a novel indicator of inflammation, but research on the links and mechanisms between the PLR and long-term health conditions is lacking. This study aimed to evaluate the relationship between phenotypic age (PhenoAge) mediated PLR and mortality among US adults. METHODS A total of 37,182 participants from the National Health and Nutrition Examination Survey (NHANES) database (1999-2018) were included to evaluate the PLR's relevance to survival by Cox regression models. The associations between the PLR and mortality were apparent using restricted cubic spline regression. Mediation analyses were conducted to investigate the mediated effects of PhenoAge on the associations of PLR with mortality. RESULTS Compared to the PLR in Quintile 1 participants, the multivariable-adjusted Cox model showed the PLR in Quintile 5 was linked with greater risks of death from all-cause (HR, 1.14; 95 % CI: 1.04-1.25), cardiovascular disease (CVD) (HR, 1.26; 95 % CI: 1.01-1.57) and respiratory disease (HR, 1.98; 95 % CI: 1.35-2.90). The risk of death from cancer was approximately 28 % lower for participants with the PLR in the fourth quintile. Restricted cubic splines showed the U-shaped relationships between PLR and all-cause and cancer mortality, and the positively linear relationships between PLR and cardiovascular disease (CVD) and respiratory mortality. Moreover, mediation analysis revealed that PhenoAge partially mediated 45.33 %, 44.26 %, and 15.35 % of the associations of PLR with all-cause, CVD, and respiratory disease mortality, respectively. CONCLUSION The PLR, a valuable index that should be recommended for use, was independently linked with all-cause and cause-specific mortality, with PhenoAge playing a partial mediating role in the relationships.
Collapse
Affiliation(s)
- Xiangjun Li
- Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jia Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mengqi Zhang
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yujing Li
- Department of Pathology, The First Hospital of China Medical University, College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning, China
| | - Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jiaqi Zhang
- Department of Medicine, Qingdao University, Qingdao, Liaoning, China
| | - Lihua Zhang
- Department of Medicine, Qingdao University, Qingdao, Liaoning, China
| | - Yixuan Zhang
- Department of Medicine, Qingdao University, Qingdao, Liaoning, China
| | - Zhenkang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
16
|
Akyel S, Korkmaz A, Yıldız A. Assessment of the Severity of Intermediate Coronary Artery Stenosis Using the Systemic Inflammatory Response Index. Diagnostics (Basel) 2025; 15:162. [PMID: 39857046 PMCID: PMC11765139 DOI: 10.3390/diagnostics15020162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Fractional Flow Reserve (FFR) is a method that enables the hemodynamic assessment of coronary artery stenosis. The Systemic Inflammatory Response Index (SIRI) is a new marker calculated by multiplying the neutrophil-to-lymphocyte ratio (NLR) with the monocyte count. It is indicative of the presence and severity of coronary artery disease. This study evaluates the relationship between the functional significance of FFR measurements and the SIRI in intermediate coronary stenosis. Methods: A total of 294 patients with 50-70% stenosis in their coronary arteries based on quantitative measurement following angiography who underwent FFR measurement were included in the study before the FFR procedure. Total and differential leukocyte counts and routine biochemical tests were performed. Results: A total of 37% of the patients were found to have a positive FFR, while 63% had a negative FFR. Significant differences were observed in the neutrophil count, monocyte count, Systemic Inflammation Response Index (SIRI), total cholesterol, and amount of adenosine used between the groups (p < 0.05). A SIRI value of 1.16 was 77% sensitive and 55% specific for FFR positivity. Multivariate logistic regression analysis identified the SIRI as an independent predictor of FFR positivity. Conclusions: Our study has demonstrated that high values of the SIRI may serve as a new biomarker for predicting FFR positivity.
Collapse
Affiliation(s)
- Serdar Akyel
- Department of Cardiology, School of Medicine, Kastamonu University, Kastamonu 37150, Turkey;
| | - Ahmet Korkmaz
- Department of Cardiology, Ankara City Hospital, Ankara 06200, Turkey;
| | - Abdülkadir Yıldız
- Department of Cardiology, School of Medicine, Kastamonu University, Kastamonu 37150, Turkey;
| |
Collapse
|
17
|
Liu K, Yang L, Liu Y, Zhang Y, Zhu J, Zhang H, He Z. Systemic Immune-Inflammation Index (SII) and Neutrophil-to-Lymphocyte Ratio (NLR): A Strong Predictor of Disease Severity in Large-Artery Atherosclerosis (LAA) Stroke Patients. J Inflamm Res 2025; 18:195-202. [PMID: 39802522 PMCID: PMC11724665 DOI: 10.2147/jir.s500474] [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: 10/12/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
Background Systemic immune-inflammation index (SII) and neutrophil-to-lymphocyte ratio (NLR) are novel inflammatory markers based on neutrophil, platelet and lymphocyte counts. Atherosclerosis is a chronic inflammatory vascular disease. This study aimed to verify the predictive value of the clinical parameters such as systemic immune-inflammation index (SII) and neutrophil-to-lymphocyte ratio (NLR) for the severity in Large Artery Atherosclerosis (LAA) stroke patients. Methods The SII is defined as platelet × (neutrophil count/lymphocyte count), the NLR is defined as neutrophil count/lymphocyte count. Univariate logistic regression was used to analyze the association between SII and NLR and NIHSS score in patients with LAA stroke. Multiple logistic regression was used to analyze the risk factors for the severity of LAA stroke. We plotted receiver operating characteristic curves to determine the diagnostic role of SII and NLR in differentiating stroke disease severity. Results We included 283 LAA stroke patients, the SII and NLR in the moderate-to-severe stroke group were significantly higher than the mild stroke group. Multiple logistic regression analysis showed that SII (OR 1.051 95% CI (1.035-1.066), P < 0.001), NLR (OR 1.077,95% CI (1.032-1.123), P < 0.001) were significantly associated with stroke severity. The SII values under the receiver operating characteristic curve (0.701, 95% CI (0.649-0.791, P < 0.001, cut-off value 912.97) and NLR values under the receiver operating characteristic curve (0.604,5% CI (0.519-0.689), P < 0.01, cut-off value 1.461), and SII values had high discrimination ability. Both SII and NLR had high diagnostic and predictive value for stroke severity, and SII was better than NLR. Conclusion The higher SII and NLR, the more severity in LAA stroke patients. SII and NLR are independent risk factors for LAA stroke, and they can also effectively predict stroke severity; moreover, SII has a higher diagnostic efficacy than NLR. However, multicenter studies with large sample size are still needed to confirm this conclusion.
Collapse
Affiliation(s)
- Keting Liu
- Department of Neurology, Chengdu Seventh People’s Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan Province, 610213, People’s Republic of China
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, People’s Republic of China
| | - Li Yang
- Department of Neurology, Chengdu Seventh People’s Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Yang Liu
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610075, People’s Republic of China
| | - Yun Zhang
- Department of Neurology, Chengdu Seventh People’s Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Juncheng Zhu
- Department of Radiology, Chengdu Seventh People’s Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Haofeng Zhang
- Department of Information, Chengdu Seventh People’s Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Zemin He
- Department of Thoracic Surgery, The First People’s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chendu, Sichuan Province, 610200, People’s Republic of China
| |
Collapse
|
18
|
Yu N, Fang R, Ding Z, Xu X, Zhang J. Preparation and structural characterization of a sulfated octasaccharide with heparin-like anticoagulant activity. Carbohydr Polym 2025; 347:122782. [PMID: 39487001 DOI: 10.1016/j.carbpol.2024.122782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 11/04/2024]
Abstract
Heparins are sulfated polysaccharides with a heterogeneous mixture derived from animal tissues, subject to supply limitations and the risk of animal virus residues. Patients using heparin also face the risks of spontaneous bleeding and thrombocytopenia. Here we reported an efficient riclinoctaose-based approach for rapid chemical synthesis of a structurally defined heparin-like anticoagulant sulfated octasaccharide (SRO). We used sulfur trioxide-pyridine, sulfur trioxide-trimethylamine, and sulfur trioxide-triethylamine complexes as solvents for one-pot O-sulfation and determined the optimal conditions for synthesizing SRO. Sulfur trioxide-trimethylamine provided reasonable control over the degree of substitution between 1.85 and 1.88, revealing a single molecule with a theoretical molecular weight of 2952.96 g/mol. The structural features of the SRO were carried out by Fourier transform infrared spectroscopy and one- and two- dimensional 1H and 13C NMR analysis, revealing sulfation repeatedly present at the fixed positions of C-6/C-2/C-3 and reducing terminals. The anticoagulant activity of SRO was demonstrated by efficiently blocking coagulation in the blood of mice and human. SRO dose-dependently decreased ferric chloride-induced experimental thrombosis in mice. Like heparin, SRO specifically inhibits coagulation factor Xa, but significantly reduces the risk of bleeding compared to heparin. Therefore, we named it octaparin. These results support that octaparin is expected to replace animal-sourced heparin.
Collapse
Affiliation(s)
- Ning Yu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China
| | - Rui Fang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China
| | - Zhao Ding
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China.
| |
Collapse
|
19
|
Patrignani P, De Michele A, Tacconelli S. New antiplatelet approach: inhibiting Pim kinase to reduce constitutive surface expression of thromboxane A 2 receptor. J Thromb Haemost 2025; 23:43-46. [PMID: 39798970 DOI: 10.1016/j.jtha.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 01/15/2025]
Affiliation(s)
- Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy.
| | - Alessandra De Michele
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy
| |
Collapse
|
20
|
Gutiérrez‐Muñoz C, Blázquez‐Serra R, Sebastian‐Jaraba IS, Sanz‐Andrea S, Fernández‐Gómez MJ, Nuñez‐Moreno G, Mínguez P, Escolá‐Gil JC, Nogales P, Ollivier V, Martín‐Ventura JL, Noe BH, Rescher U, Méndez‐Barbero N, Blanco‐Colio LM. Annexin A8 deficiency delays atherosclerosis progression. Clin Transl Med 2025; 15:e70176. [PMID: 39835780 PMCID: PMC11748212 DOI: 10.1002/ctm2.70176] [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: 01/10/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids and leukocytes within the arterial wall. By studying the aortic transcriptome of atherosclerosis-prone apolipoprotein E (ApoE-/-) mice, we aimed to identify novel players in the progression of atherosclerosis. METHODS RNA-Seq analysis was performed on aortas from ApoE-/- and wild-type mice. AnxA8 expression was assessed in human and mice atherosclerotic tissue and healthy aorta. ApoE-/- mice lacking systemic AnxA8 (ApoE-/-AnxA8-/-) were generated to assess the effect of AnxA8 deficiency on atherosclerosis. Bone marrow transplantation (BMT) was also performed to generate ApoE-/- lacking AnxA8 specifically in bone marrow-derived cells. Endothelial-specific AnxA8 silencing in vivo was performed in ApoE-/- mice. The functional role of AnxA8 was analysed in cultured murine cells. RESULTS RNA-Seq unveiled AnxA8 as one of the most significantly upregulated genes in atherosclerotic aortas of ApoE-/- compared to wild-type mice. Moreover, AnxA8 was upregulated in human atherosclerotic plaques. Germline deletion of AnxA8 decreased the atherosclerotic burden, the size and volume of atherosclerotic plaques in the aortic root. Plaques of ApoE-/-AnxA8-/- were characterized by lower lipid and inflammatory content, smaller necrotic core, thicker fibrous cap and less apoptosis compared with those in ApoE-/-AnxA8+/+. BMT showed that hematopoietic AnxA8 deficiency had no effect on atherosclerotic progression. Oxidized low-density lipoprotein (ox-LDL) increased AnxA8 expression in murine aortic endothelial cells (MAECs). In vitro experiments revealed that AnxA8 deficiency in MAECs suppressed P/E-selectin and CD31 expression and secretion induced by ox-LDL with a concomitant reduction in platelet and leukocyte adhesion. Intravital microscopy confirmed the reduction in leukocyte and platelet adhesion in ApoE-/-AnxA8-/- mice. Finally, endothelial-specific silencing of AnxA8 decreased atherosclerosis progression. CONCLUSION Our findings demonstrate that AnxA8 promotes the progression of atherosclerosis by modulating endothelial-leukocyte interactions. Interventions capable of reducing AnxA8 expression in endothelial cells may delay atherosclerotic plaque progression. KEY POINTS This study shows that AnxA8 is upregulated in aorta of atheroprone mice and in human atherosclerotic plaques. Germline AnxA8 deficiency reduces platelet and leukocyte recruitment to activated endothelium as well as atherosclerotic burden, plaque size, and macrophage accumulation in mice. AnxA8 regulates oxLDL-induced adhesion molecules expression in aortic endothelial cells. Our data strongly suggest that AnxA8 promotes disease progression through regulation of adhesion and influx of immune cells to the intima. Endothelial specific silencing of AnxA8 reduced atherosclerosis progression. Therapeutic interventions to reduce AnxA8 expression may delay atherosclerosis progression.
Collapse
Affiliation(s)
- Carmen Gutiérrez‐Muñoz
- Vascular Research Laboratory, IIS‐Fundación Jiménez DíazMadridSpain
- CIBERCV, ISCIIIMadridSpain
| | - Rafael Blázquez‐Serra
- Vascular Research Laboratory, IIS‐Fundación Jiménez DíazMadridSpain
- CIBERCV, ISCIIIMadridSpain
| | | | | | | | - Gonzalo Nuñez‐Moreno
- Bioinformatics UnitDepartment of Genetics & GenomicsIIS‐Fundación Jiménez DíazMadridSpain
- CIBERER, ISCIIIMadridSpain
| | - Pablo Mínguez
- Bioinformatics UnitDepartment of Genetics & GenomicsIIS‐Fundación Jiménez DíazMadridSpain
- CIBERER, ISCIIIMadridSpain
| | - Joan Carles Escolá‐Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant PauBarcelonaSpain
- CIBERDEM, ISCIIIMadridSpain
| | - Paula Nogales
- Centro Nacional de Investigaciones CardiovascularesMadridSpain
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Veronique Ollivier
- Laboratory for Vascular Translation ScienceInserm U1148, Paris Bichat HospitalParisFrance
| | - Jose L. Martín‐Ventura
- Vascular Research Laboratory, IIS‐Fundación Jiménez DíazMadridSpain
- CIBERCV, ISCIIIMadridSpain
| | - Benoit Ho‐Tin Noe
- Laboratory for Vascular Translation ScienceInserm U1148, Paris Bichat HospitalParisFrance
| | - Ursula Rescher
- Center for Molecular Biology of InflammationResearch Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, University of MuensterMuensterGermany
| | - Nerea Méndez‐Barbero
- Vascular Research Laboratory, IIS‐Fundación Jiménez DíazMadridSpain
- CIBERCV, ISCIIIMadridSpain
| | - Luis M. Blanco‐Colio
- Vascular Research Laboratory, IIS‐Fundación Jiménez DíazMadridSpain
- CIBERCV, ISCIIIMadridSpain
| |
Collapse
|
21
|
Momi S, Gresele P. The Role of Platelets in Atherosclerosis: A Historical Review. Semin Thromb Hemost 2024. [PMID: 39561814 DOI: 10.1055/s-0044-1795097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Atherosclerosis is a chronic, multifactorial inflammatory disorder of large and medium-size arteries, which is the leading cause of cardiovascular mortality and morbidity worldwide. Although platelets in cardiovascular disease have mainly been studied for their crucial role in the thrombotic event triggered by atherosclerotic plaque rupture, over the last two decades it has become clear that platelets participate also in the development of atherosclerosis, owing to their ability to interact with the damaged arterial wall and with leukocytes. Platelets participate in all phases of atherogenesis, from the initial functional damage to endothelial cells to plaque unstabilization. Platelets deposit at atherosclerosis predilection sites before the appearance of manifest lesions to the endothelium and contribute to induce endothelial dysfunction, thus supporting leukocyte adhesion to the vessel wall. In particular, platelets release matrix metalloproteinases, which interact with protease-activated receptor 1 on endothelial cells triggering adhesion molecule expression. Moreover, P-selectin and glycoprotein Ibα expressed on the surface of vessel wall-adhering platelets bind PSGL-1 and β2 integrins on leukocytes, favoring their arrest and transendothelial migration. Platelet-leukocyte interactions promote the formation of radical oxygen species which are strongly involved in the lipid peroxidation associated with atherosclerosis. Platelets themselves actively migrate through the endothelium toward the plaque core where they release chemokines that modify the microenvironment by modulating the function of other inflammatory cells, such as macrophages. While current antiplatelet agents seem unable to prevent the contribution of platelets to atherogenesis, the inhibition of platelet secretion, of the release of MMPs, and of some specific pathways of platelet adhesion to the vessel wall may represent promising future strategies for the prevention of atheroprogression.
Collapse
Affiliation(s)
- Stefania Momi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Gresele
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
22
|
Bouhaddou N, Mabrouk M, Atifi F, Bouyahya A, Zaid Y. The link between BDNF and platelets in neurological disorders. Heliyon 2024; 10:e39278. [PMID: 39568824 PMCID: PMC11577193 DOI: 10.1016/j.heliyon.2024.e39278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Platelets are considered one of the most important reservoirs not only of growth factors, but also of neurotrophic factors that could contribute to the repair of vascular lesions and the prevention of neurological deterioration. Among these factors, Brain-Derived Neurotrophic Factor (BDNF) - a protein belonging to the neurotrophin family - is widely expressed both in the hippocampus and in platelets. Platelets constitute an important reservoir of BDNF; however, little is known about the factors modulating its release into the circulation and whether anti-platelet drugs affect this secretion. In this review, we have discussed the link between BDNF and platelets and their role in neurological disorders.
Collapse
Affiliation(s)
- Nezha Bouhaddou
- Physiology and Physiopathology Team, Genomics of Human Pathologies Research Center, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Meryem Mabrouk
- Laboratory of Materials, Nanotechnology and Environment, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Farah Atifi
- Laboratory of Materials, Nanotechnology and Environment, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Younes Zaid
- Laboratory of Materials, Nanotechnology and Environment, Faculty of Sciences, Mohammed V University, Rabat, Morocco
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| |
Collapse
|
23
|
Dahan J, Pinthus J, Delouya G, Taussky D, Duceppe E, de Jesus A, Leong D. Investigation of association between clinically significant prostate cancer, obesity and platelet to-lymphocyte ratio and neutrophil -to-lymphocyte ratio. BMC Urol 2024; 24:226. [PMID: 39407194 PMCID: PMC11481316 DOI: 10.1186/s12894-024-01617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
INTRODUCTION Several blood markers of inflammation are elevated in prostate cancer (PCa) and have prognostic value. Little is known about the relationship between these markers, PCa, and other factors associated with chronic inflammation, such as smoking and obesity. We analyzed the interaction between neutrophil and platelet counts indexed to lymphocyte count (NLR and PLR, resp.) and clinically significant PCa (csPCa), accounting for the potential confounding factors of systemic inflammation. METHODS NLR and PLR were evaluated in a multicenter prospective study in 443 patients. CsPCa was defined as a Gleason ≥ 4 + 3. Differences between patients with csPCa and non-csPCA were evaluated using the chi-square test, analysis of variance or the Kruskal-Wallis test. Multivariable logistic regression analysis adjusted for smoking, hypertension, diabetes, and cardiovascular disease, and in separate models, either body mass index or waist-to-hip ratio was used to characterize the relationship between inflammation and csPCa. RESULTS None of the factors such as plateletcrit, NLR, and PLR were significantly different between patients with csPCa or non-significant PCa. After adjustment, there was no association between PLR, NLR, plateletcrit or platelet count and csPCa. In an exploratory analysis, there was no association between markers of inflammation and PSA levels > 10 ng/mL. When testing different NLR cutoffs to predict csPCa in ROC analysis, none reached a clinically meaningful value. CONCLUSION In contrast to previous studies, we found no significant association between easily available blood markers of inflammation and indices of PCa aggressiveness. Further research is required to determine whether inflammation promotes PCa. (ClinicalTrials.gov: NCT03127631. Date of registration: April 25, 2017.
Collapse
Affiliation(s)
- Johanna Dahan
- Department of Radiation Oncology, Centre hospitalier de l'Université de Montréal, 1000 rue St Denis, Montréal, QC, H2X 0C1, Canada
| | - Jehonathan Pinthus
- Department of Surgery, Juravinski Cancer Center/Hamilton Health Sciences, McMaster University, Hamilton, Canada
- Department of Surgery, Division of Urology, McMaster University, St. Joseph's Healthcare, Hamilton, Canada
| | - Guila Delouya
- Department of Radiation Oncology, Centre hospitalier de l'Université de Montréal, 1000 rue St Denis, Montréal, QC, H2X 0C1, Canada
| | - Daniel Taussky
- Department of Radiation Oncology, Centre hospitalier de l'Université de Montréal, 1000 rue St Denis, Montréal, QC, H2X 0C1, Canada.
| | - Emmanuelle Duceppe
- Department of Medicine, Centre hospitalier de l'Université de Montréal, Montréal, Canada
| | - Amanda de Jesus
- Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Darryl Leong
- Departments of Medicine and Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| |
Collapse
|
24
|
Pan D, Chen H. Relationship between serum albumin level and hospitalization duration following percutaneous coronary intervention for acute coronary syndrome. Sci Rep 2024; 14:23883. [PMID: 39396090 PMCID: PMC11470946 DOI: 10.1038/s41598-024-74955-8] [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: 05/30/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024] Open
Abstract
Low serum albumin levels increase coronary morbidity, mortality, and postoperative cardiovascular risk. Therefore, this study investigated the relationship between these levels and the length of hospital stay in patients undergoing percutaneous coronary intervention (PCI) for acute coronary syndrome. A total of 350 patients were divided into quartiles according to serum albumin levels. Univariate and multivariate analyses were performed to identify factors associated with the length of hospital stay. A non-linear regression analysis of serum albumin and length of hospital stay was also performed. The results of the multifactorial analysis revealed low serum albumin levels as an independent predictor of longer hospital stay, even in the fully adjusted model. In the segmented linear regression model, serum albumin level showed a U-shaped relationship with the length of hospital stay. In conclusion, low serum albumin level was an independent predictor of longer hospital stay in patients undergoing PCI for acute coronary syndrome, with shorter stays observed for increasing serum albumin levels. Low serum albumin can be used to identify patients who require longer hospitalization and may need additional nutritional support or interventions to improve their prognosis.
Collapse
Affiliation(s)
- Di Pan
- Department of Cardiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No. 3002, Sungang West Road, Futian District, Shenzhen, 518035, China
- Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Haibo Chen
- Department of Cardiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No. 3002, Sungang West Road, Futian District, Shenzhen, 518035, China.
| |
Collapse
|
25
|
Tao Q, Fan LP, Feng J, Zhang ZJ, Liu XW, Qin Z, Li JY, Yang YJ. Platelet Proteomics and Tissue Metabolomics Investigation for the Mechanism of Aspirin Eugenol Ester on Preventive Thrombosis Mechanism in a Rat Thrombosis Model. Int J Mol Sci 2024; 25:10747. [PMID: 39409077 PMCID: PMC11476519 DOI: 10.3390/ijms251910747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Platelet activation is closely related to thrombosis. Aspirin eugenol ester (AEE) is a novel medicinal compound synthesized by esterifying aspirin with eugenol using the pro-drug principle. Pharmacological and pharmacodynamic experiments showed that AEE has excellent anti-inflammatory, antioxidant, and inhibitory platelet activation effects, preventing thrombosis. However, the regulatory network and action target of AEE in inhibiting platelet activation remain unknown. This study aimed to investigate the effects of AEE on platelets of thrombosed rats to reveal its regulatory mechanism via a multi-omics approach. The platelet proteomic results showed that 348 DEPs were identified in the AEE group compared with the model group, of which 87 were up- and 261 down-regulated. The pathways in this result were different from previous results, including mTOR signaling and ADP signaling at P2Y purinoceptor 12. The metabolomics of heart and abdominal aortic tissue results showed that the differential metabolites were mainly involved in steroid biosynthesis, the citric acid cycle, phenylalanine metabolism, phenylalanine, tyrosine, and tryptophan biosynthesis, and glutathione metabolism. Molecular docking results showed that AEE had a better binding force to both the COX-1 and P2Y12 protein. AEE could effectively inhibit platelet activation by inhibiting COX-1 protein and P2Y12 protein activity, thereby inhibiting platelet aggregation. Therefore, AEE can have a positive effect on inhibiting platelet activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jian-Yong Li
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (L.-P.F.); (J.F.); (Z.-J.Z.); (X.-W.L.); (Z.Q.)
| | - Ya-Jun Yang
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (L.-P.F.); (J.F.); (Z.-J.Z.); (X.-W.L.); (Z.Q.)
| |
Collapse
|
26
|
Gangavarapu RR, Mahmud SA, Manandhar A, Sabir G, Abdelhady HA, Oumar Abakar A, Nassar ST. Comparative Efficacy and Safety of Novel Antiplatelets and Standard Therapy in Patients With Coronary Artery Disease. Cureus 2024; 16:e71333. [PMID: 39534810 PMCID: PMC11554594 DOI: 10.7759/cureus.71333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Coronary artery disease (CAD) is a significant health concern that has affected approximately 110 million people worldwide. CAD is defined as persistent narrowing of the coronary arteries as a result of atherosclerotic plaque build-up. Acute coronary syndrome (ACS), which encompasses ST-elevation myocardial infarction (STEMI), non-ST-elevation myocardial infarction (NSTEMI), and unstable angina, often results from plaque ruptures. Platelets are crucial for atherogenesis, vascular inflammation, and oxidative stress. Antiplatelet therapy aimed at reducing thrombotic events is vital for ACS treatment. Clinical guidelines advise the use of dual antiplatelet therapy (DAPT) that combines aspirin and a P2Y12 receptor inhibitor (clopidogrel, prasugrel, or ticagrelor) in ACS patients undergoing percutaneous intervention (PCI). This study aimed to assess comprehensively the effectiveness and safety of ticagrelor and prasugrel in comparison to clopidogrel in patients with ACS. An extensive literature search was conducted using PubMed, PubMed Central (PMC), ScienceDirect, and EBSCO databases. The search revealed studies that compared ticagrelor and prasugrel to clopidogrel in ACS patients, and we selected these studies based on specific inclusion and exclusion criteria, which included observational studies, clinical trials, literature reviews, and meta-analyses involving adult ACS patients treated with ticagrelor, prasugrel, or clopidogrel. The efficacy outcomes were defined as major adverse cardiovascular events (MACE) and thrombotic events, whereas the safety outcomes were measured by major and minor bleeding and hemorrhagic stroke. After a rigorous quality assessment to minimize bias, 23 studies were selected for analysis. The findings indicated that novel antiplatelets reduced MACE but increased bleeding complications, with ticagrelor consistently associated with dyspnea. In conclusion, novel P2Y12 inhibitors provide cardiovascular benefits but require careful patient selection and monitoring due to gastrointestinal bleeding (GIB) risks. Future research should standardize bleeding definitions and assess long-term outcomes. Ticagrelor and prasugrel may be more effective and safer than clopidogrel in ACS patients. Given the high risk of GIB, especially among older individuals or those with a past stroke, it is advisable to suggest a lower prasugrel dose without raising the bleeding rates. Since fewer patients use the novel antiplatelet regimen compared to clopidogrel, future clinical trials should include a broader patient population and compare these regimens.
Collapse
Affiliation(s)
| | - Sayed A Mahmud
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Anura Manandhar
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Ghadeer Sabir
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Hala A Abdelhady
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Adoum Oumar Abakar
- Internal Medicine and Clinical Research, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Sondos T Nassar
- Medicine and Surgery, Jordan University of Science and Technology, Amman, JOR
| |
Collapse
|
27
|
Ye W, Shen B, Tang Q, Fang C, Wang L, Xie L, He Q. Identification of a novel immune infiltration-related gene signature, MCEMP1, for coronary artery disease. PeerJ 2024; 12:e18135. [PMID: 39346078 PMCID: PMC11438437 DOI: 10.7717/peerj.18135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024] Open
Abstract
Background This study aims to identify a novel gene signature for coronary artery disease (CAD), explore the role of immune cell infiltration in CAD pathogenesis, and assess the cell function of mast cell-expressed membrane protein 1 (MCEMP1) in human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL). Methods To identify differentially expressed genes (DEGs) of CAD, datasets GSE24519 and GSE61145 were downloaded from the Gene Expression Omnibus (GEO) database using the R "limma" package with p < 0.05 and |log2 FC| > 1. Gene ontology (GO) and pathway analyses were conducted to determine the biological functions of DEGs. Hub genes were identified using support vector machine-recursive feature elimination (SVM-RFE) and least absolute shrinkage and selection operator (LASSO). The expression levels of these hub genes in CAD were validated using the GSE113079 dataset. CIBERSORT program was used to quantify the proportion of immune cell infiltration. Western blot assay and qRT-PCR were used to detect the expression of hub genes in ox-LDL-treated HUVECs to validate the bioinformatics results. Knockdown interference sequences for MCEMP1 were synthesized, and cell proliferation and apoptosis were examined using a CCK8 kit and Muse® Cell Analyzer, respectively. The concentrations of IL-1β, IL-6, and TNF-α were measured with respective enzyme-linked immunosorbent assay (ELISA) kits. Results A total of 73 DEGs (four down-regulated genes and 69 up-regulated genes) were identified in the metadata (GSE24519 and GSE61145) cohort. GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis results indicated that these DEGs might be associated with the regulation of platelet aggregation, defense response or response to bacterium, NF-kappa B signaling pathway, and lipid and atherosclerosis. Using SVM-RFE and LASSO, seven hub genes were obtained from the metadata. The upregulated expression of DIRC2 and MCEMP1 in CAD was confirmed in the GSE113079 dataset and in ox-LDL-treated HUVECs. The associations between the two hub genes (DIRC2 and MCEMP1) and the 22 types of immune cell infiltrates in CAD were found. MCEMP1 knockdown accelerated cell proliferation and suppressed cell apoptosis for ox-LDL-treated HUVECs. Additionally, MCEMP1 knockdown appeared to decrease the expression of inflammatory factors IL-1β, IL-6, and TNF-α. Conclusions The results of this study indicate that MCEMP1 may play an important role in CAD pathophysiology.
Collapse
Affiliation(s)
- Wei Ye
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China
| | - Chengzhi Fang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Cardiology, HanChuan Hospital, Hanchuan, China
| | - Lili Xie
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qi He
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Ye H, Chen Z, Li K, Zhang Y, Li H, Tian N. Non-linear association of the platelet/high-density lipoprotein cholesterol ratio with bone mineral density a cross-sectional study. Lipids Health Dis 2024; 23:300. [PMID: 39285435 PMCID: PMC11403790 DOI: 10.1186/s12944-024-02291-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Numerous studies have demonstrated shared risk factors and pathophysiologic mechanisms between osteoporosis and cardiovascular disease. High-density lipoprotein cholesterol (HDL-C) and platelets have long been recognized as crucial factors for cardiovascular health. The platelet to HDL-C ratio (PHR) combines platelet count and high-density lipoprotein cholesterol (HDL-C) level, It is a novel biomarker for metabolic syndrome and cardiovascular disease. The platelet to HDL-C ratio (PHR) possibly reflects the balance between proinflammatory and anti-inflammatory states in the body. Therefore, we hypothesized that changes in PHR ratios may predict a predisposition to pro-inflammatory and increased bone resorption. However, the relationship between the platelet to HDL-C ratio (PHR) and bone mineral density (BMD) remains insufficiently understood. This study aimed to elucidate the relationship between the platelet to HDL-C ratio (PHR) index and bone mineral density (BMD). METHODS Data from the NHANES 2005-2018 were analyzed, excluding adults with missing key variables and specific conditions. Nonlinear relationships were explored by fitting smoothed curves and generalized additive models, with threshold effects employed to calculate inflection points. Additionally, subgroup analyses and interaction tests were conducted. RESULTS The study included 13,936 individuals with a mean age of 51.19 ± 16.65 years. Fitted smoothed curves and generalized additive models revealed a nonlinear, inverted U-shaped relationship between the two variables. Threshold effect analysis showed a significant negative association between PHR and total femur bone mineral density (BMD) beyond the inflection point of platelet to HDL-C ratio (PHR) 33.301. Subgroup analyses showed that a significant interaction between these two variables was observed only in the age and sex subgroups (P-interaction < 0.05). CONCLUSIONS Our study identified a complex, nonlinear, inverted U-shaped relationship between platelet to HDL-C ratio (PHR) and total femur bone mineral density (BMD). These findings underscore the importance of maintaining optimal PHR levels to support bone health, especially in high-risk populations.
Collapse
Affiliation(s)
- Haobo Ye
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Yuying Children Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zihao Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Yuying Children Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Kaiyu Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Yuying Children Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yekai Zhang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Yuying Children Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hualin Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Yuying Children Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Naifeng Tian
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Yuying Children Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
29
|
Liu Y, Wang Z, Fang L, Xu Y, Zhao B, Kang X, Zhao Y, Han J, Zhang Y, Dong E, Wang N. Deficiency of 5-HT 2B receptors alleviates atherosclerosis by regulating macrophage phenotype through inhibiting interferon signalling. Br J Pharmacol 2024. [PMID: 39232850 DOI: 10.1111/bph.17315] [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: 12/30/2023] [Revised: 06/09/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND AND PURPOSE Elevated levels of 5-HT have been correlated with coronary artery disease and cardiac events, suggesting 5-HT is a potential novel factor in the development of atherosclerotic cardiovascular disease. However, the underlying pathological mechanisms of the 5-HT system in atherosclerosis remain unclear. The 5-HT2B receptor (5-HT2BR), which establishes a positive feedback loop with 5-HT, has been identified as a contributor to pathophysiological processes in various vascular disorders. In this study, we investigated the immunological impact of 5-HT2BR in atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice. EXPERIMENTAL APPROACH Plasma levels of 5-HT were measured in mice using an ELISA kit. Atherosclerotic plaque formation, macrophage infiltration and inflammatory signalling were assessed in ApoE-/- mice by employing both pharmacological inhibition and genetic deficiency of 5-HT2BR. Inflammasome activation was elucidated using peritoneal macrophages isolated from 5-HT2BR-deficient mice. KEY RESULTS An upregulation of 5-HT2BR expression was observed in the aortas of ApoE-/- mice, exhibiting a strong correlation with the presence of macrophages in plaques. Atherosclerosis was attenuated in mice through pharmacological inhibition and genetic deficiency of 5-HT2BR. Additionally, a significant reduction in atherosclerotic plaque size was achieved through bone marrow reconstitution with 5-HT2BR-deficient cells. 5-HT2BR-deficient macrophages showed attenuated interferon (IFN) signalling, NLRP3 inflammasome activation, and interleukin-1β release. Moreover, macrophages primed with 5-HT2BR deficiency displayed an anti-inflammatory phenotype. CONCLUSION AND IMPLICATIONS These findings support the hypothesis that 5-HT2BR in macrophages plays a causal role in the development of atherosclerosis, revealing a novel perspective for potential therapeutic strategies in atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Yahan Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhipeng Wang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Fang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yaohua Xu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Beilei Zhao
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xuya Kang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yanqing Zhao
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Jintao Han
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Institute of Cardiovascular Diseases, The first affiliated Hospital of Dalian Medical University, Dalian, China
| | - Erdan Dong
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital); School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Nanping Wang
- Wuhu Hospital, East China Normal University (ECNU), Wuhu, China
- East China Normal University Health Science Center, Shanghai, China
| |
Collapse
|
30
|
Martinez Bravo G, Annarapu G, Carmona E, Nawarskas J, Clark R, Novelli E, Mota Alvidrez RI. Platelets in Thrombosis and Atherosclerosis: A Double-Edged Sword. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1608-1621. [PMID: 38885926 PMCID: PMC11373056 DOI: 10.1016/j.ajpath.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
This review focuses on the dual role of platelets in atherosclerosis and thrombosis, exploring their involvement in inflammation, angiogenesis, and plaque formation, as well as their hemostatic and prothrombotic functions. Beyond their thrombotic functions, platelets engage in complex interactions with diverse cell types, influencing disease resolution and progression. The contribution of platelet degranulation helps in the formation of atheromatous plaque, whereas the reciprocal interaction with monocytes adds complexity. Alterations in platelet membrane receptors and signaling cascades contribute to advanced atherosclerosis, culminating in atherothrombotic events. Understanding these multifaceted roles of platelets will lead to the development of targeted antiplatelet strategies for effective cardiovascular disease prevention and treatment. Understanding platelet functions in atherosclerosis and atherothrombosis at different stages of disease will be critical for designing targeted treatments and medications to prevent or cure the disease Through this understanding, platelets can be targeted at specific times in the atherosclerosis process, possibly preventing the development of atherothrombosis.
Collapse
Affiliation(s)
| | - Gowtham Annarapu
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Emely Carmona
- School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - James Nawarskas
- Pharmaceutical Sciences-Pharmacy Practice, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | - Ross Clark
- Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico; Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico
| | - Enrico Novelli
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania; School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Roberto I Mota Alvidrez
- Biomedical Engineering Department, University of New Mexico, Albuquerque, New Mexico; Pharmaceutical Sciences-Pharmacy Practice, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico; Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico.
| |
Collapse
|
31
|
Mueller KAL, Langnau C, Harm T, Sigle M, Mott K, Droppa M, Borst O, Rohlfing AK, Gekeler S, Günter M, Goebel N, Franke UF, Radwan M, Schlensak C, Janning H, Scheuermann S, Seitz CM, Rath D, Kreisselmeier KP, Castor T, Mueller II, Schulze H, Autenrieth SE, Gawaz MP. Macrophage Migration Inhibitory Factor Promotes Thromboinflammation and Predicts Fast Progression of Aortic Stenosis. Arterioscler Thromb Vasc Biol 2024; 44:2118-2135. [PMID: 38989580 PMCID: PMC11335082 DOI: 10.1161/atvbaha.124.321000] [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: 03/25/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Aortic stenosis (AS) is driven by progressive inflammatory and fibrocalcific processes regulated by circulating inflammatory and valve resident endothelial and interstitial cells. The impact of platelets, platelet-derived mediators, and platelet-monocyte interactions on the acceleration of local valvular inflammation and mineralization is presently unknown. METHODS We prospectively enrolled 475 consecutive patients with severe symptomatic AS undergoing aortic valve replacement. Clinical workup included repetitive echocardiography, analysis of platelets, monocytes, chemokine profiling, aortic valve tissue samples for immunohistochemistry, and gene expression analysis. RESULTS The patients were classified as fast-progressive AS by the median ∆Vmax of 0.45 m/s per year determined by echocardiography. Immunohistological aortic valve analysis revealed enhanced cellularity in fast-progressive AS (slow- versus fast-progressive AS; median [interquartile range], 247 [142.3-504] versus 717.5 [360.5-1234]; P<0.001) with less calcification (calcification area, mm2: 33.74 [27.82-41.86] versus 20.54 [13.52-33.41]; P<0.001). MIF (macrophage migration inhibitory factor)-associated gene expression was significantly enhanced in fast-progressive AS accompanied by significantly elevated MIF plasma levels (mean±SEM; 6877±379.1 versus 9959±749.1; P<0.001), increased platelet activation, and decreased intracellular MIF expression indicating enhanced MIF release upon platelet activation (CD62P, %: median [interquartile range], 16.8 [11.58-23.8] versus 20.55 [12.48-32.28], P=0.005; MIF, %: 4.85 [1.48-9.75] versus 2.3 [0.78-5.9], P<0.001). Regression analysis confirmed that MIF-associated biomarkers are strongly associated with an accelerated course of AS. CONCLUSIONS Our findings suggest a key role for platelet-derived MIF and its interplay with circulating and valve resident monocytes/macrophages in local and systemic thromboinflammation during accelerated AS. MIF-based biomarkers predict an accelerated course of AS and represent a novel pharmacological target to attenuate progression of AS.
Collapse
Affiliation(s)
- Karin Anne Lydia Mueller
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Carolin Langnau
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Kristina Mott
- Institute for Experimental Biomedicine, Chair I University Hospital Würzburg, Germany (K.M., H.S.)
| | - Michal Droppa
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
- DFG Heisenberg Group Thrombocardiology (O.B.), University of Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Sarah Gekeler
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Manina Günter
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology (M.G., S.E.A.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
- Dendritic Cells in Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany (M.G., S.E.A.)
| | - Nora Goebel
- Robert-Bosch Hospital, Department of Cardiovascular Surgery, Stuttgart, Germany (N.G., U.F.W.F.)
| | - Ulrich F.W. Franke
- Robert-Bosch Hospital, Department of Cardiovascular Surgery, Stuttgart, Germany (N.G., U.F.W.F.)
| | - Medhat Radwan
- Department of Thoracic and Cardiovascular Surgery (M.R., C.S.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery (M.R., C.S.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Henrik Janning
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Sophia Scheuermann
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies (S.S., C.M.S.), University of Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Tuebingen, Germany (S.S., C.M.S.)
| | - Christian M. Seitz
- Cluster of Excellence iFIT (EXC 2180) Image-Guided and Functionally Instructed Tumor Therapies (S.S., C.M.S.), University of Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Tuebingen, Germany (S.S., C.M.S.)
| | - Dominik Rath
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Klaus-Peter Kreisselmeier
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Iris Irmgard Mueller
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| | - Harald Schulze
- Institute for Experimental Biomedicine, Chair I University Hospital Würzburg, Germany (K.M., H.S.)
| | - Stella E. Autenrieth
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology (M.G., S.E.A.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
- Dendritic Cells in Infection and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany (M.G., S.E.A.)
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology (K.A.L.M., C.L., T.H., M.S., M.D., O.B., A.-K.R., S.G., H.J., D.R., K.-P.K., T.C., I.I.M., M.P.G.), University Hospital Tuebingen, Eberhard Karls University Tuebingen, Germany
| |
Collapse
|
32
|
Zhou ZF, Hu CF, Gao MF, Jin X, Chen Y, Wang D, Shen CX, Liu L. The predictive value of serum fibrinogen and platelet distribution width for long-term cardiac death in acute myocardial infarction patients. J Thorac Dis 2024; 16:5073-5085. [PMID: 39268112 PMCID: PMC11388258 DOI: 10.21037/jtd-24-204] [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: 02/03/2024] [Accepted: 06/24/2024] [Indexed: 09/15/2024]
Abstract
Background Acute myocardial infarction (AMI) is the most severe manifestation of coronary heart disease (CHD), and timely and effective opening of the culprit vessels has been effective in reducing its mortality, but long-term death still threatens the life of patients. Therefore, finding biomarkers to predict death post-myocardial infarction (MI) is crucial. The aim of our study is to find biomarkers that predicted long-term death in Chinese AMI patients. Methods This retrospective analysis included patients with AMI from 1 January 2017 to 30 September 2019. All patients were followed up at least 4 years. Propensity score matching was used to mitigate the influence of nonrandom selection in MI-survival and MI-death groups. Cox analysis was applied for analyzing the risk factors of death post-MI. The receiver operating characteristic (ROC) curve was used to analyze the predictive value of biomarkers. Results Of the 1,059 AMI patients analyzed, 130 died during follow-up. After propensity matching, there were 116 patients in each of the two groups. In addition to the traditional risk factors for long-term death post-MI, two important risk factors platelet distribution width (PDW) [hazard ratio (HR) =1.210, 95% confidence interval (CI): 1.080-1.356, P=0.001] and fibrinogen (HR =1.218, 95% CI: 1.027-1.444, P=0.02) were found. The area under the curve (AUC) of PDW and fibrinogen was 0.604 (P=0.007) and 0.684 (P<0.001) respectively. The optimal thresholds were 13.05% and 3.562 g/L respectively. Conclusions PDW and fibrinogen seem to be useful as biomarkers for long-term death prediction post-MI. The current research provides new insight into the prevention and treatment of death in Chinese patients post-MI.
Collapse
Affiliation(s)
- Zhen-Fa Zhou
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cui-Fen Hu
- Department of Ultrasound in Medicine, Minhang Hospital, Fudan University, Shanghai, China
| | - Mei-Fang Gao
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xian Jin
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Chen
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Wang
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Xing Shen
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Liu
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Băghină RM, Crișan S, Luca S, Pătru O, Lazăr MA, Văcărescu C, Negru AG, Luca CT, Gaiță D. Association between Inflammation and New-Onset Atrial Fibrillation in Acute Coronary Syndromes. J Clin Med 2024; 13:5088. [PMID: 39274304 PMCID: PMC11396258 DOI: 10.3390/jcm13175088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
Acute coronary syndrome (ACS) is a complex clinical syndrome that encompasses acute myocardial infarction (AMI) and unstable angina (UA). Its underlying mechanism refers to coronary plaque disruption, with consequent platelet aggregation and thrombosis. Inflammation plays an important role in the progression of atherosclerosis by mediating the removal of necrotic tissue following myocardial infarction and shaping the repair processes that are essential for the recovery process after ACS. As a chronic inflammatory disorder, atherosclerosis is characterized by dysfunctional immune inflammation involving interactions between immune (macrophages, T lymphocytes, and monocytes) and vascular cells (endothelial cells and smooth muscle cells). New-onset atrial fibrillation (NOAF) is one of the most common arrhythmic complications in the setting of acute coronary syndromes, especially in the early stages, when the myocardial inflammatory reaction is at its maximum. The main changes in the atrial substrate are due to atrial ischemia and acute infarcts that can be attributed to neurohormonal factors. The high incidence of atrial fibrillation (AF) post-myocardial infarction may be secondary to inflammation. Inflammatory response and immune system cells have been involved in the initiation and development of atrial fibrillation. Several inflammatory indexes, such as C-reactive protein and interleukins, have been demonstrated to be predictive of prognosis in patients with ACS. The cell signaling activation patterns associated with fibrosis, apoptosis, and hypertrophy are forms of cardiac remodeling that occur at the atrial level, predisposing to AF. According to a recent study, the presence of fibrosis and lymphomononuclear infiltration in the atrial tissue was associated with a prior history of AF. However, inflammation may contribute to both the occurrence/maintenance of AF and its thromboembolic complications.
Collapse
Affiliation(s)
- Ruxandra-Maria Băghină
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Simina Crișan
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Silvia Luca
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Oana Pătru
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Mihai-Andrei Lazăr
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Cristina Văcărescu
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Alina Gabriela Negru
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Constantin-Tudor Luca
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Dan Gaiță
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
- Research Center of the Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| |
Collapse
|
34
|
Yagin FH, Aygun U, Algarni A, Colak C, Al-Hashem F, Ardigò LP. Platelet Metabolites as Candidate Biomarkers in Sepsis Diagnosis and Management Using the Proposed Explainable Artificial Intelligence Approach. J Clin Med 2024; 13:5002. [PMID: 39274215 PMCID: PMC11395774 DOI: 10.3390/jcm13175002] [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: 08/01/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Background: Sepsis is characterized by an atypical immune response to infection and is a dangerous health problem leading to significant mortality. Current diagnostic methods exhibit insufficient sensitivity and specificity and require the discovery of precise biomarkers for the early diagnosis and treatment of sepsis. Platelets, known for their hemostatic abilities, also play an important role in immunological responses. This study aims to develop a model integrating machine learning and explainable artificial intelligence (XAI) to identify novel platelet metabolomics markers of sepsis. Methods: A total of 39 participants, 25 diagnosed with sepsis and 14 control subjects, were included in the study. The profiles of platelet metabolites were analyzed using quantitative 1H-nuclear magnetic resonance (NMR) technology. Data were processed using the synthetic minority oversampling method (SMOTE)-Tomek to address the issue of class imbalance. In addition, missing data were filled using a technique based on random forests. Three machine learning models, namely extreme gradient boosting (XGBoost), light gradient boosting machine (LightGBM), and kernel tree boosting (KTBoost), were used for sepsis prediction. The models were validated using cross-validation. Clinical annotations of the optimal sepsis prediction model were analyzed using SHapley Additive exPlanations (SHAP), an XAI technique. Results: The results showed that the KTBoost model (0.900 accuracy and 0.943 AUC) achieved better performance than the other models in sepsis diagnosis. SHAP results revealed that metabolites such as carnitine, glutamate, and myo-inositol are important biomarkers in sepsis prediction and intuitively explained the prediction decisions of the model. Conclusion: Platelet metabolites identified by the KTBoost model and XAI have significant potential for the early diagnosis and monitoring of sepsis and improving patient outcomes.
Collapse
Affiliation(s)
- Fatma Hilal Yagin
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, Malatya 44280, Türkiye
| | - Umran Aygun
- Department of Anesthesiology and Reanimation, Malatya Yesilyurt Hasan Calık State Hospital, Malatya 44929, Türkiye
| | - Abdulmohsen Algarni
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia
| | - Cemil Colak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, Malatya 44280, Türkiye
| | - Fahaid Al-Hashem
- Department of Physiology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Luca Paolo Ardigò
- Department of Teacher Education, NLA University College, 0166 Oslo, Norway
| |
Collapse
|
35
|
Berger JS, Cornwell MG, Xia Y, Muller MA, Smilowitz NR, Newman JD, Schlamp F, Rockman CB, Ruggles KV, Voora D, Hochman JS, Barrett TJ. A Platelet Reactivity ExpreSsion Score derived from patients with peripheral artery disease predicts cardiovascular risk. Nat Commun 2024; 15:6902. [PMID: 39164233 PMCID: PMC11336089 DOI: 10.1038/s41467-024-50994-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 07/22/2024] [Indexed: 08/22/2024] Open
Abstract
Platelets are key mediators of atherothrombosis, yet, limited tools exist to identify individuals with a hyperreactive platelet phenotype. In this study, we investigate the association of platelet hyperreactivity and cardiovascular events, and introduce a tool, the Platelet Reactivity ExpreSsion Score (PRESS), which integrates platelet aggregation responses and RNA sequencing. Among patients with peripheral artery disease (PAD), those with a hyperreactive platelet response (>60% aggregation) to 0.4 µM epinephrine had a higher incidence of the 30 day primary cardiovascular endpoint (37.2% vs. 15.3% in those without hyperreactivity, adjusted HR 2.76, 95% CI 1.5-5.1, p = 0.002). PRESS performs well in identifying a hyperreactive phenotype in patients with PAD (AUC [cross-validation] 0.81, 95% CI 0.68 -0.94, n = 84) and in an independent cohort of healthy participants (AUC [validation] 0.77, 95% CI 0.75 -0.79, n = 35). Following multivariable adjustment, PAD individuals with a PRESS score above the median are at higher risk for a future cardiovascular event (adjusted HR 1.90, CI 1.07-3.36; p = 0.027, n = 129, NCT02106429). This study derives and validates the ability of PRESS to discriminate platelet hyperreactivity and identify those at increased cardiovascular risk. Future studies in a larger independent cohort are warranted for further validation. The development of a platelet reactivity expression score opens the possibility for a personalized approach to antithrombotic therapy for cardiovascular risk reduction.
Collapse
Affiliation(s)
- Jeffrey S Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Surgery, New York University Langone Health, New York, NY, USA.
| | - Macintosh G Cornwell
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA
| | - Yuhe Xia
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Matthew A Muller
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA
| | - Nathaniel R Smilowitz
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Cardiology Section, Department of Medicine, Veterans Affairs New York Harbor Health Care System, New York, NY, USA
| | - Jonathan D Newman
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Florencia Schlamp
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Caron B Rockman
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Kelly V Ruggles
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA
| | - Deepak Voora
- Department of Medicine, Duke Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Judith S Hochman
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Tessa J Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
36
|
Wang Q, Zhang G. Platelet count as a prognostic marker for acute respiratory distress syndrome. BMC Pulm Med 2024; 24:396. [PMID: 39153980 PMCID: PMC11330071 DOI: 10.1186/s12890-024-03204-5] [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: 01/17/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND This study aimed to evaluate the role of platelet count (PLT) in the prognosis of patients with acute respiratory distress syndrome (ARDS). METHODS The data were extracted from the Medical Information Mart for Intensive Care database (version 2.2). Patients diagnosed with ARDS according to criteria from Berlin Definition and had the platelet count (PLT) measured within the first day after intensive care unit admission were analyzed. Based on PLT, ARDS patients were divided into four groups: PLT ≤ 100 × 109/L, PLT 101-200 × 109/L, PLT 201-300 × 109/L, and PLT > 300 × 109/L. The primary outcome was 28-day mortality. Survival probabilities were analyzed using Kaplan-Meier. Furthermore, the association between PLT and mortality in ARDS patients was assessed using a univariate and multivariable Cox proportional hazards model. RESULTS Overall, the final analysis included 3,207 eligible participants with ARDS. According to the Kaplan-Meier curves for 28-day mortality of PLT, PLT ≤ 100 × 109/L was associated with a higher incidence of mortality (P = 0.001), the same trends were observed in the 60-day (P = 0.001) and 90-day mortality (P = 0.001). In the multivariate model adjusted for the potential factors, the adjusted hazard ratio at PLT 101-200 × 109/L group, PLT 201-300 × 109/L, and PLT > 300 × 109/L was 0.681 [95% confidence interval (CI): 0.576-0.805, P < 0.001], 0.733 (95% CI: 0.604-0.889, P = 0.002), and 0.787 (95% CI: 0.624-0.994, P = 0.044) compared to the reference group (PLT ≤ 100 × 109/L), respectively. Similar relationships between the PLT ≤ 100 × 109/L group and 28-day mortality were obtained in most subgroups. CONCLUSION PLT appeared to be an independent predictor of mortality in critically ill patients with ARDS.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Intensive care unit, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hang Zhou, Zhe Jiang, 310000, China, No 3 East Road Qingchun
| | - Ge Zhang
- Department of Intensive care unit, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hang Zhou, Zhe Jiang, 310000, China, No 3 East Road Qingchun.
| |
Collapse
|
37
|
Kou H, Yang H. Molecular imaging nanoprobes and their applications in atherosclerosis diagnosis. Theranostics 2024; 14:4747-4772. [PMID: 39239513 PMCID: PMC11373619 DOI: 10.7150/thno.96037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/06/2024] [Indexed: 09/07/2024] Open
Abstract
Molecular imaging has undergone significant development in recent years for its excellent ability to image and quantify biologic processes at cellular and molecular levels. Its application is of significance in cardiovascular diseases, particularly in diagnosing them at early stages. Atherosclerosis is a complex, chronic, and progressive disease that can lead to serious consequences such as heart strokes or infarctions. Attempts have been made to detect atherosclerosis with molecular imaging modalities. Not only do imaging modalities develop rapidly, but research of relevant nanomaterials as imaging probes has also been increasingly studied in recent years. This review focuses on the latest developments in the design and synthesis of probes that can be utilized in computed tomography, positron emission tomography, magnetic resonance imaging, ultrasound imaging, photoacoustic imaging and combined modalities. The challenges and future developments of nanomaterials for molecular imaging modalities are also discussed.
Collapse
Affiliation(s)
| | - Hu Yang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, MO 65409, United States
| |
Collapse
|
38
|
An JY, Ma XN, Wen HL, Hu HD. Identification of key genes and long non‑coding RNA expression profiles in osteoporosis with rheumatoid arthritis based on bioinformatics analysis. BMC Musculoskelet Disord 2024; 25:634. [PMID: 39118036 PMCID: PMC11312199 DOI: 10.1186/s12891-024-07738-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Although rheumatoid arthritis (RA) is a chronic systemic tissue disease often accompanied by osteoporosis (OP), the molecular mechanisms underlying this association remain unclear. This study aimed to elucidate the pathogenesis of RA and OP by identifying differentially expressed mRNAs (DEmRNAs) and long non-coding RNAs (lncRNAs) using a bioinformatics approach. METHODS Expression profiles of individuals diagnosed with OP and RA were retrieved from the Gene Expression Omnibus database. Differential expression analysis was conducted. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) pathway enrichment analyses were performed to gain insights into the functional categories and molecular/biochemical pathways associated with DEmRNAs. We identified the intersection of common DEmRNAs and lncRNAs and constructed a protein-protein interaction (PPI) network. Correlation analysis between the common DEmRNAs and lncRNAs facilitated the construction of a coding-non-coding network. Lastly, serum peripheral blood mononuclear cells (PBMCs) from patients with RA and OP, as well as healthy controls, were obtained for TRAP staining and qRT-PCR to validate the findings obtained from the online dataset assessments. RESULTS A total of 28 DEmRNAs and 2 DElncRNAs were identified in individuals with both RA and OP. Chromosomal distribution analysis of the consensus DEmRNAs revealed that chromosome 1 had the highest number of differential expression genes. GO and KEGG analyses indicated that these DEmRNAs were primarily associated with " platelets (PLTs) degranulation", "platelet alpha granules", "platelet activation", "tight junctions" and "leukocyte transendothelial migration", with many genes functionally related to PLTs. In the PPI network, MT-ATP6 and PTGS1 emerged as potential hub genes, with MT-ATP6 originating from mitochondrial DNA. Co-expression analysis identified two key lncRNA-mRNA pairs: RP11 - 815J21.2 with MT - ATP6 and RP11 - 815J21.2 with PTGS1. Experimental validation confirmed significant differential expression of RP11-815J21.2, MT-ATP6 and PTGS1 between the healthy controls and the RA + OP groups. Notably, knockdown of RP11-815J21.2 attenuated TNF + IL-6-induced osteoclastogenesis. CONCLUSIONS This study successfully identified shared dysregulated genes and potential therapeutic targets in individuals with RA and OP, highlighting their molecular similarities. These findings provide new insights into the pathogenesis of RA and OP and suggest potential avenues for further research and targeted therapies.
Collapse
Affiliation(s)
- Jin-Yu An
- Department of Orthopedics, Changzhou Fourth People's Hospital, Changzhou, 213000, China.
| | - Xing-Na Ma
- Department of Pediatric, Changzhou Fourth People's Hospital, Changzhou, 213000, China
| | - Hui-Long Wen
- Department of Orthopedics, Changzhou Fourth People's Hospital, Changzhou, 213000, China
| | - Hui-Dong Hu
- Department of Orthopedics, Changzhou Fourth People's Hospital, Changzhou, 213000, China
| |
Collapse
|
39
|
Maaniitty E, Jalkanen J, Sinisilta S, Gunn J, Vasankari T, Biancari F, Jalkanen S, Airaksinen KEJ, Hollmen M, Kiviniemi T. Differential circulating cytokine profiles in acute coronary syndrome versus stable coronary artery disease. Sci Rep 2024; 14:17269. [PMID: 39068298 PMCID: PMC11283453 DOI: 10.1038/s41598-024-68333-7] [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: 01/09/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Chronic inflammation plays a crucial role in coronary artery disease (CAD), but differences in specific cytokine profiles between acute coronary syndrome (ACS) and stable CAD remain unknown. We investigated cytokine differences between these two manifestations of CAD. The study included 308 patients with angiographically detected, hemodynamically significant CAD: 150 patients undergone angiography for ACS, 158 patients undergone angiography for stable CAD. To assess dynamic changes, 116 patients had index angiogram at least 3 months earlier. We measured the serum concentrations of 48 circulating cytokines. The ACS group had decreased interleukin (IL) 4 (p = 0.005), and increased IL-8 (p = 0.008), hepatocyte growth factor (HGF) (p < 0.001) and macrophage colony-stimulating factor (M-CSF) (p = 0.002) levels compared with the stable CAD group. Multivariable logistic regression revealed increased levels of HGF (OR 18.050 [95% CI 4.372-74.517], p < 0.001), M-CSF (OR 2.257 [1.375-3.705], p = 0.001) and IL-6 (OR 1.586 [1.131-2.224], p = 0.007), independently associated with ACS. In the post-angiography group, only diminished platelet-derived growth factor-BB levels in ACS-manifested patients were observed (OR 0.478, [0.279-0.818], p = 0.007). Cytokine profiles differ between ACS and stable CAD. Such differences seem to be mainly reversible within 3 months after ACS. Thus, targeting one or two cytokines only might not offer one-size fits all-therapeutic approach for CAD-associated inflammation.Trial registration: NCT03444259.
Collapse
Affiliation(s)
- Eveliina Maaniitty
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland.
| | - Juho Jalkanen
- Vascular Surgery, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Sami Sinisilta
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Jarmo Gunn
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Tuija Vasankari
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Fausto Biancari
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
- Department of Medicine, South Karelia Central Hospital, University of Helsinki, Valto Käkelän Katu 1, 53130, Lappeenranta, Finland
| | - Sirpa Jalkanen
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - K E Juhani Airaksinen
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Maija Hollmen
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Tuomas Kiviniemi
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland.
| |
Collapse
|
40
|
Chee YJ, Dalan R. Novel Therapeutics for Type 2 Diabetes Mellitus-A Look at the Past Decade and a Glimpse into the Future. Biomedicines 2024; 12:1386. [PMID: 39061960 PMCID: PMC11274090 DOI: 10.3390/biomedicines12071386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024] Open
Abstract
Cardiovascular disease (CVD) and kidney disease are the main causes of morbidity and mortality in type 2 diabetes mellitus (T2DM). Globally, the incidence of T2DM continues to rise. A substantial increase in the burden of CVD and renal disease, alongside the socioeconomic implications, would be anticipated. Adopting a purely glucose-centric approach focusing only on glycemic targets is no longer adequate to mitigate the cardiovascular risks in T2DM. In the past decade, significant advancement has been achieved in expanding the pharmaceutical options for T2DM, with novel agents such as the sodium-glucose cotransporter type 2 (SGLT2) inhibitors and glucagon-like peptide receptor agonists (GLP-1 RAs) demonstrating robust evidence in cardiorenal protection. Combinatorial approaches comprising multiple pharmacotherapies combined in a single agent are an emerging and promising way to not only enhance patient adherence and improve glycemic control but also to achieve the potential synergistic effects for greater cardiorenal protection. In this review, we provide an update on the novel antidiabetic agents in the past decade, with an appraisal of the mechanisms contributing to cardiorenal protection. Additionally, we offer a glimpse into the landscape of T2DM management in the near future by providing a comprehensive summary of upcoming agents in early-phase trials.
Collapse
Affiliation(s)
- Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
41
|
Al-Nuaimi AMA. Role of hematological indices in predicting preeclampsia and its severity: retrospective case-control study. Medicine (Baltimore) 2024; 103:e38557. [PMID: 38905404 PMCID: PMC11192010 DOI: 10.1097/md.0000000000038557] [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: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/23/2024] Open
Abstract
Preeclampsia (PE) is a serious condition that threatens pregnancy with severe sequelae on both the mother and infant. Early detection of PE will lead to favorable outcomes, and using readily available markers like hematological indices is an attractive choice. Examine the diagnostic utility of hematological indices in pregnant women to predict preeclampsia and its severity. In a retrospective case-control study that included 252 women, all had their complete blood picture evaluated during their first and third trimesters as part of their outpatient antenatal care during their pregnancy. They were also divided into 3 groups: healthy pregnant women (control), non-severe PE, and severe PE, each involving 84 women. The changes in platelet to lymphocyte ratio (PLR) between 1st and 3rd trimesters showed an excellent ability to differentiate between severe PE and control (area under the curve = 0.954, cutoff ≤ -5.45%) and a good ability to differentiate between severe PE and non-severe PE (area under the curve = 0.841, cutoff ≤ -7.89%). Neutrophil to lymphocyte ratio showed a good to excellent ability to differentiate between severe PE and non-severe PE compared to control in the first and third trimesters and the percentage change between them. Changes in neutrophil to lymphocyte ratio and PLR strongly predict preeclampsia and its severity since they offer more predictive values than measuring NLP and PLR at different stages of pregnancy individually.
Collapse
|
42
|
Serban D, Papanas N, Dascalu AM, Kempler P, Raz I, Rizvi AA, Rizzo M, Tudor C, Silviu Tudosie M, Tanasescu D, Pantea Stoian A, Gouveri E, Ovidiu Costea D. Significance of Neutrophil to Lymphocyte Ratio (NLR) and Platelet Lymphocyte Ratio (PLR) in Diabetic Foot Ulcer and Potential New Therapeutic Targets. INT J LOW EXTR WOUND 2024; 23:205-216. [PMID: 34791913 DOI: 10.1177/15347346211057742] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Diabetic foot ulcer (DFU) is a well-known complication of diabetes and a significant burden on the national health systems. The neutrophil/lymphocyte ratio (NLR) and platelet/lymphocyte ratio are inexpensive and easily accessible biomarkers that have proved to be useful in several inflammatory, infectious and cardiovascular diseases. We carried out a comprehensive review examining the association of NLR and PLR with the onset and progression of DFU. PLR and NLR were significantly increased in patients with DFU, compared with a control group of T2DM patients without DFU, and correlate well with DFU severity, evaluated by Wagner and IWGDF grading scales. In patients with diabetic foot infections (DFI), elevated NLR and PLR were correlated with osteomyelitis, increased risk of amputation, and septic complications. The significance of the elevated value of these biomarkers in DFU is related to chronic hyperglycemia and low-grade systemic inflammation, atherosclerotic and vascular complications, and also the associated septic factor. Serial, dynamic follow-up can provide useful information in planning and monitoring DFU treatment, as well as in risk stratification of these vulnerable patients. Further randomized studies are needed to set the cut-off values with clinical significance.
Collapse
Affiliation(s)
- Dragos Serban
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania, ; ;
- Fourth Department of General Surgery, Emergency University Hospital, Romania,
| | - Nikolaos Papanas
- Diabetes Centre-Diabetic Foot Clinic, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece,
| | - Ana Maria Dascalu
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania, ; ;
- Department of Ophthalmology, Emergency University Hospital Bucharest, Romania
| | | | | | - Ali A Rizvi
- Emory University, Atlanta, GA, USA,
- University of South Carolina School of Medicine Columbia, SC, USA
| | - Manfredi Rizzo
- University of South Carolina School of Medicine Columbia, SC, USA
- Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy,
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania,
| | - Corneliu Tudor
- Fourth Department of General Surgery, Emergency University Hospital, Romania,
| | - Mihail Silviu Tudosie
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania, ; ;
- Clinical Emergency Hospital, Bucharest, Romania
| | | | | | | | - Daniel Ovidiu Costea
- Ovidius University, Constanta, Romania,
- Emergency County Hospital, Constanta, Romania
| |
Collapse
|
43
|
Yaşan M, Özel R, Yildiz A, Savaş G, Korkmaz A. The predictive value of systemic immune-inflammation index for long-term cardiovascular mortality in non-ST segment elevation myocardial infarction. Coron Artery Dis 2024; 35:179-185. [PMID: 38451553 DOI: 10.1097/mca.0000000000001355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
BACKGROUND Increased levels of inflammatory markers have been found in association with the severity of coronary atherosclerosis. Systemic immuneinflammation index (SII), which is calculated by multiplying neutrophil and platelet counts and then dividing the result by the lymphocyte count, can also be used as a prognostic indicator in different cardiovascular diseases. In this study, we investigated SII levels and long-term mortality of patients with non-ST segment elevation myocardial infarction (NSTEMI). METHODS This is an observational, single-center study. Two hundred-eight patients who underwent coronary angiography for NSTEMI were included in the study. Patients were divided into 3 tertiles based on SII levels. We researched the relationship between level level and 1, 3 and 5 years mortality (NSTEMI). RESULTS One-year mortality of the patients was significantly higher among patients in the upper SII tertile when compared with the lower and middle SII tertile groups [11 (15.9%) vs. 2 (2.9%) and 6 (8.7%); P = 0.008, P = 0.195, respectively). Three-year mortality of the patients was significantly higher among patients in the upper SII tertile when compared with the lower and middle SII tertile groups [21 (30.4%) vs. 5 (7.1%) and 12 (17.4%); P < 0.001, P = 0.072, respectively). Five-year mortality of the patients was significantly higher among patients in the upper SII tertile when compared with the lower and middle SII tertile groups [26 (37.7%) vs. 8 (11.4%) and 15 (21.7%); P < 0.001, P = 0.040, respectively). CONCLUSION Our study showed that NSTEMI patients with higher SII had worse long-term mortality.
Collapse
Affiliation(s)
- Mustafa Yaşan
- Department of Cardiology, Kastamonu Training and Research Hospital, Kastamonu
| | - Ramime Özel
- Department of Cardiology, Kastamonu Training and Research Hospital, Kastamonu
| | - Abdulkadir Yildiz
- Department of Cardiology, Kastamonu Training and Research Hospital, Kastamonu
| | - Göktuğ Savaş
- Department of Cardiology, Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Education and Research Hospital, Istanbul
| | - Ahmet Korkmaz
- Department of Cardiology, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
44
|
Tian S, Wang Y, Wan J, Yang M, Fu Z. Co-stimulators CD40-CD40L, a potential immune-therapy target for atherosclerosis: A review. Medicine (Baltimore) 2024; 103:e37718. [PMID: 38579073 PMCID: PMC10994492 DOI: 10.1097/md.0000000000037718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/04/2024] [Indexed: 04/07/2024] Open
Abstract
The interaction between CD40 and CD40 ligand (CD40L) a crucial co-stimulatory signal for activating adaptive immune cells, has a noteworthy role in atherosclerosis. It is well-known that atherosclerosis is linked to immune inflammation in blood vessels. In atherosclerotic lesions, there is a multitude of proinflammatory cytokines, adhesion molecules, and collagen, as well as smooth muscle cells, macrophages, and T lymphocytes, particularly the binding of CD40 and CD40L. Therefore, research on inhibiting the CD40-CD40L system to prevent atherosclerosis has been ongoing for more than 30 years. However, it's essential to note that long-term direct suppression of CD40 or CD40L could potentially result in immunosuppression, emphasizing the critical role of the CD40-CD40L system in atherosclerosis. Thus, specifically targeting the CD40-CD40L interaction on particular cell types or their downstream signaling pathways may be a robust strategy for mitigating atherosclerosis, reducing potential side effects. This review aims to summarize the potential utility of the CD40-CD40L system as a viable therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Simeng Tian
- Department of Immunology, Basic Medicine College, Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University, Heilongjiang Academy of Medical Science, Harbin, China
- The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yufei Wang
- Department of Neurosurgery & Nursing Teaching and Research Office, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jie Wan
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mao Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenkun Fu
- Department of Immunology, Basic Medicine College, Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University, Heilongjiang Academy of Medical Science, Harbin, China
| |
Collapse
|
45
|
Boccatonda A, Del Cane L, Marola L, D’Ardes D, Lessiani G, di Gregorio N, Ferri C, Cipollone F, Serra C, Santilli F, Piscaglia F. Platelet, Antiplatelet Therapy and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Narrative Review. Life (Basel) 2024; 14:473. [PMID: 38672744 PMCID: PMC11051088 DOI: 10.3390/life14040473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is not only related to traditional cardiovascular risk factors like type 2 diabetes mellitus and obesity, but it is also an independent risk factor for the development of cardiovascular disease. MASLD has been shown to be independently related to endothelial dysfunction and atherosclerosis. MASLD is characterized by a chronic proinflammatory response that, in turn, may induce a prothrombotic state. Several mechanisms such as endothelial and platelet dysfunction, changes in the coagulative factors, lower fibrinolytic activity can contribute to induce the prothrombotic state. Platelets are players and addresses of metabolic dysregulation; obesity and insulin resistance are related to platelet hyperactivation. Furthermore, platelets can exert a direct effect on liver cells, particularly through the release of mediators from granules. Growing data in literature support the use of antiplatelet agent as a treatment for MASLD. The use of antiplatelets drugs seems to exert beneficial effects on hepatocellular carcinoma prevention in patients with MASLD, since platelets contribute to fibrosis progression and cancer development. This review aims to summarize the main data on the role of platelets in the pathogenesis of MASLD and its main complications such as cardiovascular events and the development of liver fibrosis. Furthermore, we will examine the role of antiplatelet therapy not only in the prevention and treatment of cardiovascular events but also as a possible anti-fibrotic and anti-tumor agent.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Internal Medicine, Bentivoglio Hospital, AUSL Bologna, 40010 Bentivoglio, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Lorenza Del Cane
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Lara Marola
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Damiano D’Ardes
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, 66100 Chieti, Italy (F.C.)
| | | | - Nicoletta di Gregorio
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Claudio Ferri
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Francesco Cipollone
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, 66100 Chieti, Italy (F.C.)
| | - Carla Serra
- Interventional, Diagnostic and Therapeutic Ultrasound Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Francesca Santilli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, 66100 Chieti, Italy;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
46
|
Seizer P, von Ungern-Sternberg SNI, Haug V, Dicenta V, Rosa A, Butt E, Nöthel M, Rohlfing AK, Sigle M, Nawroth PP, Nussbaum C, Sperandio M, Kusch C, Meub M, Sauer M, Münzer P, Bieber K, Stanger A, Mack AF, Huber R, Brand K, Lehners M, Feil R, Poso A, Krutzke K, Schäffer TE, Nieswandt B, Borst O, May AE, Zernecke A, Gawaz M, Heinzmann D. Cyclophilin A is a ligand for RAGE in thrombo-inflammation. Cardiovasc Res 2024; 120:385-402. [PMID: 38175781 DOI: 10.1093/cvr/cvad189] [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: 01/30/2023] [Revised: 10/08/2023] [Accepted: 10/20/2023] [Indexed: 01/06/2024] Open
Abstract
AIMS Cyclophilin A (CyPA) induces leucocyte recruitment and platelet activation upon release into the extracellular space. Extracellular CyPA therefore plays a critical role in immuno-inflammatory responses in tissue injury and thrombosis upon platelet activation. To date, CD147 (EMMPRIN) has been described as the primary receptor mediating extracellular effects of CyPA in platelets and leucocytes. The receptor for advanced glycation end products (RAGE) shares inflammatory and prothrombotic properties and has also been found to have similar ligands as CD147. In this study, we investigated the role of RAGE as a previously unknown interaction partner for CyPA. METHODS AND RESULTS Confocal imaging, proximity ligation, co-immunoprecipitation, and atomic force microscopy were performed and demonstrated an interaction of CyPA with RAGE on the cell surface. Static and dynamic cell adhesion and chemotaxis assays towards extracellular CyPA using human leucocytes and leucocytes from RAGE-deficient Ager-/- mice were conducted. Inhibition of RAGE abrogated CyPA-induced effects on leucocyte adhesion and chemotaxis in vitro. Accordingly, Ager-/- mice showed reduced leucocyte recruitment and endothelial adhesion towards CyPA in vivo. In wild-type mice, we observed a downregulation of RAGE on leucocytes when endogenous extracellular CyPA was reduced. We furthermore evaluated the role of RAGE for platelet activation and thrombus formation upon CyPA stimulation. CyPA-induced activation of platelets was found to be dependent on RAGE, as inhibition of RAGE, as well as platelets from Ager-/- mice showed a diminished activation and thrombus formation upon CyPA stimulation. CyPA-induced signalling through RAGE was found to involve central signalling pathways including the adaptor protein MyD88, intracellular Ca2+ signalling, and NF-κB activation. CONCLUSION We propose RAGE as a hitherto unknown receptor for CyPA mediating leucocyte as well as platelet activation. The CyPA-RAGE interaction thus represents a novel mechanism in thrombo-inflammation.
Collapse
Affiliation(s)
- Peter Seizer
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- Department of Cardiology and Angiology, Ostalbklinikum Aalen, Aalen, Germany
| | - Saskia N I von Ungern-Sternberg
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Verena Haug
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Valerie Dicenta
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Annabelle Rosa
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Elke Butt
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Moritz Nöthel
- Department of Internal Medicine II, Cardiology, Pneumology, Angiology, University Hospital Bonn, Bonn, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Peter P Nawroth
- Department of Internal Medicine 1 and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
- Joint Heidelberg-ICD Translational Diabetes Program, Helmholtz-Zentrum, Munich, Germany
| | - Claudia Nussbaum
- Division of Neonatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians University Munich, Munich, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Charly Kusch
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Mara Meub
- Department of Biotechnology und Biophysics, Julius-Maximilians University, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology und Biophysics, Julius-Maximilians University, Würzburg, Germany
| | - Patrick Münzer
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Tübingen, Germany
| | - Kristin Bieber
- Department of Hematology, Oncology, Immunology und Pulmonology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anna Stanger
- Department of Hematology, Oncology, Immunology und Pulmonology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas F Mack
- Institute of Clinical Anatomy and Cell Analytics, Eberhard Karls University Tübingen, Tübingen, Germany
| | - René Huber
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Korbinian Brand
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Moritz Lehners
- Interfakultäres Institut für Biochemie, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Antti Poso
- Department of Internal Medicine VIII, University Hospital of Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls University, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Tübingen, Germany
- Excellence Cluster 'Controlling Microbes to Fight Infections' (CMFI), Tübingen, Germany
| | - Konstantin Krutzke
- Institute of Applied Physics, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tilman E Schäffer
- Institute of Applied Physics, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- DFG Heisenberg Group Cardiovascular Thromboinflammation and Translational Thrombocardiology, University of Tübingen, Tübingen, Germany
| | - Andreas E May
- Department of Cardiology, Innere Medizin I, Klinikum Memmingen, Memmingen, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - David Heinzmann
- Department of Cardiology and Angiology, Universitätsklinikum Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| |
Collapse
|
47
|
Jiang Y, Yu M, Song ZF, Wei ZY, Huang J, Qian HY. Targeted Delivery of Mesenchymal Stem Cell-Derived Bioinspired Exosome-Mimetic Nanovesicles with Platelet Membrane Fusion for Atherosclerotic Treatment. Int J Nanomedicine 2024; 19:2553-2571. [PMID: 38505171 PMCID: PMC10949310 DOI: 10.2147/ijn.s452824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/05/2024] [Indexed: 03/21/2024] Open
Abstract
Purpose Accumulating evidence indicates that mesenchymal stem cells (MSCs)-derived exosomes hold significant potential for the treatment of atherosclerosis. However, large-scale production and organ-specific targeting of exosomes are still challenges for further clinical applications. This study aims to explore the targeted efficiency and therapeutic potential of biomimetic platelet membrane-coated exosome-mimetic nanovesicles (P-ENVs) in atherosclerosis. Methods To produce exosome-mimetic nanovesicles (ENVs), MSCs were successively extruded through polycarbonate porous membranes. P-ENVs were engineered by fusing MSC-derived ENVs with platelet membranes and characterized using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot. The stability and safety of P-ENVs were also assessed. The targeted efficacy of P-ENVs was evaluated using an in vivo imaging system (IVIS) spectrum imaging system and immunofluorescence. Histological analyses, Oil Red O (ORO) staining, and Western blot were used to investigate the anti-atherosclerotic effectiveness of P-ENVs. Results Both ENVs and P-ENVs exhibited similar characteristics to exosomes. Subsequent miRNA sequencing of P-ENVs revealed their potential to mitigate atherosclerosis by influencing biological processes related to cholesterol metabolism. In an ApoE-/- mice model, the intravenous administration of P-ENVs exhibited enhanced targeting of atherosclerotic plaques, resulting in a significant reduction in lipid deposition and necrotic core area. Our in vitro experiments showed that P-ENVs promoted cholesterol efflux and reduced total cholesterol content in foam cells. Further analysis revealed that P-ENVs attenuated intracellular cholesterol accumulation by upregulating the expression of the critical cholesterol transporters ABCA1 and ABCG1. Conclusion This study highlighted the potential of P-ENVs as a novel nano-drug delivery platform for enhancing drug delivery efficiency while concurrently mitigating adverse reactions in atherosclerotic therapy.
Collapse
Affiliation(s)
- Yu Jiang
- Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Miao Yu
- Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Zhi-Feng Song
- Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Zhi-Yao Wei
- Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Ji Huang
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, National Clinical Research Center for Cardiovascular Diseases, Beijing, People’s Republic of China
| | - Hai-Yan Qian
- Center for Coronary Heart Disease, Department of Cardiology, Fu Wai Hospital, National Center for Cardiovascular Diseases of China, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
48
|
Lyytinen G, Melnikov G, Brynedal A, Anesäter E, Antoniewicz L, Blomberg A, Wallén H, Bosson JA, Hedman L, Tehrani S, Lundbäck M. Use of heated tobacco products (IQOS) causes an acute increase in arterial stiffness and platelet thrombus formation. Atherosclerosis 2024; 390:117335. [PMID: 37872010 DOI: 10.1016/j.atherosclerosis.2023.117335] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/19/2023] [Accepted: 10/06/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND AND AIMS Heated tobacco products (HTPs) are novel alternative tobacco products being promoted as an alternative to cigarettes. To evaluate the impact of HTP use on vascular function, we investigated the effects of a brief HTP usage on arterial stiffness and platelet thrombus formation in healthy volunteers. METHODS In a randomised crossover study, twenty-four healthy young adults with occasional tobacco use smoked the HTP IQOS 3 Multi (Phillip Morris Int.) and "no-exposure" was used as a control, with a wash-out period of at least one week in-between. Arterial stiffness was assessed through pulse wave velocity and pulse wave analysis. Blood samples, collected at baseline and 5 min following exposure, were analysed with the Total-Thrombus-formation analysis system evaluating platelet and fibrin-rich thrombus formation tendency. RESULTS HTP exposure caused immediate heightened pulse wave velocity (+0.365 m/s, 95% CI: +0.188 to 0.543; p = 0.004) and enhanced augmentation index corrected to heart rate (+6.22%, 95% CI: +2.33 to 10.11; p = 0.003) compared to the no-exposure occasion. Similarly, blood pressure and heart rate transiently increased immediately following HTP inhalation. Platelet thrombus formation significantly increased following HTP exposure (area under the curve +59.5, 95% CI: +25.6 to 93.4; p < 0.001) compared to no-exposure. No effect was seen on fibrin-rich thrombus formation following HTP-exposure. CONCLUSIONS Brief HTP use in healthy young adults had immediate adverse effects on vascular function resulting in increased arterial stiffness and platelet thrombus formation, known risk factors for the development of atherosclerosis. Further research is needed to address long term health impacts.
Collapse
Affiliation(s)
- Gustaf Lyytinen
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Georgy Melnikov
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Amelie Brynedal
- Dept. of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Erik Anesäter
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Lukasz Antoniewicz
- Department of Medicine II, Division of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Anders Blomberg
- Dept. of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Håkan Wallén
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jenny A Bosson
- Dept. of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Linnea Hedman
- Department of Public Health and Clinical Medicine, Section of Sustainable Health, The OLIN Unit, Umeå University, Umeå, Sweden
| | - Sara Tehrani
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Lundbäck
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
49
|
Cetinkaya Z, Kelesoglu S, Tuncay A, Yilmaz Y, Karaca Y, Karasu M, Secen O, Cinar A, Harman M, Sahin S, Akin Y, Yavcin O. The Role of Pan-Immune-Inflammation Value in Determining the Severity of Coronary Artery Disease in NSTEMI Patients. J Clin Med 2024; 13:1295. [PMID: 38592192 PMCID: PMC10931938 DOI: 10.3390/jcm13051295] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Even though medication and interventional therapy have improved the death rate for non-ST elevation myocardial infarction (NSTEMI) patients, these patients still have a substantial residual risk of cardiovascular events. Early identification of high-risk individuals is critical for improving prognosis, especially in this patient group. The focus of recent research has switched to finding new related indicators that can help distinguish high-risk patients. For this purpose, we examined the relationship between the pan-immune-inflammation value (PIV) and the severity of coronary artery disease (CAD) defined by the SYNTAX score (SxS) in NSTEMI patients. METHODS Based on the SxS, CAD patients were split into three groups. To evaluate the risk variables of CAD, multivariate logistic analysis was employed. RESULTS The PIV (odds ratio: 1.003; 95% CI: 1.001-1.005; p = 0.005) was found to be an independent predictor of a high SxS in the multivariate logistic regression analysis. Additionally, there was a positive association between the PIV and SxS (r: 0.68; p < 0.001). The PIV predicted the severe coronary lesion in the receiver-operating characteristic curve analysis with a sensitivity of 91% and specificity of 81.1%, using an appropriate cutoff value of 568.2. CONCLUSIONS In patients with non-STEMI, the PIV, a cheap and easily measured laboratory variable, was substantially correlated with a high SxS and the severity of CAD.
Collapse
Affiliation(s)
- Zeki Cetinkaya
- Department of Cardiology, Ministry of Health, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey; (Z.C.); (Y.K.); (M.K.); (O.S.); (S.S.); (Y.A.); (O.Y.)
| | - Saban Kelesoglu
- Department of Cardiology, Erciyes University Faculty of Medicine, Kayseri 38039, Turkey
| | - Aydin Tuncay
- Department of Cardiovascular Surgery, Erciyes University Faculty of Medicine, Kayseri 38039, Turkey
| | - Yucel Yilmaz
- Department of Cardiology, University of Health Sciences, Kayseri Education and Research Hospital, Kayseri 38100, Turkey; (Y.Y.); (A.C.)
| | - Yucel Karaca
- Department of Cardiology, Ministry of Health, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey; (Z.C.); (Y.K.); (M.K.); (O.S.); (S.S.); (Y.A.); (O.Y.)
| | - Mehdi Karasu
- Department of Cardiology, Ministry of Health, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey; (Z.C.); (Y.K.); (M.K.); (O.S.); (S.S.); (Y.A.); (O.Y.)
| | - Ozlem Secen
- Department of Cardiology, Ministry of Health, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey; (Z.C.); (Y.K.); (M.K.); (O.S.); (S.S.); (Y.A.); (O.Y.)
| | - Ahmet Cinar
- Department of Cardiology, University of Health Sciences, Kayseri Education and Research Hospital, Kayseri 38100, Turkey; (Y.Y.); (A.C.)
| | - Murat Harman
- Department of Cardiology, Fırat University Faculty of Medicine, Elazıg 23119, Turkey;
| | - Seyda Sahin
- Department of Cardiology, Ministry of Health, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey; (Z.C.); (Y.K.); (M.K.); (O.S.); (S.S.); (Y.A.); (O.Y.)
| | - Yusuf Akin
- Department of Cardiology, Ministry of Health, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey; (Z.C.); (Y.K.); (M.K.); (O.S.); (S.S.); (Y.A.); (O.Y.)
| | - Ozkan Yavcin
- Department of Cardiology, Ministry of Health, Elazıg Fethi Sekin City Hospital, Elazıg 23280, Turkey; (Z.C.); (Y.K.); (M.K.); (O.S.); (S.S.); (Y.A.); (O.Y.)
| |
Collapse
|
50
|
Schulte C, Pieper L, Frye M, Waldeyer C, Neumann JT, Brunner FJ, Pula G. Antiplatelet drugs do not protect from platelet-leukocyte aggregation in coronary artery disease. J Thromb Haemost 2024; 22:553-557. [PMID: 37225020 DOI: 10.1016/j.jtha.2023.04.041] [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: 02/04/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/26/2023]
Abstract
BACKGROUND Despite advances in cardiovascular medicine, coronary artery disease (CAD) remains a leading cause of mortality. Among the pathophysiological features of this condition, platelet-leukocyte aggregates (PLAs) require further attention, either as diagnostic/prognostic disease markers or as potential interventional targets. OBJECTIVES In this study, we characterized PLAs in patients with CAD. Primarily, we investigated the association of PLA levels with CAD diagnosis. In addition, the basal levels of platelet activation and degranulation were assessed in patients with CAD and controls, and their correlation with PLA levels was analyzed. Finally, the effect of antiplatelet treatments on circulating PLA numbers, basal platelet activation, and degranulation was studied in patients with CAD. METHODS Participants were recruited at the Department of Cardiology of the University Heart and Vascular Centre Hamburg Eppendorf. Among patients admitted with severe chest pain, the diagnosis of CAD was made angiographically, and patients without CAD were used as controls. PLAs, platelet activation, and platelet degranulation were assessed by flow cytometry. RESULTS Circulating PLAs and basal platelet degranulation levels were significantly higher in patients with CAD than in controls. Surprisingly, there was no significant correlation between PLA levels and platelet degranulation (or any other measured parameter). In addition, patients with CAD on antiplatelet therapy did not display lower PLA or platelet degranulation levels compared with those in controls. CONCLUSION Overall, these data suggest a mechanism of PLA formation that is independent of platelet activation or degranulation and highlights the inefficiency of current antiplatelet treatments for the prevention of basal platelet degranulation and PLA formation.
Collapse
Affiliation(s)
- Christian Schulte
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Luise Pieper
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maike Frye
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Eppendorf, Hamburg, Germany
| | - Christoph Waldeyer
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Johannes T Neumann
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Fabian J Brunner
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg, Luebeck, Kiel, Hamburg, Germany
| | - Giordano Pula
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Eppendorf, Hamburg, Germany; Centre for Biomedicine, Hull York Medical School, Hull, UK.
| |
Collapse
|