1
|
Maekawa K, Nakamura E, Saito Y, Matsuura Y, Gi T, Nishihira K, Oguri N, Moriguchi-Goto S, Sato Y, Hatakeyama K, Shibata Y, Komohara Y, Kaikita K, Asada Y, Yamashita A. Inflammatory stimuli and hypoxia on atherosclerotic plaque thrombogenicity: Linking macrophage tissue factor and glycolysis. PLoS One 2025; 20:e0316474. [PMID: 40036261 DOI: 10.1371/journal.pone.0316474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/11/2024] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The thrombogenic potential of cells within atherosclerotic plaques is critical in the formation of a coronary thrombus. We hypothesized that a combination of inflammatory and hypoxic stimuli enhances tissue factor (TF) expression and glycolysis in cells in atherosclerotic plaques and contributes to coronary thrombus formation. AIMS To identify TF- and hexokinase (HK)-II-expressing cells in coronary atherosclerotic plaques and thrombi and determine the effects of combined inflammatory and hypoxic stimuli and glycolysis on TF expression in peripheral blood mononuclear cell-derived macrophages. METHODS We immunohistochemically assessed TF and HK-II expression in stable (n = 20) and unstable (n = 24) human coronary plaques and aspirated acute coronary thrombi (n = 15). The macrophages were stimulated with tumor necrosis factor-α, interferon-γ, or interleukin-10 under normoxic (21% O2) or hypoxic (1% O2) conditions, and TF expression was assessed. RESULTS TF and HK-II expression were increased in unstable plaques compared with stable plaques. The number of CD68- and HK-II-immunopositive cells positively correlated with the number of TF-immunopositive cells. TF- and HK-II-expressing macrophages, which expressed M1- or M2-like markers, were involved in platelet-fibrin thrombus formation in ruptured plaques. The combination of inflammatory and hypoxic conditions additively augmented TF expression and procoagulant activity in the cultured macrophages. Inhibition of glycolysis with 2-deoxyglucose reduced the augmented TF expression and procoagulant activity. CONCLUSION Combined inflammatory and hypoxic conditions in atherosclerotic plaques may markedly enhance procoagulant activity in macrophages and contribute to coronary thrombus formation following plaque disruption. Macrophage TF expression may be associated with glycolysis.
Collapse
Affiliation(s)
- Kazunari Maekawa
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Eriko Nakamura
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Yoichi Saito
- Bioengineering Lab, Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Kumamoto, Japan
| | - Yunosuke Matsuura
- Division of Cardiovascular Medicine and Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Toshihiro Gi
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Kensaku Nishihira
- Department of Cardiology, Miyazaki Medical Association Hospital, Miyazaki, Miyazaki, Japan
| | - Nobuyuki Oguri
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Sayaka Moriguchi-Goto
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Yuichiro Sato
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Kinta Hatakeyama
- Department of Pathology, National Cerebral and Cardiovascular Center Hospital, Suita, Osaka, Japan
| | - Yoshisato Shibata
- Department of Cardiology, Miyazaki Medical Association Hospital, Miyazaki, Miyazaki, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Kumamoto, Japan
| | - Koichi Kaikita
- Division of Cardiovascular Medicine and Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Yujiro Asada
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
- Department of Diagnostic Pathology, Miyazaki Medical Association Hospital, Miyazaki, Miyazaki, Japan
| | - Atsushi Yamashita
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| |
Collapse
|
2
|
Lu MJ, Zhang JQ, Nie ZY, Yan TH, Cao YB, Zhang LC, Li L. Monocyte/macrophage-mediated venous thrombus resolution. Front Immunol 2024; 15:1429523. [PMID: 39100675 PMCID: PMC11297357 DOI: 10.3389/fimmu.2024.1429523] [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: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Venous thromboembolism (VTE) poses a notable risk of morbidity and mortality. The natural resolution of the venous thrombus might be a potential alternative treatment strategy for VTE. Monocytes/macrophages merge as pivotal cell types in the gradual resolution of the thrombus. In this review, the vital role of macrophages in inducing inflammatory response, augmenting neovascularization, and facilitating the degradation of fibrin and collagen during thrombus resolution was described. The two phenotypes of macrophages involved in thrombus resolution and their dual functions were discussed. Macrophages expressing various factors, including cytokines and their receptors, adhesion molecules, chemokine receptors, vascular endothelial growth factor receptors, profibrinolytic- or antifibrinolytic-related enzymes, and other elements, are explored for their potential to promote or attenuate thrombus resolution. Furthermore, this review provides a comprehensive summary of new and promising therapeutic candidate drugs associated with monocytes/macrophages that have been demonstrated to promote or impair thrombus resolution. However, further clinical trials are essential to validate their efficacy in VTE therapy.
Collapse
Affiliation(s)
- Meng-Jiao Lu
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Jia-Qi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhou-Yu Nie
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Hua Yan
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Yong-Bing Cao
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Li
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Kral-Pointner JB, Haider P, Szabo PL, Salzmann M, Brekalo M, Schneider KH, Schrottmaier WC, Kaun C, Bleichert S, Kiss A, Sickha R, Hengstenberg C, Huber K, Brostjan C, Bergmeister H, Assinger A, Podesser BK, Wojta J, Hohensinner P. Reduced Monocyte and Neutrophil Infiltration and Activation by P-Selectin/CD62P Inhibition Enhances Thrombus Resolution in Mice. Arterioscler Thromb Vasc Biol 2024; 44:954-968. [PMID: 38385292 PMCID: PMC11020038 DOI: 10.1161/atvbaha.123.320016] [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: 08/16/2023] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Venous thromboembolism is a major health problem. After thrombus formation, its resolution is essential to re-establish blood flow, which is crucially mediated by infiltrating neutrophils and monocytes in concert with activated platelets and endothelial cells. Thus, we aimed to modulate leukocyte function during thrombus resolution post-thrombus formation by blocking P-selectin/CD62P-mediated cell interactions. METHODS Thrombosis was induced by inferior vena cava stenosis through ligation in mice. After 1 day, a P-selectin-blocking antibody or isotype control was administered and thrombus composition and resolution were analyzed. RESULTS Localizing neutrophils and macrophages in thrombotic lesions of wild-type mice revealed that these cells enter the thrombus and vessel wall from the caudal end. Neutrophils were predominantly present 1 day and monocytes/macrophages 3 days after vessel ligation. Blocking P-selectin reduced circulating platelet-neutrophil and platelet-Ly6Chigh monocyte aggregates near the thrombus, and diminished neutrophils and Ly6Chigh macrophages in the cranial thrombus part compared with isotype-treated controls. Depletion of neutrophils 1 day after thrombus initiation did not phenocopy P-selectin inhibition but led to larger thrombi compared with untreated controls. In vitro, P-selectin enhanced human leukocyte function as P-selectin-coated beads increased reactive oxygen species production by neutrophils and tissue factor expression of classical monocytes. Accordingly, P-selectin inhibition reduced oxidative burst in the thrombus and tissue factor expression in the adjacent vessel wall. Moreover, blocking P-selectin reduced thrombus density determined by scanning electron microscopy and increased urokinase-type plasminogen activator levels in the thrombus, which accelerated caudal fibrin degradation from day 3 to day 14. This accelerated thrombus resolution as thrombus volume declined more rapidly after blocking P-selectin. CONCLUSIONS Inhibition of P-selectin-dependent activation of monocytes and neutrophils accelerates venous thrombosis resolution due to reduced infiltration and activation of innate immune cells at the site of thrombus formation, which prevents early thrombus stabilization and facilitates fibrinolysis.
Collapse
Affiliation(s)
- Julia B. Kral-Pointner
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Division of Cardiology, Department of Internal Medicine II (J.B.K.-P., P. Haider, M.S., M.B., C.K., C.H., J.W.), Medical University of Vienna, Austria
| | - Patrick Haider
- Division of Cardiology, Department of Internal Medicine II (J.B.K.-P., P. Haider, M.S., M.B., C.K., C.H., J.W.), Medical University of Vienna, Austria
| | - Petra L. Szabo
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Centre for Biomedical Research and Translational Surgery (P.L.S., K.H.S., A.K., H.B., B.K.P., P. Hohensinner), Medical University of Vienna, Austria
| | - Manuel Salzmann
- Division of Cardiology, Department of Internal Medicine II (J.B.K.-P., P. Haider, M.S., M.B., C.K., C.H., J.W.), Medical University of Vienna, Austria
| | - Mira Brekalo
- Centre for Biomedical Research and Translational Surgery (P.L.S., K.H.S., A.K., H.B., B.K.P., P. Hohensinner), Medical University of Vienna, Austria
| | - Karl H. Schneider
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Centre for Biomedical Research and Translational Surgery (P.L.S., K.H.S., A.K., H.B., B.K.P., P. Hohensinner), Medical University of Vienna, Austria
| | - Waltraud C. Schrottmaier
- Institute for Vascular Biology and Thrombosis Research (W.C.S., A.A.), Medical University of Vienna, Austria
| | - Christoph Kaun
- Division of Cardiology, Department of Internal Medicine II (J.B.K.-P., P. Haider, M.S., M.B., C.K., C.H., J.W.), Medical University of Vienna, Austria
| | - Sonja Bleichert
- Division of Vascular Surgery, Department of General Surgery (S.B., C.B.), Medical University of Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Centre for Biomedical Research and Translational Surgery (P.L.S., K.H.S., A.K., H.B., B.K.P., P. Hohensinner), Medical University of Vienna, Austria
| | - Romana Sickha
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christian Hengstenberg
- Division of Cardiology, Department of Internal Medicine II (J.B.K.-P., P. Haider, M.S., M.B., C.K., C.H., J.W.), Medical University of Vienna, Austria
| | - Kurt Huber
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital, Vienna, Austria (K.H.)
- Medical Faculty, Sigmund Freud University, Vienna, Austria (K.H.)
| | - Christine Brostjan
- Division of Vascular Surgery, Department of General Surgery (S.B., C.B.), Medical University of Vienna, Austria
| | - Helga Bergmeister
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Centre for Biomedical Research and Translational Surgery (P.L.S., K.H.S., A.K., H.B., B.K.P., P. Hohensinner), Medical University of Vienna, Austria
| | - Alice Assinger
- Institute for Vascular Biology and Thrombosis Research (W.C.S., A.A.), Medical University of Vienna, Austria
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Centre for Biomedical Research and Translational Surgery (P.L.S., K.H.S., A.K., H.B., B.K.P., P. Hohensinner), Medical University of Vienna, Austria
| | - Johann Wojta
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Division of Cardiology, Department of Internal Medicine II (J.B.K.-P., P. Haider, M.S., M.B., C.K., C.H., J.W.), Medical University of Vienna, Austria
| | - Philipp Hohensinner
- Ludwig Boltzmann Institute for Cardiovascular Research (J.B.K.-P., P.L.S., K.H.S., A.K., R.S., K.H., H.B., B.K.P., J.W., P. Hohensinner), Medical University of Vienna, Austria
- Centre for Biomedical Research and Translational Surgery (P.L.S., K.H.S., A.K., H.B., B.K.P., P. Hohensinner), Medical University of Vienna, Austria
| |
Collapse
|
4
|
Rehill AM, Leon G, McCluskey S, Schoen I, Hernandez-Santana Y, Annett S, Klavina P, Robson T, Curtis AM, Renné T, Hussey S, O'Donnell JS, Walsh PT, Preston RJS. Glycolytic reprogramming fuels myeloid cell-driven hypercoagulability. J Thromb Haemost 2024; 22:394-409. [PMID: 37865288 DOI: 10.1016/j.jtha.2023.10.006] [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: 04/25/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Myeloid cell metabolic reprogramming is a hallmark of inflammatory disease; however, its role in inflammation-induced hypercoagulability is poorly understood. OBJECTIVES We aimed to evaluate the role of inflammation-associated metabolic reprogramming in regulating blood coagulation. METHODS We used novel myeloid cell-based global hemostasis assays and murine models of immunometabolic disease. RESULTS Glycolysis was essential for enhanced activated myeloid cell tissue factor expression and decryption, driving increased cell-dependent thrombin generation in response to inflammatory challenge. Similarly, inhibition of glycolysis enhanced activated macrophage fibrinolytic activity through reduced plasminogen activator inhibitor 1 activity. Macrophage polarization or activation markedly increased endothelial protein C receptor (EPCR) expression on monocytes and macrophages, leading to increased myeloid cell-dependent protein C activation. Importantly, inflammation-dependent EPCR expression on tissue-resident macrophages was also observed in vivo. Adipose tissue macrophages from obese mice fed a high-fat diet exhibited significantly enhanced EPCR expression and activated protein C generation compared with macrophages isolated from the adipose tissue of healthy mice. Similarly, the induction of colitis in mice prompted infiltration of EPCR+ innate myeloid cells within inflamed colonic tissue that were absent from the intestinal tissue of healthy mice. CONCLUSION Collectively, this study identifies immunometabolic regulation of myeloid cell hypercoagulability, opening new therapeutic possibilities for targeted mitigation of thromboinflammatory disease.
Collapse
Affiliation(s)
- Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland. https://twitter.com/aislingrehill
| | - Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland
| | - Sean McCluskey
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland
| | - Ingmar Schoen
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Yasmina Hernandez-Santana
- National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland; Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Stephanie Annett
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Paula Klavina
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Tracy Robson
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Annie M Curtis
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Thomas Renné
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland; Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Seamus Hussey
- National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland; Department of Paediatrics, University College Dublin and Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Patrick T Walsh
- National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland; Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland.
| |
Collapse
|
5
|
Aloi N, Drago G, Ruggieri S, Cibella F, Colombo P, Longo V. Extracellular Vesicles and Immunity: At the Crossroads of Cell Communication. Int J Mol Sci 2024; 25:1205. [PMID: 38256278 PMCID: PMC10816988 DOI: 10.3390/ijms25021205] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024] Open
Abstract
Extracellular vesicles (EVs), comprising exosomes and microvesicles, are small membranous structures secreted by nearly all cell types. They have emerged as crucial mediators in intercellular communication, playing pivotal roles in diverse physiological and pathological processes, notably within the realm of immunity. These roles go beyond mere cellular interactions, as extracellular vesicles stand as versatile and dynamic components of immune regulation, impacting both innate and adaptive immunity. Their multifaceted involvement includes immune cell activation, antigen presentation, and immunomodulation, emphasising their significance in maintaining immune homeostasis and contributing to the pathogenesis of immune-related disorders. Extracellular vesicles participate in immunomodulation by delivering a wide array of bioactive molecules, including proteins, lipids, and nucleic acids, thereby influencing gene expression in target cells. This manuscript presents a comprehensive review that encompasses in vitro and in vivo studies aimed at elucidating the mechanisms through which EVs modulate human immunity. Understanding the intricate interplay between extracellular vesicles and immunity is imperative for unveiling novel therapeutic targets and diagnostic tools applicable to various immunological disorders, including autoimmune diseases, infectious diseases, and cancer. Furthermore, recognising the potential of EVs as versatile drug delivery vehicles holds significant promise for the future of immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (N.A.); (G.D.); (S.R.); (F.C.); (V.L.)
| | | |
Collapse
|
6
|
Anijs RJS, Nguyen YN, Cannegieter SC, Versteeg HH, Buijs JT. MicroRNAs as prognostic biomarkers for (cancer-associated) venous thromboembolism. J Thromb Haemost 2023; 21:7-17. [PMID: 36695398 DOI: 10.1016/j.jtha.2022.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 01/11/2023]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs with gene regulatory functions and are commonly dysregulated in disease states. As miRNAs are relatively stable, easily measured, and accessible from plasma or other body fluids, they are promising biomarkers for the diagnosis and prediction of cancer and cardiovascular diseases. Venous thromboembolism (VTE) is the third most common cardiovascular disease worldwide with high morbidity and mortality. The suggested roles of miRNAs in regulating the pathophysiology of VTE and as VTE biomarkers are nowadays more evidenced. Patients with cancer are at increased risk of developing VTE compared to the general population. However, current risk prediction models for cancer-associated thrombosis (CAT) perform suboptimally, and novel biomarkers are therefore urgently needed to identify which patients may benefit the most from thromboprophylaxis. This review will first discuss how miRNAs mechanistically contribute to the pathophysiology of VTE. Next, the potential use of miRNAs as predictive biomarkers for VTE in subjects without cancer is reviewed, followed by an in-depth focus on CAT. Several of the identified miRNAs in CAT were found to be differentially regulated in VTE as well, giving clues on the pathophysiology of CAT. We propose that subsequent studies should be adequately sized to determine which panel of miRNAs best predicts VTE and CAT. Thereafter, validation studies using comparable patient populations are required to ultimately unveil whether miRNAs-as standalone or incorporated into existing risk models-are promising valuable VTE and CAT biomarkers.
Collapse
Affiliation(s)
- Rayna J S Anijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yen Nhi Nguyen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne C Cannegieter
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands; Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen T Buijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
7
|
Hwang EH, Hur GH, Koo BS, Oh H, Kim G, Jung H, Baek SH, An YJ, Park JH, Hong JJ. Monocytes as suitable carriers for dissemination of dengue viral infection. Heliyon 2022; 8:e11212. [PMCID: PMC9615040 DOI: 10.1016/j.heliyon.2022.e11212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/10/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
Dengue viruses (DENVs) exploit monocytes and macrophages for tropism and replication, therefore, establishing a long-term reservoir. However, their roles in dengue pathogenesis remains unclear. Here, using the human monocytic cell line THP-1, human primary monocytes, and non-human primate models, we show that DENV-infected monocytes represent suitable carriers for circulatory viral dissemination. Monocyte-derived macrophages expressing M2 surface markers at the gene level efficiently replicated, while the productivity of monocyte replication was low. However, attachment of DENVs to the cellular surface of monocytes was similar to that of macrophages. Furthermore, after differentiation with type-2 cytokines, DENV-attached monocytes could replicate DENVs. Productive DENV infection was confirmed by intravenous injection of DENVs into nonhuman primate model, in which, DENV attachment to monocytes was positively correlated with viremia. These results provide insight into the role of circulating monocytes in DENV infection, suggesting that monocytes directly assist in DENV dissemination and replication during viremia and could be applied to design antiviral intervention.
Collapse
Affiliation(s)
- Eun-Ha Hwang
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea,Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Jeolla, Republic of Korea
| | | | - Bon-Sang Koo
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea
| | - Hanseul Oh
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea,College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Green Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea,Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Jeolla, Republic of Korea
| | - Hoyin Jung
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea
| | - Seung Ho Baek
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea
| | - You Jung An
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Jeolla, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk, Republic of Korea,KRIBB School of Bioscience, Korea University of Science & Technology (UST), Daejeon, Republic of Korea,Corresponding author.
| |
Collapse
|
8
|
Haider P, Kral-Pointner JB, Salzmann M, Moik F, Bleichert S, Schrottmaier WC, Kaun C, Brekalo M, Fischer MB, Speidl WS, Hengstenberg C, Podesser BK, Huber K, Pabinger I, Knapp S, Brombacher F, Brostjan C, Ay C, Wojta J, Hohensinner PJ. Interleukin-4 receptor alpha signaling regulates monocyte homeostasis. FASEB J 2022; 36:e22532. [PMID: 36063138 PMCID: PMC9544925 DOI: 10.1096/fj.202101672rr] [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: 10/29/2021] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 12/01/2022]
Abstract
Interleukin‐4 (IL‐4) and its receptors (IL‐4R) promote the proliferation and polarization of macrophages. However, it is unknown if IL‐4R also influences monocyte homeostasis and if steady state IL‐4 levels are sufficient to affect monocytes. Employing full IL‐4 receptor alpha knockout mice (IL‐4Rα−/−) and mice with a myeloid‐specific deletion of IL‐4Rα (IL‐4Rαf/f LysMcre), we show that IL‐4 acts as a homeostatic factor regulating circulating monocyte numbers. In the absence of IL‐4Rα, murine monocytes in blood were reduced by 50% without altering monocytopoiesis in the bone marrow. This reduction was accompanied by a decrease in monocyte‐derived inflammatory cytokines in the plasma. RNA sequencing analysis and immunohistochemical staining of splenic monocytes revealed changes in mRNA and protein levels of anti‐apoptotic factors including BIRC6 in IL‐4Rα−/− knockout animals. Furthermore, assessment of monocyte lifespan in vivo measuring BrdU+ cells revealed that the lifespan of circulating monocytes was reduced by 55% in IL‐4Rα−/− mice, whereas subcutaneously applied IL‐4 prolonged it by 75%. Treatment of human monocytes with IL‐4 reduced the amount of dying monocytes in vitro. Furthermore, IL‐4 stimulation reduced the phosphorylation of proteins involved in the apoptosis pathway, including the phosphorylation of the NFκBp65 protein. In a cohort of human patients, serum IL‐4 levels were significantly associated with monocyte counts. In a sterile peritonitis model, reduced monocyte counts resulted in an attenuated recruitment of monocytes upon inflammatory stimulation in IL‐4Rαf/f LysMcre mice without changes in overall migratory function. Thus, we identified a homeostatic role of IL‐4Rα in regulating the lifespan of monocytes in vivo.
Collapse
Affiliation(s)
- Patrick Haider
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Julia B Kral-Pointner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Manuel Salzmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Florian Moik
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sonja Bleichert
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Waltraud C Schrottmaier
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Christoph Kaun
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Mira Brekalo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Michael B Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria.,Department of Biomedical Research, Danube University Krems, Krems, Austria
| | - Walter S Speidl
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christian Hengstenberg
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria.,Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Kurt Huber
- 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminenhospital, Vienna, Austria.,Medical Faculty, Sigmund Freud University, Vienna, Austria
| | - Ingrid Pabinger
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sylvia Knapp
- Laboratory of Infection Biology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Frank Brombacher
- Institute of Infectious Disease and Molecular Medicine, International Center for Genetic and Biotechnology Cape Town Component & University of Cape Town, Cape Town, South Africa
| | - Christine Brostjan
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Cihan Ay
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria.,Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Philipp J Hohensinner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria.,Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Al-Koussa H, AlZaim I, El-Sabban ME. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. J Clin Med 2022; 11:jcm11164932. [PMID: 36013171 PMCID: PMC9410115 DOI: 10.3390/jcm11164932] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
The notion of blood coagulation dates back to the ancient Greek civilization. However, the emergence of innovative scientific discoveries that started in the seventeenth century formulated the fundamentals of blood coagulation. Our understanding of key coagulation processes continues to evolve, as novel homeostatic and pathophysiological aspects of hemostasis are revealed. Hemostasis is a dynamic physiological process, which stops bleeding at the site of injury while maintaining normal blood flow within the body. Intrinsic and extrinsic coagulation pathways culminate in the homeostatic cessation of blood loss, through the sequential activation of the coagulation factors. Recently, the cell-based theory, which combines these two pathways, along with newly discovered mechanisms, emerged to holistically describe intricate in vivo coagulation mechanisms. The complexity of these mechanisms becomes evident in coagulation diseases such as hemophilia, Von Willebrand disease, thrombophilia, and vitamin K deficiency, in which excessive bleeding, thrombosis, or unnecessary clotting, drive the development and progression of diseases. Accumulating evidence implicates cell-derived and platelet-derived extracellular vesicles (EVs), which comprise microvesicles (MVs), exosomes, and apoptotic bodies, in the modulation of the coagulation cascade in hemostasis and thrombosis. As these EVs are associated with intercellular communication, molecular recycling, and metastatic niche creation, emerging evidence explores EVs as valuable diagnostic and therapeutic approaches in thrombotic and prothrombotic diseases.
Collapse
Affiliation(s)
- Houssam Al-Koussa
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan E. El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Correspondence: ; Tel.: +961-01-350-000 (ext. 4765)
| |
Collapse
|
10
|
Kawai K, Vozenilek AE, Kawakami R, Sato Y, Ghosh SKB, Virmani R, Finn AV. Understanding the role of alternative macrophage phenotypes in human atherosclerosis. Expert Rev Cardiovasc Ther 2022; 20:689-705. [PMID: 35942866 DOI: 10.1080/14779072.2022.2111301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Atherosclerosis-based ischemic heart disease is still the primary cause of death throughout the world. Over the past decades there has been no significant changes in the therapeutic approaches to atherosclerosis, which are mainly based on lipid lowering therapies and management of comorbid conditions such as diabetes and hypertension. The involvement of macrophages in atherosclerosis has been recognized for decades. More recently, a more detailed and sophisticated understanding of their various phenotypes and roles in the atherosclerotic process has been recognized. This new data is revealing how specific subtypes of macrophage-induced inflammation may have distinct effects on atherosclerosis progression and may provide new approaches for treatment, based upon targeting of specific macrophage subtypes. AREAS COVERED We will comprehensively review the spectrum of macrophage phenotypes and how they contribute to atherosclerotic plaque development and progression. EXPERT OPINION Various signals derived from atherosclerotic lesions drive macrophages into complex subsets with different gene expression profiles, phenotypes, and functions, not all of which are understood. Macrophage phenotypes include those that enhance, heal, and regress the atherosclerotic lesions though various mechanisms. Targeting of specific macrophage phenotypes may provide a promising and novel approach to prevent atherosclerosis progression.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA.,University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Chua CLL, Khoo SKM, Ong JLE, Ramireddi GK, Yeo TW, Teo A. Malaria in Pregnancy: From Placental Infection to Its Abnormal Development and Damage. Front Microbiol 2021; 12:777343. [PMID: 34867919 PMCID: PMC8636035 DOI: 10.3389/fmicb.2021.777343] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/20/2021] [Indexed: 11/24/2022] Open
Abstract
Malaria remains a global health burden with Plasmodium falciparum accounting for the highest mortality and morbidity. Malaria in pregnancy can lead to the development of placental malaria, where P. falciparum-infected erythrocytes adhere to placental receptors, triggering placental inflammation and subsequent damage, causing harm to both mother and her infant. Histopathological studies of P. falciparum-infected placentas revealed various placental abnormalities such as excessive perivillous fibrinoid deposits, breakdown of syncytiotrophoblast integrity, trophoblast basal lamina thickening, increased syncytial knotting, and accumulation of mononuclear immune cells within intervillous spaces. These events in turn, are likely to impair placental development and function, ultimately causing placental insufficiency, intrauterine growth restriction, preterm delivery and low birth weight. Hence, a better understanding of the mechanisms behind placental alterations and damage during placental malaria is needed for the design of effective interventions. In this review, using evidence from human studies and murine models, an integrated view on the potential mechanisms underlying placental pathologies in malaria in pregnancy is provided. The molecular, immunological and metabolic changes in infected placentas that reflect their responses to the parasitic infection and injury are discussed. Finally, potential models that can be used by researchers to improve our understanding on the pathogenesis of malaria in pregnancy and placental pathologies are presented.
Collapse
Affiliation(s)
| | | | - Jun Long Ernest Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Tsin Wen Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Center for Infectious Diseases, Singapore, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - Andrew Teo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Medicine at Royal Melbourne Hospital, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Hernández-Huerta MT, Pérez-Santiago AD, Pérez-Campos Mayoral L, Sánchez Navarro LM, Rodal Canales FJ, Majluf-Cruz A, Matias-Cervantes CA, Pérez-Campos Mayoral E, Romero Díaz C, Mayoral-Andrade G, Martínez Cruz M, Luna Ángel J, Pérez-Campos E. Mechanisms of Immunothrombosis by SARS-CoV-2. Biomolecules 2021; 11:1550. [PMID: 34827548 PMCID: PMC8615366 DOI: 10.3390/biom11111550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 12/20/2022] Open
Abstract
SARS-CoV-2 contains certain molecules that are related to the presence of immunothrombosis. Here, we review the pathogen and damage-associated molecular patterns. We also study the imbalance of different molecules participating in immunothrombosis, such as tissue factor, factors of the contact system, histones, and the role of cells, such as endothelial cells, platelets, and neutrophil extracellular traps. Regarding the pathogenetic mechanism, we discuss clinical trials, case-control studies, comparative and translational studies, and observational studies of regulatory or inhibitory molecules, more specifically, extracellular DNA and RNA, histones, sensors for RNA and DNA, as well as heparin and heparinoids. Overall, it appears that a network of cells and molecules identified in this axis is simultaneously but differentially affecting patients at different stages of COVID-19, and this is characterized by endothelial damage, microthrombosis, and inflammation.
Collapse
Affiliation(s)
- María Teresa Hernández-Huerta
- CONACyT, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.T.H.-H.); (C.A.M.-C.)
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
| | | | - Laura Pérez-Campos Mayoral
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | | | - Francisco Javier Rodal Canales
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | | | - Carlos Alberto Matias-Cervantes
- CONACyT, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.T.H.-H.); (C.A.M.-C.)
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
| | - Eduardo Pérez-Campos Mayoral
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | - Carlos Romero Díaz
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | - Gabriel Mayoral-Andrade
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico;
| | - Margarito Martínez Cruz
- Tecnológico Nacional de México/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (A.D.P.-S.); (M.M.C.)
| | - Judith Luna Ángel
- Hospital General Dr. Aurelio Valdivieso, Oaxaca de Juárez, Oaxaca 68000, Mexico;
| | - Eduardo Pérez-Campos
- Grupo de Investigación Biomedicina y Salud, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (E.P.-C.M.); (C.R.D.); (G.M.-A.)
- Tecnológico Nacional de México/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico; (A.D.P.-S.); (M.M.C.)
- Laboratorio de Patología Clinica “Eduardo Pérez Ortega”, Oaxaca de Juárez, Oaxaca 68000, Mexico
| |
Collapse
|
13
|
MiRNA Let-7a and Let-7d Are Induced by Globotriaosylceramide via NF-kB Activation in Fabry Disease. Genes (Basel) 2021; 12:genes12081184. [PMID: 34440358 PMCID: PMC8394417 DOI: 10.3390/genes12081184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Fabry disease is a hereditary genetic defect resulting in reduced activity of the enzyme α-galactosidase-A and the accumulation of globotriaosylceramide (Gb3) in body fluids and cells. Gb3 accumulation was especially reported for the vascular endothelium in several organs. METHODS Three Fabry disease patients were screened using a micro-RNA screen. An in vitro approach in human endothelial cells was used to determine miRNA regulation by Gb3. RESULTS In a micro-RNA screen of three Fabry patients undergoing enzyme replacement therapy, we found that miRNAs let-7a and let-7d were significantly increased after therapy. We demonstrate in vitro in endothelial cells that Gb3 induced activation of NF-κB and activated downstream targets. In addition, NF-κB activity directly reduced let-7a and let-7d miRNA expression as inhibiting NF-kB nuclear entry abolished the Gb3 effects. CONCLUSION We suggest that let-7a and let-7d are potential markers for enzyme activity and inflammation in Fabry disease patients.
Collapse
|
14
|
Hohensinner PJ, Lenz M, Haider P, Mayer J, Richter M, Kaun C, Goederle L, Brekalo M, Salzmann M, Sharma S, Fischer MB, Stojkovic S, Ramsmayer D, Hengstenberg C, Podesser BK, Huber K, Binder CJ, Wojta J, Speidl WS. Pharmacological inhibition of fatty acid oxidation reduces atherosclerosis progression by suppression of macrophage NLRP3 inflammasome activation. Biochem Pharmacol 2021; 190:114634. [PMID: 34058186 DOI: 10.1016/j.bcp.2021.114634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Inflammation is a key process during atherosclerotic lesion development and propagation. Recent evidence showed clearly that especially the inhibition of interleukin (IL)-1β reduced atherosclerotic adverse events in human patients. Fatty acid oxidation (FAO) was previously demonstrated to interact with the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) pathway which is required for mature IL-1β secretion. To understand possible anti-inflammatory properties of FAO inhibition, we tested the effect of pharmacological FAO inhibition using the inhibitor for long-chain 3-ketoacyl coenzyme A thiolase trimetazidine on atherosclerotic plaque development and inflammation. EXPERIMENTAL APPROACH The effect of FAO inhibition was determined in LDL-R-/- male mice on a C57/BL6 background. In vitro effects of trimetazidine treatment were analyzed in human umbilical vein endothelial cells and human monocyte derived macrophages. KEY RESULTS We were able to demonstrate that inhibition of FAO reduced atherosclerotic plaque growth. We did not find direct anti-inflammatory properties of trimetazidine in endothelial cells or macrophages in vitro. However, we found that the activation of the NLRP3 system and the secretion of IL-1β were significantly reduced in macrophages after FAO inhibition. These results were confirmed in atherosclerotic lesions of mice treated with trimetazidine as they showed a significant reduction of IL-1β and cleaved caspase-1 in the atherosclerotic lesion as well as of IL-1β and IL-18 in the circulation. CONCLUSION Overall, we therefore suggest that the main mechanism of reducing inflammation of trimetazidine and FAO inhibition is the reduction of the NLRP-3 activation leading to reduced levels of the proinflammatory cytokine IL-1β.
Collapse
Affiliation(s)
- Philipp J Hohensinner
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Max Lenz
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Patrick Haider
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Julia Mayer
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Manuela Richter
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Christoph Kaun
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Laura Goederle
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Mira Brekalo
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Smriti Sharma
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Michael B Fischer
- Clinic for Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria; Department for Health Science and Biomedicine, Danube University Krems, Krems, Austria
| | - Stefan Stojkovic
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Daniel Ramsmayer
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Christian Hengstenberg
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Bruno K Podesser
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Kurt Huber
- 3(rd) Medical Department, Wilhelminenhospital, Vienna, Austria; Sigmund Freud University, Medical Faculty, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria; Core Facilities, Medical University of Vienna, Vienna, Austria.
| | - Walter S Speidl
- Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Li Q, Hu L, Li J, Yu P, Hu F, Wan B, Xu M, Cheng H, Yu W, Jiang L, Shi Y, Li J, Duan M, Long Y, Liu WT. Hydrogen Attenuates Endotoxin-Induced Lung Injury by Activating Thioredoxin 1 and Decreasing Tissue Factor Expression. Front Immunol 2021; 12:625957. [PMID: 33767697 PMCID: PMC7985449 DOI: 10.3389/fimmu.2021.625957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/18/2021] [Indexed: 01/19/2023] Open
Abstract
Endotoxin-induced lung injury is one of the major causes of death induced by endotoxemia, however, few effective therapeutic options exist. Hydrogen inhalation has recently been shown to be an effective treatment for inflammatory lung injury, but the underlying mechanism is unknown. In the current study we aim to investigate how hydrogen attenuates endotoxin-induced lung injury and provide reference values for the clinical application of hydrogen. LPS was used to establish an endotoxin-induced lung injury mouse model. The survival rate and pulmonary pathologic changes were evaluated. THP-1 and HUVECC cells were cultured in vitro. The thioredoxin 1 (Trx1) inhibitor was used to evaluate the anti-inflammatory effects of hydrogen. Hydrogen significantly improved the survival rate of mice, reduced pulmonary edema and hemorrhage, infiltration of neutrophils, and IL-6 secretion. Inhalation of hydrogen decreased tissue factor (TF) expression and MMP-9 activity, while Trx1 expression was increased in the lungs and serum of endotoxemia mice. LPS-stimulated THP-1 and HUVEC-C cells in vitro and showed that hydrogen decreases TF expression and MMP-9 activity, which were abolished by the Trx1 inhibitor, PX12. Hydrogen attenuates endotoxin-induced lung injury by decreasing TF expression and MMP-9 activity via activating Trx1. Targeting Trx1 by hydrogen may be a potential treatment for endotoxin-induced lung injury.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, Jinling College Affiliated to Nanjing Medical University, Nanjing, China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Juan Li
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Pan Yu
- Department of Burn and Plastic Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Fan Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Bing Wan
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Miaomiao Xu
- Department of Anesthesiology, Jinling College Affiliated to Nanjing Medical University, Nanjing, China
| | - Huixian Cheng
- Department of Anesthesiology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Wanyou Yu
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Liping Jiang
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yadan Shi
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Jincan Li
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Manlin Duan
- Department of Anesthesiology, Jinling College Affiliated to Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
- Department of Anesthesiology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Yun Long
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Wen-Tao Liu
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Zhou Y, Zeng J, Tu Y, Li L, Du S, Zhu L, Cang X, Lu J, Zhu M, Liu X. CSF1/CSF1R-mediated Crosstalk Between Choroidal Vascular Endothelial Cells and Macrophages Promotes Choroidal Neovascularization. Invest Ophthalmol Vis Sci 2021; 62:37. [PMID: 33764399 PMCID: PMC7995352 DOI: 10.1167/iovs.62.3.37] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose This study examined the role of the CSF1/CSF1Raxis in the crosstalk between choroidal vascular endothelial cells (CVECs) and macrophages during the formation of choroidal neovascularization (CNV). Methods Quantitative reverse transcriptase (QRT)-PCR, Western blot and ELISA measured the production and release of CSF1 from human choroidal vascular endothelial cells (HCVECs) under hypoxic conditions. Western blot detected CSF1 released from HCVECs under hypoxic conditions that activated the PI3K/AKT/FOXO1 axis in human macrophages via binding to CSF1R. Transwell migration assay, qRT-PCR, and Western blot detected the effect of CSF1 released from HCVECs on macrophage migration and M2 polarization via the CSF1R/PI3K/AKT/FOXO1 pathway. Incorporation of 5-ethynyl-20-deoxyuridine, transwell migration, and tube formation assays detected the effects of CSF1/CSF1R on the behaviors of HCVECs. Fundus fluorescein angiography (FFA), indocyanine green angiography (ICGA), and immunofluorescence detected the effect of blockade of CSF1/CSF1R on mouse laser-induced CNV. Color fundus photograph, ICGA, and FFA detected CNV lesions in neovascular AMD (nAMD) patients. ELISA detected CSF1 and CSF1R in the aqueous humor of age-related cataract and nAMD patients. Results CSF1 released from HCVECs under hypoxic conditions activated the PI3K/AKT/FOXO1 axis in human macrophages via binding to CSF1R, promoting macrophage migration and M2 polarization via up-regulation of the CSF1R/PI3K/AKT/FOXO1 pathway. Human macrophages promoted the proliferation, migration, and tube formation of HCVECs in a CSF1/CSFR1-dependent manner under hypoxic conditions. CSF1/CSF1R blockade ameliorated the formation of mouse laser-induced CNV. CSF1 and CSF1R were increased in the aqueous humor of nAMD patients. Conclusions Our results affirmed the crucial role of CSF1/CSF1R in boosting the formation of CNV and offered potential molecular targets for the treatment of nAMD.
Collapse
Affiliation(s)
- Yamei Zhou
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jia Zeng
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yuanyuan Tu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lele Li
- Department of Ophthalmology, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Shu Du
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Linling Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaomin Cang
- Department of Endocrinology, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jiajie Lu
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Manhui Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
17
|
Dietze R, Hammoud MK, Gómez-Serrano M, Unger A, Bieringer T, Finkernagel F, Sokol AM, Nist A, Stiewe T, Reinartz S, Ponath V, Preußer C, von Strandmann EP, Müller-Brüsselbach S, Graumann J, Müller R. Phosphoproteomics identify arachidonic-acid-regulated signal transduction pathways modulating macrophage functions with implications for ovarian cancer. Am J Cancer Res 2021; 11:1377-1395. [PMID: 33391540 PMCID: PMC7738879 DOI: 10.7150/thno.52442] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022] Open
Abstract
Arachidonic acid (AA) is a polyunsaturated fatty acid present at high concentrations in the ovarian cancer (OC) microenvironment and associated with a poor clinical outcome. In the present study, we have unraveled a potential link between AA and macrophage functions. Methods: AA-triggered signal transduction was studied in primary monocyte-derived macrophages (MDMs) by phosphoproteomics, transcriptional profiling, measurement of intracellular Ca2+ accumulation and reactive oxygen species production in conjunction with bioinformatic analyses. Functional effects were investigated by actin filament staining, quantification of macropinocytosis and analysis of extracellular vesicle release. Results: We identified the ASK1 - p38δ/α (MAPK13/14) axis as a central constituent of signal transduction pathways triggered by non-metabolized AA. This pathway was induced by the Ca2+-triggered activation of calmodulin kinase II, and to a minor extent by ROS generation in a subset of donors. Activated p38 in turn was linked to a transcriptional stress response associated with a poor relapse-free survival. Consistent with the phosphorylation of the p38 substrate HSP27 and the (de)phosphorylation of multiple regulators of Rho family GTPases, AA impaired actin filament organization and inhibited actin-driven macropinocytosis. AA also affected the phosphorylation of proteins regulating vesicle biogenesis, and consistently, AA enhanced the release of tetraspanin-containing exosome-like vesicles. Finally, we identified phospholipase A2 group 2A (PLA2G2A) as the clinically most relevant enzyme producing extracellular AA, providing further potentially theranostic options. Conclusion: Our results suggest that AA contributes to an unfavorable clinical outcome of OC by impacting the phenotype of tumor-associated macrophages. Besides critical AA-regulated signal transduction proteins identified in the present study, PLA2G2A might represent a potential prognostic tool and therapeutic target to interfere with OC progression.
Collapse
|
18
|
Haider P, Kral-Pointner JB, Mayer J, Richter M, Kaun C, Brostjan C, Eilenberg W, Fischer MB, Speidl WS, Hengstenberg C, Huber K, Wojta J, Hohensinner P. Neutrophil Extracellular Trap Degradation by Differently Polarized Macrophage Subsets. Arterioscler Thromb Vasc Biol 2020; 40:2265-2278. [PMID: 32673525 PMCID: PMC7447175 DOI: 10.1161/atvbaha.120.314883] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Supplemental Digital Content is available in the text. Macrophages are immune cells, capable to remodel the extracellular matrix, which can harbor extracellular DNA incorporated into neutrophil extracellular traps (NETs). To study the breakdown of NETs we studied the capability of macrophage subsets to degrade these structures in vitro and in vivo in a murine thrombosis model. Furthermore, we analyzed human abdominal aortic aneurysm samples in support of our in vitro and in vivo results.
Collapse
Affiliation(s)
- Patrick Haider
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Julia B Kral-Pointner
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Austria (J.B.K.-P., J.W.)
| | - Julia Mayer
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Manuela Richter
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christoph Kaun
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christine Brostjan
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery (C.B., W.E.), Medical University of Vienna, Austria
| | - Wolf Eilenberg
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery (C.B., W.E.), Medical University of Vienna, Austria
| | - Michael B Fischer
- Department of Blood Group Serology and Transfusion Medicine (M.B.F.), Medical University of Vienna, Austria.,Department of Biomedical Research, Danube University Krems, Austria (M.B.F.)
| | - Walter S Speidl
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christian Hengstenberg
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Kurt Huber
- Wilhelminenhospital, 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Vienna, Austria (K.H.).,Sigmund Freud University, Medical Faculty, Vienna, Austria (K.H.)
| | - Johann Wojta
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Austria (J.B.K.-P., J.W.).,Medical University of Vienna, Core Facilities, Austria (J.W.)
| | - Philipp Hohensinner
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| |
Collapse
|