1
|
Nickel KF, Jämsä A, Konrath S, Papareddy P, Butler LM, Stavrou EX, Frye M, Gelderblom M, Nieswandt B, Hammerschmidt S, Herwald H, Renné T. Factor XII-driven coagulation traps bacterial infections. J Exp Med 2025; 222:e20250049. [PMID: 40261297 PMCID: PMC12013512 DOI: 10.1084/jem.20250049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/24/2025] Open
Abstract
Blood coagulation is essential for stopping bleeding but also drives thromboembolic disorders. Factor XII (FXII)-triggered coagulation promotes thrombosis while being dispensable for hemostasis, making it a potential anticoagulant target. However, its physiological role remains unclear. Here, we demonstrate that FXII-driven coagulation enhances innate immunity by trapping pathogens and restricting bacterial infection in mice. Streptococcus pneumoniae infection was more severe in FXII-deficient (F12-/-) mice, with increased pulmonary bacterial burden, systemic spread, and mortality. Similarly, Staphylococcus aureus skin infections and systemic dissemination were exacerbated in F12-/- mice. Reconstitution with human FXII restored bacterial containment. Plasma kallikrein amplifies FXII activation, and its deficiency aggravated S. aureus skin infections, similarly to F12-/- mice. FXII deficiency impaired fibrin deposition in abscess walls, leading to leaky capsules and bacterial escape. Bacterial long-chain polyphosphate activated FXII, triggering fibrin formation. Deficiency in FXII substrate factor XI or FXII/factor XI co-deficiency similarly exacerbated S. aureus infection. The data reveal a protective role for FXII-driven coagulation in host defense, urging caution in developing therapeutic strategies targeting this pathway.
Collapse
Affiliation(s)
- Katrin F. Nickel
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Clinical Chemistry, Department of Molecular Medicine and Surgery, and Center of Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Anne Jämsä
- Clinical Chemistry, Medical Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Praveen Papareddy
- Department of Laboratory Medicine, Biomedical Center (BMC), Lund University, Lund, Sweden
| | - Lynn M. Butler
- Clinical Chemistry, Department of Molecular Medicine and Surgery, and Center of Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Department of Clinical Medicine, The Arctic University of Norway, Tromsø, Norway
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Evi X. Stavrou
- Medicine Service, Section of Hematology-Oncology, Louis Stokes Veterans Administration Medical Center, Cleveland, OH, USA
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Heiko Herwald
- Department of Laboratory Medicine, Biomedical Center (BMC), Lund University, Lund, Sweden
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
2
|
Boulaftali Y, Massberg S, Nicolai L. Platelets in vascular inflammation: fire-fighters or pyromaniacs? Curr Opin Hematol 2025; 32:221-230. [PMID: 40359086 DOI: 10.1097/moh.0000000000000877] [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: 05/15/2025]
Abstract
PURPOSE OF REVIEW In this review, we aim to highlight recent insights into the mechanisms through which platelets contribute to vascular inflammation. We will discuss how platelets interact with other cellular players in the vascular milieu, their role in shaping inflammatory responses, and the potential therapeutic implications of targeting platelet function in inflammatory vascular diseases. RECENT FINDINGS Platelets are essential components in the processes of hemostasis and thrombosis. Their role is now widely acknowledged as far more complex than merely acting as "band-aids" or helping to "clog a pipe". Platelets are now recognized as crucial mediators in inflammatory reactions, particularly in various diseases of the vasculature, where they contribute to the onset and progression of injury. Through their interactions with leukocytes, vascular cells, and by supporting the coagulation cascade, platelets are able to finely regulate the extent and intensity of vascular damage. SUMMARY Recent findings underscore the remarkable diversity and functionality of platelets in vascular diseases. Mechanistic studies in preclinical models reveal promising therapeutic opportunities, which require further validation before being translated into clinical practice.
Collapse
Affiliation(s)
- Yacine Boulaftali
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
3
|
Andraska E, Fields A, Nunez-Garcia B, Moore E, Wade CE, Knudson MM, Neal MD, Kornblith L, CLOTT (Consortium of Leaders in the Study of Traumatic Thromboembolism) Study Group. Platelet flux in trauma-associated venous thromboembolism: A secondary analysis of the Consortium of Leaders in the Study of Traumatic Thromboembolism (CLOTT) studies. J Trauma Acute Care Surg 2025; 98:957-965. [PMID: 40107968 PMCID: PMC12097933 DOI: 10.1097/ta.0000000000004592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BACKGROUND Venous thromboembolism (VTE) after traumatic injury is morbid. Evaluating changes in platelets in injured patients who develop VTE could identify platelet-based strategies for management of thrombotic complications after injury. METHODS In a prospectively designed secondary analysis of a multicenter cohort study conducted by the Consortium of Leaders in the Study of Traumatic Thromboembolism (CLOTT1) study group, injured patients aged 18 to 40 years admitted for a minimum of 48 hours with at least one risk factor for VTE were evaluated. A subset of CLOTT1 patients had platelet aggregometry and thromboelastography performed to examine platelet function (CLOTT2). Patients who developed VTE were compared with those who did not. RESULTS Of 7,805 patients from CLOTT1 (mean [SD] age, 29.1 [6.4] years; 1,987 [25.5%] female), 425 (5.4%) developed VTE. Platelet count was lower at each time point for patients with VTE (admission: 242 [234-251] vs. 254 [252-256], p < 0.01; hospital day 1: 157 [150-164] vs. 197 [195-198], p < 0.01; all counts ×10 9 /L). An initial 10-point reduction in platelet count was associated with development of VTE (odds ratio, 1.32 [1.13-1.53]; p < 0.01) controlling for shock, injury severity, coagulopathy, sex, and product transfusion. When evaluating 129 CLOTT2 patients, velocity of platelet aggregation was higher on admission in the VTE group (18.5 vs. 12.8 aggregation units/min; p < 0.01) in response to adenosine diphosphate stimulation. In response to thrombin stimulation, velocity of platelet aggregation was higher at 48 hours (34.4 vs. 12.3 aggregation units/min; p < 0.01), and overall aggregation was higher in the VTE group at 72 hours (area under the curve, 173.2 vs. 129.6; p < 0.01). Thromboelastography results were not different between groups. CONCLUSION This study identified an association of early reduction in platelet count with the development of VTE in injured patients at risk for VTE. P2Y 1/2 and protease-activated receptor 1 receptor stimulation changes in platelet aggregation responses are altered in VTE patients. Interrogating platelet count and functional responses may be beneficial in evaluating thrombotic complications after injury. LEVEL OF EVIDENCE Diagnostic Test/Criteria; Level III.
Collapse
Affiliation(s)
- Elizabeth Andraska
- From the University of Pittsburgh, Pittsburgh Trauma and Transfusion Medicine Research Center (E.A., M.D.N.), University of California, San Francisco and Zuckerberg San Francisco General Hospital (B.N.-G., A.F., M.M.K, L.K.), Ernest E Moore Shock Trauma Center at Denver Health (E.M.), University of Texas Health Sciences Center, Houston (C.E.W.)
| | | | | | | | | | | | | | | | | |
Collapse
Collaborators
Scott Brakenridge, Sherry L Sixta, Brandon R Bruns, Thomas M Scalea, Todd W Costantini, Bruce A Crookes, Elliott R Haut, George C Velmahos, Andrew Kerwin, Alicia Mohr, Lazio N Kiraly, David A Spain, Lisa M Knowlton, Matthew J Martin, David J Milia, Ram Nirula, Frederick B Rogers, Charles E Wade, Michelle K McNutt, Mark D Cipolle,
Collapse
|
4
|
Tang S, Ye JX, Li RY, Wang JL, Xie HC, Zhang YQ, Wang M, Sun GB. Formononetin attenuates myocardial ischemia/reperfusion injury by regulating neutrophil extracellular traps formation and platelet activation via platelet CD36. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156736. [PMID: 40250000 DOI: 10.1016/j.phymed.2025.156736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/11/2025] [Accepted: 04/06/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Prothrombotic and proinflammatory responses are crucial in the pathology of myocardial ischemia-reperfusion injury (MIRI). Platelets and neutrophil extracellular traps (NETs) are essential to linking inflammation with thrombosis. Formononetin (FMN), an isoflavone extracted from Astragalus membranaceus, has anti-inflammatory and anti-thrombotic effects and confers benefits on MIRI. However, the mechanisms of FMN against MIRI remain unclear. PURPOSE This study explored FMN's roles and mechanisms in modulating platelet activation and NETs formation to mitigate MIRI. STUDY DESIGN AND METHODS A rat model of MIRI by the left anterior descending coronary artery ligation was utilized to evaluate the role of FMN. 60 Sprague-Dawley male rats were randomly divided into 7 groups. Proteomics, flow cytometry, immunofluorescence, ELISA, and western blotting assays were performed to reveal the potential mechanisms of FMN. Neutrophils treated with platelet-rich plasma were applied to further explore the mechanisms of FMN in vitro. RESULTS We showed that FMN administration declined myocardial infarct size and improved cardiac function. Moreover, FMN significantly reduced MIRI-induced platelet activation including platelet aggregation, platelet adhesion, platelet granule secretion, and platelet-leukocyte aggregation without affecting tail bleeding time. Additionally, FMN inhibited microthrombus, platelet-neutrophil aggregation, and NETs formation in myocardial tissue. Mechanistically, FMN attenuated MIRI-induced CD36 expression and phosphorylation of ERK5 in platelets. Furthermore, up-regulation of CD36 content in vitro counteracted the potency of FMN to inhibit platelet activation and NETs formation. CONCLUSION FMN mitigates thrombosis and inflammation in MIRI by inhibiting platelet activation and NETs formation via the CD36 pathway. This research offers important insights for future studies on MIRI prevention.
Collapse
Affiliation(s)
- Shuang Tang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China
| | - Jing-Xue Ye
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China
| | - Ruo-Yun Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China
| | - Jia-Lu Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China
| | - Hao-Chen Xie
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China
| | - Ya-Qi Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China
| | - Min Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China.
| | - Gui-Bo Sun
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100193, PR China; Key Laboratory of new drug discovery based on Classic Chinese medicine prescription, Chinese Academy of Medical Sciences, Beijing, 100193, PR China.
| |
Collapse
|
5
|
Wu W, Tong D, Xia W, Song B, Li G, Zhou L, Xie F, Zhang C, Liu Y, Wang H, Du Z, Shao Y, Li J. Procoagulant Effect of Neutrophil Extracellular Traps, Activated Platelets, and Endothelial Cells in Patients After TAVR. Arterioscler Thromb Vasc Biol 2025; 45:1006-1019. [PMID: 40177776 DOI: 10.1161/atvbaha.124.322376] [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: 12/23/2024] [Accepted: 03/13/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Patients with severe aortic stenosis, undergoing transcatheter aortic valve replacement (TAVR), are more likely to develop thrombotic complications. However, the definite mechanisms underlying the hypercoagulation state remain unclear to date. Our objectives were to explore whether and how neutrophil extracellular traps (NETs) play a procoagulant role in patients after TAVR alone or TAVR with percutaneous coronary intervention within 1 year and further to evaluate their interactions with platelets and endothelial cells. METHODS The levels of plasma NETs, platelets, and endothelial cell activation markers were analyzed by ELISA. NET formation was observed by immunofluorescence. Procoagulant activity was measured by clotting time, fibrin, and TAT (thrombin-antithrombin) complex generation assays. Phosphatidylserine exposure on cells was assessed by flow cytometry. RESULTS Compared with pre-TAVR, controls, or severe aortic stenosis without TAVR patients, the plasma NET levels in patients after TAVR alone, especially TAVR with percutaneous coronary intervention, increased from 7 days, peaking at 3 months, and then gradually decreased until the 12th month. Furthermore, neutrophils and plasma from patients post-TAVR are more prone to promote NET formation; NETs from these patients markedly decreased clotting time and increased fibrin and TAT generation. Additionally, a high concentration of NETs induced platelet aggregation and exerted a strong cytotoxic effect on endothelial cells and transformed them into a procoagulant phenotype. CONCLUSIONS These results lead us to believe that NETs contribute to the hypercoagulability in patients post-TAVR. Our study may provide a new target for preventing thrombotic complications in patients post-TAVR by blocking NET generation.
Collapse
Affiliation(s)
- Wei Wu
- Department of Cardiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, China (W.W.)
| | - Dongxia Tong
- Departments of Oncology (D.T.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Wei Xia
- Cardiology (W.X., F.X., C.Z., Y.L., H.W., Y.S., J.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Bin Song
- Department of Geriatrics, Tai'an City Second Hospital of Traditional Chinese Medicine, China (B.S.)
| | - Guangwen Li
- Rheumatology and Immunology (G.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Lihui Zhou
- Department of General Surgery, China-Japan Union Hospital of Jilin University, Norman Bethune Health Science Center, Jilin University, Changchun (L.Z.)
| | - Fangyu Xie
- Cardiology (W.X., F.X., C.Z., Y.L., H.W., Y.S., J.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Chunquan Zhang
- Cardiology (W.X., F.X., C.Z., Y.L., H.W., Y.S., J.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Yvhao Liu
- Cardiology (W.X., F.X., C.Z., Y.L., H.W., Y.S., J.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Haiyang Wang
- Cardiology (W.X., F.X., C.Z., Y.L., H.W., Y.S., J.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Zhaona Du
- Department of Cardiology, Qingdao Municipal Hospital, School of Clinical Medicine, Weifang Medical University, China (Z.D.)
| | - Yibing Shao
- Cardiology (W.X., F.X., C.Z., Y.L., H.W., Y.S., J.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| | - Jihe Li
- Cardiology (W.X., F.X., C.Z., Y.L., H.W., Y.S., J.L.), Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, China
| |
Collapse
|
6
|
Nakazawa D, Masuda S, Nishibata Y, Watanabe-Kusunoki K, Tomaru U, Ishizu A. Neutrophils and NETs in kidney disease. Nat Rev Nephrol 2025; 21:383-398. [PMID: 40102634 DOI: 10.1038/s41581-025-00944-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Neutrophils, conventionally regarded as a homogeneous immune cell population, have emerged as a heterogeneous group of cells with distinct gene profiles and immune properties. Activated neutrophils release a spectrum of bioactive substances, including cytokines, chemokines, proteolytic enzymes, reactive oxygen species and neutrophil extracellular traps (NETs), which are composed of decondensed DNA and antimicrobial proteins. NETs have a pivotal role in innate immunity, including in preventing the ascent of uropathogenic bacteria into the kidneys, as they efficiently trap pathogenic microorganisms. However, although indispensable for defence against pathogens, NETs also pose risks of self-damage owing to their cytotoxicity, thrombogenicity and autoantigenicity. Accordingly, neutrophils and NETs have been implicated in the pathogenesis of various disorders that affect the kidneys, including acute kidney injury, vasculitis, systemic lupus erythematosus, thrombotic microangiopathy and in various aetiologies of chronic kidney disease. Pathological alterations in the glomerular vascular wall can promote the infiltration of neutrophils, which can cause tissue damage and inflammation through their interactions with kidney-resident cells, including mesangial cells and podocytes, leading to local cell death. Targeting neutrophil activation and NET formation might therefore represent a new therapeutic strategy for these conditions.
Collapse
Affiliation(s)
- Daigo Nakazawa
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sakiko Masuda
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yuka Nishibata
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Kanako Watanabe-Kusunoki
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
7
|
Sato Y, Kato Y, Kanoke A, Sun JY, Nishijima Y, Wang RK, Stryker M, Endo H, Liu J. Type 2 diabetes abates retrograde collateral flow and promotes leukocyte adhesion following ischemic stroke. J Cereb Blood Flow Metab 2025:271678X251338203. [PMID: 40439073 PMCID: PMC12122487 DOI: 10.1177/0271678x251338203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/25/2025] [Accepted: 04/07/2025] [Indexed: 06/02/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with impaired leptomeningeal collateral compensation and poor stroke outcome. Neutrophils tethering and rolling on endothelium after stroke can also independently reduce flow velocity. However, the chronology and topological changes in collateral circulation in T2DM is not yet defined. Here, we describe the spatial and temporal blood flow dynamics and vessel diameter changes in pial arteries and veins and leukocyte-endothelial adhesion following middle cerebral artery (MCA) stroke using two-photon microscopy in awake control and T2DM mice. Relative to control mice, T2DM mice already exhibited smaller pial vessels with reduced flow velocity prior to stroke. Following stroke, T2DM mice displayed persistently reduced blood flow in pial arteries and veins, resulting in a poor recovery of downstream penetrating arterial flow and a sustained deficit in microvascular flow. There was also persistent increase of leukocyte adhesion to the endothelium of veins, coincided with elevated neutrophils infiltration into brain parenchyma in T2DM mice compared to control mice after stroke. Our data suggest that T2DM-induced increase in inflammation and chronic remodeling of leptomeningeal vessels may contribute to the observed hemodynamics deficiency after stroke and subsequent poor stroke outcome.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yuya Kato
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Atsushi Kanoke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jennifer Y Sun
- Institute of Ophthalmology, University College London, London, UK
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Yasuo Nishijima
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ruikang K Wang
- Department of Bioengineering, College of Engineering and School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Michael Stryker
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Hidenori Endo
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jialing Liu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- SFVAMC, San Francisco, CA, USA
| |
Collapse
|
8
|
Dinc R, Ardic N. Relationship Between Neutrophil Extracellular Traps and Venous Thromboembolism: Pathophysiological and Therapeutic Role. Br J Hosp Med (Lond) 2025; 86:1-15. [PMID: 40405858 DOI: 10.12968/hmed.2024.0660] [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: 05/24/2025]
Abstract
Venous thromboembolism (VTE), which includes pulmonary embolism (PE) and deep vein thrombosis (DVT), is a serious vascular disease that ranks third in cardiovascular-related deaths. Inflammation along with neutrophil extracellular traps (NETs) play a key role in the pathophysiology of VTE. This review focuses on articles that that evaluate the role of NETs in the development of VTE and their potential as therapeutic targets. Research has demonstrated that when NETs become overactivated, they take part in thrombotic activities, which is the opposite of their defensive functions under healthy conditions. When endothelial cells are activated, neutrophils are recruited very early, releasing NETs and initiating a thrombotic process. NETs promote thrombosis by directly activating factor XII (FXII), ultimately triggering platelet recruitment, and initiating the intrinsic coagulant pathway. Subsequently, monocytes and factors such as tissue factor join the process, further increasing NET formation, the inflammatory reactions and progression of venous thrombus. NETs play a crucial part in the intricate interaction between inflammation and thrombosis, where each triggers the other. High levels of NETs also correlate with the severity of VTE. These properties of NETs make them potential therapeutic targets for VTE prevention and treatment. This article aims to describe NETs, their occurrence, and how they relate to VTE. Taking into account what is now known, the function of NETs as targets for treatment in VTE using various approaches, the benefits and drawbacks of these approaches, and suggestions for the future are examined.
Collapse
Affiliation(s)
- Rasit Dinc
- INVAMED Medical Innovation Institute, New York, NY, USA
| | - Nurittin Ardic
- Med-International UK Health Agency Ltd., Leicestershire, UK
| |
Collapse
|
9
|
Ebrahimi R, Kalantari T, Sarkari F, Nematzadeh M, Sharifi MJ, Mohammadian K, Alipour P, Safari F, Namdari S, Borhani-Haghighi A. Serum levels of inflammatory markers, sP-selectin, IL-1β, IL-6, and hsCRP are positively correlated with tissue factor transcript level of peripheral blood mononuclear cells in stroke. Blood Coagul Fibrinolysis 2025:00001721-990000000-00205. [PMID: 40396731 DOI: 10.1097/mbc.0000000000001370] [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: 01/20/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVES Stroke is an injury occurring due to a sudden interruption of blood supply to the brain. It is the leading cause of disability worldwide and the second most prevalent cause of mortality. The objective of this study is whether momentous inflammatory and coagulation markers may have a correlation with each other in stroke patients. MATERIAL AND METHODS Sixty stroke patients and twenty sex-age matched normal controls were sampled. Interleukin (IL)-6, IL-1β, sP-selectin, and high-sensitivity C-reactive protein (hsCRP), key inflammatory markers, were selected and measured by ELISA, and tissue factor gene expression level was evaluated by real-time PCR in peripheral blood mononuclear cells. RESULTS The serum levels of sP-selectin, IL-1β, IL-6, and hsCRP increased significantly in patients (P-value < 0.05). In the patient group, a significant correlation was observed between these inflammatory markers and coagulant tissue factor gene expression in peripheral blood mononuclear cells (P-values < 0.05), while it was not significant in the control group. CONCLUSION This study proposed that the main inflammatory markers in connection with tissue factor may play a role in the occurrence of thrombosis in stroke patients. Therefore, targeting and inhibiting the key inflammatory factors along with existing anticoagulants may greatly reduce the complications associated with stroke.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- Student Research Committee
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences
| | - Tahereh Kalantari
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences
| | - Fatemeh Sarkari
- Student Research Committee
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences
| | - Mahdieh Nematzadeh
- Student Research Committee
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences
| | - Mohammad Jafar Sharifi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences
| | - Kiana Mohammadian
- Student Research Committee
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences
| | - Parisa Alipour
- Student Research Committee
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences
| | - Sepide Namdari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences
| | | |
Collapse
|
10
|
Zhang J, Wang N, Xin T, Zhu X, Lang S, Ge X. Liquiritin mitigates lower extremity deep vein thrombosis by inhibiting inflammation and oxidative stress via the NF-κB signaling pathway. Thromb J 2025; 23:51. [PMID: 40394684 PMCID: PMC12090432 DOI: 10.1186/s12959-025-00739-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Lower extremity deep vein thrombosis (LEDVT) is a common vascular disease, with its pathogenesis mainly involving inflammatory responses and oxidative stress. Liquiritin (LIQ) is a flavonoid that exhibits pharmacological effects such as anti-inflammatory and antioxidant properties. This study aimed to investigate the role of LIQ in LEDVT and its potential mechanisms. METHODS We established an LEDVT model in mice by ligating the inferior vena cava (IVC) and performed in vitro experiments by stimulating human umbilical vein endothelial cells (HUVECs) with IL-1β (10 ng/mL) to simulate endothelial cell injury. RESULTS We found that LIQ significantly reduced the size and weight of thrombi and decreased the concentrations of inflammatory factors TNF-α and IL-6 in the IVC of LEDVT mice. Furthermore, LIQ inhibited the secretion of prothrombotic mediators such as tissue factor (TF) and vascular cell adhesion molecule-1 (VCAM-1). Administration of LIQ resulted in a notable reduction in immune inflammatory cells in the IVC of LEDVT mice. LIQ also demonstrated antioxidant properties, as the treatment of LIQ enhanced SOD activity and restored ROS levels to normal in the IVC. Similarly, LIQ reduced the formation of inflammatory factors and the secretion of prothrombotic mediators by HUVECs while inhibiting oxidative stress in HUVECs. Finally, LIQ effectively suppressed the levels of phosphorylated p65 in both the IVC and HUVECs. CONCLUSIONS LIQ reduces inflammatory responses and oxidative stress in LEDVT by inhibiting the NF-κB signaling pathway. This finding provides new insights into the prevention and treatment of LEDVT.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Emergency and Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Nan Wang
- Department of Emergency and Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Tianyou Xin
- Department of Ultrasound, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, People's Republic of China
| | - Xiaojun Zhu
- Department of Vascular Surgery, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, People's Republic of China
| | - Shengkun Lang
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Fuxing Road 28, Beijing, People's Republic of China.
| | - Xin Ge
- Department of Emergency and Critical Care Medicine, Wuxi 9th People's Hospital Affiliated to Soochow University, Liangxi Road 999, Wuxi, Jiangsu, 214000, People's Republic of China.
- Orthopedic Institution of Wuxi City, Liangxi Road 999, Wuxi, Jiangsu, 214000, People's Republic of China.
| |
Collapse
|
11
|
Sun GJ, Xu F, Jiao XY, Yin Y. Advances in research of neutrophil extracellular trap formation in osteoarticular diseases. World J Orthop 2025; 16. [DOI: 10.5312/wjo.v16.i5.106377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025] Open
Abstract
Neutrophil extracellular traps (NETs) have been the subject of research in the field of innate immunity since they were first described two decades ago. NETs are fibrous network structures released by neutrophils under specific stimuli, including DNA, histones, and a variety of granular proteins. NETs have been widely studied in the fields of infectious and immune diseases, and new breakthroughs have been made in the understanding of disease pathogenesis and treatment. In recent years, studies have found that NETs play an important role in the occurrence and development of osteoarticular diseases. This article reviews the progress in the research of NETs in common osteoarticular diseases such as rheumatoid arthritis, ankylosing spondylitis, gouty arthritis, osteonecrosis of the femoral head, osteoarthritis, and joint fibrosis, including the formation mechanism of NETs and its role in inflammation, joint destruction, pain and other pathological processes. The problems existing in current research are discussed, along with future research directions, to provide a reference for the in-depth study of osteoarticular diseases and the development of new treatment strategies.
Collapse
Affiliation(s)
- Guan-Jun Sun
- Department of Joint and Sports Medicine, Suining Central Hospital, Suining 629000, Sichuan Province, China
| | - Feng Xu
- Department of Joint and Sports Medicine, Suining Central Hospital, Suining 629000, Sichuan Province, China
| | - Xiao-Yi Jiao
- Department of Joint and Sports Medicine, Suining Central Hospital, Suining 629000, Sichuan Province, China
| | - Yi Yin
- Department of Joint and Sports Medicine, Suining Central Hospital, Suining 629000, Sichuan Province, China
| |
Collapse
|
12
|
Blümke J, Schameitat M, Verma A, Limbecker C, Arlt E, Kessler SM, Kielstein H, Krug S, Bazwinsky-Wutschke I, Haemmerle M. Innate Immunity and Platelets: Unveiling Their Role in Chronic Pancreatitis and Pancreatic Cancer. Cancers (Basel) 2025; 17:1689. [PMID: 40427186 PMCID: PMC12110028 DOI: 10.3390/cancers17101689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic instability, acinar-ductal metaplasia, and pancreatic intraepithelial neoplasia (PanIN) formation. Components of the extracellular matrix, including immune cells, can modulate this progression phase. This includes cells of the innate immune system, such as natural killer (NK) cells, macrophages, dendritic cells, mast cells, neutrophils, and myeloid-derived suppressor cells (MDSCs), either promoting or inhibiting tumor growth. On one hand, innate immune cells can trigger inflammatory responses that support tumor progression by releasing cytokines and growth factors, fostering tumor cell proliferation, invasion, and metastasis. On the other hand, they can also activate immune surveillance mechanisms, which can limit tumor development. For example, NK cells are cytotoxic innate lymphoid cells that are able to kill tumor cells, and active dendritic cells are crucial for a functioning anti-tumor immune response. In contrast, mast cells and MDSCs rather support a pro-tumorigenic tumor microenvironment that is additionally sustained by platelets. Once thought to play a role in hemostasis only, platelets are now recognized as key players in inflammation and cancer progression. By releasing cytokines, growth factors, and pro-angiogenic mediators, platelets help shape an immunosuppressive microenvironment that promotes fibrotic remodeling, tumor initiation, progression, metastasis, and immune evasion. Neutrophils and macrophages exist in different functional subtypes that can both act pro- and anti-tumorigenic. Understanding the complex interactions between innate immune cells, platelets, and early precursor lesions, as well as PDAC cells, is crucial for developing new therapeutic approaches that can harness the immune and potentially also the coagulation system to target and eliminate tumors, offering hope for improved patient outcomes.
Collapse
Affiliation(s)
- Juliane Blümke
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| | - Moritz Schameitat
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Atul Verma
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
| | - Celina Limbecker
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sonja M. Kessler
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Faculty of Natural Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sebastian Krug
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Monika Haemmerle
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| |
Collapse
|
13
|
Liu S, Shen Y, Chen J, Ruan Z, Hua L, Wang K, Xi X, Mao J. The critical role of platelets in venous thromboembolism: Pathogenesis, clinical status, and emerging therapeutic strategies. Blood Rev 2025:101302. [PMID: 40382294 DOI: 10.1016/j.blre.2025.101302] [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: 03/03/2025] [Revised: 05/08/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
Venous thromboembolism (VTE), encompassing deep vein thrombosis (DVT) and pulmonary embolism (PE), is a complex vascular disorder with high morbidity and mortality, driven by Virchow's Triad: blood stasis, hypercoagulability, and endothelial injury. VTE is now recognized as an inflammatory process involving multiple components. Platelets are involved in the process of VTE, contributing to thrombosis initiation, progression, resolution and recurrence through coagulation activation, and interactions with immune and endothelial cells. Anticoagulation remains the cornerstone of VTE treatment; however, antiplatelet agents like aspirin have demonstrated therapeutic potential, particularly following major orthopedic surgeries. Furthermore, emerging platelet-targeted therapies and biomarkers offer new opportunities for improving VTE diagnosis and treatment. This review explores the evolving role of platelets in VTE pathophysiology, assesses current antiplatelet strategies, and highlights novel therapeutic approaches. Advancing platelet research in VTE may lead to safer, more effective interventions, optimizing outcomes for patients with this life-threatening condition.
Collapse
Affiliation(s)
- Shuang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan Shen
- Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiayi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zheng Ruan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Hua
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xiaodong Xi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jianhua Mao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
14
|
Xiang Y, Wu Y, Liu H, Chen Z, Pan J. Association of coagulation-related indicators with postoperative venous thromboembolism occurrence in patients with pituitary tumors. Sci Rep 2025; 15:16694. [PMID: 40368990 PMCID: PMC12078525 DOI: 10.1038/s41598-025-01029-8] [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: 08/16/2024] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
This study aimed to analyze risk factors for postoperative VTE in pituitary tumor resection patients, focusing on coagulation indicators and their predictive value. This study collected clinical data from 300 patients who underwent pituitary adenoma resection from January 2021 to August 2023 in the Department of Neurosurgery, the Second Clinical College of the Army Medical University, China. Logistic regression modeling was used to identify risk factors for VTE. Restricted cubic spline curves were used to characterize the dose-response relationship between coagulation-related indicators and the risk of venous thromboembolism. The area under the curve (AUC) was calculated using the receiver operating characteristic (ROC) curve to evaluate the predictive power of coagulation-related indicators. Multivariate analysis showed that D-dimer, platelet count and hemoglobin (Hb) were significant predictors of VTE with OR (95% CI) of 1.967 (1.441-2.808), 1.020 (1.013-1.029), and 0.952 (0.914-0.994), respectively. The AUCs for D-dimer, Platelet Count, and Hb were 0.708, 0.731, and 0.712, respectively. The AUC for combining the three coagulation indices was the largest, 0.838. The combined use of D-dimer, Hb, and platelet count can identify high-risk patients early, enabling timely implementation of antithrombotic strategies.
Collapse
Affiliation(s)
- Yi Xiang
- Chongqing General Hospital, Chongqing University, 118 Star Avenue, Liangjiang New Area, Chongqing, China
| | - Ya Wu
- The Second Clinical College of the Army Medical University, Chongqing, China
| | - Huan Liu
- The Second Clinical College of the Army Medical University, Chongqing, China
| | - Zheng Chen
- The Second Clinical College of the Army Medical University, Chongqing, China
| | - JinYu Pan
- Chongqing General Hospital, Chongqing University, 118 Star Avenue, Liangjiang New Area, Chongqing, China.
| |
Collapse
|
15
|
Blair KM, Bohinc DJ, Bane KL, Warnock M, Abuaita B, Gura C, Grinsztejn E, Marshall SH, Wilson BM, Bonomo RA, Tambralli A, Knight JS, O'Riordan MX, Lawrence DA, Stavrou EX, Sandkvist M. Acinetobacter Baumannii Secreted Protease CpaA Inhibits Factor XII-Mediated Bradykinin Generation and Neutrophil Activation. Circ Res 2025. [PMID: 40357548 DOI: 10.1161/circresaha.124.324764] [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: 04/19/2024] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND FXII (coagulation factor XII) is best known for its roles in the contact and kallikrein-kinin pathways. FXII is converted to its active enzyme (FXIIa [activated factor XII]) by PKa (plasma kallikrein) or its unique ability to autoactivate on bacterial or other biologic surfaces. In vivo, FXIIa initiates the intrinsic coagulation pathway and promotes inflammation by reciprocal activation of prekallikrein, which cleaves HK (high-molecular-weight kininogen) to liberate bradykinin. CpaA (coagulation targeting metallo-endopeptidase of A baumannii) is a secreted metalloprotease identified in a human clinical isolate of Acinetobacter baumannii that cleaves FXII at O-linked glycosylated sites, inhibiting contact activation. While CpaA facilitates a modest in vivo fitness advantage in mice, the role of CpaA in human infection remains unclear. As such, the objectives of this study were to characterize the structural details of the interaction between CpaA, FXII, and the KKSs (kallikrein-kinin systems) and to determine the downstream consequences on thromboinflammatory responses. METHODS The effect of purified CpaA on the coagulant activity of FXII and the generation of bradykinin was characterized. Neutrophil signaling, flow cytometry, and functional assays were performed to define how CpaA-mediated cleavage of FXII affects innate immune functions. Bacterial killing by human neutrophils was performed with wild-type and mutant A baumannii strains lacking CpaA. RESULTS We found that CpaA cleaves both FXII zymogen and FXIIa but not beta Factor XII. However, cleavage of FXIIa by CpaA does not significantly inhibit its clotting activity, demonstrating that CpaA does not inactivate FXIIa, but rather prevents activation of zymogen FXII. CpaA also cleaves HK, resulting in reduced kallikrein activation and bradykinin generation. We previously identified that zymogen FXII interacts with the urokinase receptor on neutrophils and upregulates neutrophil activation. Here, we demonstrate that CpaA cleaves neutrophil FXII, resulting in reduced Akt2 phosphorylation, chemotaxis, oxidative burst, and neutrophil extracellular trap formation. Importantly, CpaA decreases the human neutrophil killing efficiency of A baumannii in culture. CONCLUSIONS These data identify a role for FXII in responding to bacterial infection and suggest that by inhibiting the contact and KKSs and impairing neutrophil activation, CpaA may blunt the innate immune response and help prevent the elimination of A baumannii from the human host.
Collapse
Affiliation(s)
- Kris M Blair
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
- Now with Fred Hutch/University of Washington/Seattle Children's Cancer Consortium (K.M.B.)
| | - Dillon J Bohinc
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
| | - Kara L Bane
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
| | - Mark Warnock
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (M.W., D.A.L.)
| | - Basel Abuaita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge (B.A.)
| | - Colby Gura
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| | - Eduarda Grinsztejn
- Hematology and Oncology Division, Department of Medicine, University Hospitals Cleveland Medical Center, OH (E.G.)
| | - Steven H Marshall
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
| | - Brigid M Wilson
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
| | - Robert A Bonomo
- Research Service, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH. (S.H.M., B.M.W., R.A.B.)
- Clinician Scientist Investigator, Louis Stokes Cleveland Department, Veterans Affairs Medical Center, Cleveland, OH.(R.A.B.)
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- Department of Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH. (R.A.B.)
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), OH (R.A.B.)
| | - Ajay Tambralli
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (A.T., J.S.K.)
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (A.T., J.S.K.)
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor. (M.W., D.A.L.)
| | - Evi X Stavrou
- Hematology and Oncology Division, Department of Medicine, Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH (D.J.B., K.L.B., E.X.S.)
- Medicine Service, Section of Hematology-Oncology, Louis Stokes Veterans Administration Medical Center, Cleveland, OH (E.X.S.)
| | - Maria Sandkvist
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor. (K.M.B., B.A., C.G., M.X.O., M.S.)
| |
Collapse
|
16
|
Xie P, Liu Y, Bai P, Ming Y, Zheng Q, Zhu L, Qi Y. TMEM132A: a novel susceptibility gene for lung adenocarcinoma combined with venous thromboembolism identified through comprehensive bioinformatic analysis. Front Oncol 2025; 15:1564114. [PMID: 40432920 PMCID: PMC12107398 DOI: 10.3389/fonc.2025.1564114] [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: 01/21/2025] [Accepted: 04/09/2025] [Indexed: 05/29/2025] Open
Abstract
Background Mounting evidence indicates that lung adenocarcinoma (LUAD) patients are at elevated risk for venous thromboembolism (VTE), presenting a major clinical challenge. This study utilized public databases to identify crosstalk genes (CGs) between LUAD and VTE, applied machine learning methods to discover shared diagnostic biomarkers, and explored their underlying mechanisms. Methods Disease-specific genes for VTE were extracted from comprehensive genomic databases (CTD, DisGeNET, GeneCards, OMIM), while transcriptomic profiles of LUAD and VTE cohorts were retrieved from GEO via GEOquery implementation. Molecular crosstalk analysis identified candidate genes through differential expression algorithms and disease-association metrics. Functional annotation employed GO and KEGG analyses to elucidate the biological significance of identified CGs. LASSO regression analysis of VTE and LUAD matrices yielded overlapping diagnostic biomarkers. Immune contexture was characterized via CIBERSORT deconvolution, followed by correlation analyses between hub genes and immune infiltration profiles. Hub genes expression was corroborated through independent cohort validation and serological quantification. Diagnostic utility was evaluated through receiver operating characteristic (ROC) curve and nomogram. Therapeutic potential was assessed via DSigDB-based drug sensitivity profiling. Result Through transcriptomic analysis, we identified 381 CGs, which demonstrated significant enrichment in inflammatory cascades, immunological processes, and coagulation pathways. LASSO regression analysis of LUAD and VTE cohorts revealed TIMP1 and TMEM132A as putative shared diagnostic biomarkers. TMEM132A exhibited significant correlation with immune cell infiltration patterns across both diseases, modulating the immune microenvironment. Validation cohorts and serological assessment confirmed elevated TMEM132A expression in LUAD and LUAD combined with VTE phenotypes. The diagnostic accuracy of TMEM132A was substantiated by ROC curves and nomogram analyses. Pharmacological sensitivity analysis indicated that TMEM132A may serve as a potential target for the therapeutic agents birabresib and abemaciclib. Conclusion TMEM132A demonstrates diagnostic utility as a predictive biomarker for VTE occurrence in LUAD, suggesting its potential role as a susceptibility gene in this patient cohort.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yong Qi
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital CN, Zhengzhou, China
| |
Collapse
|
17
|
Pekayvaz K, Kilani B, Joppich M, Eivers L, Brambs S, Knottenberg V, Akgöl S, Yue K, Li L, Martinez-Navarro A, Kaiser R, Meißner N, Schulz H, Belz L, Akhalkatsi A, Stockhausen S, Mueller TT, Millonig S, Hartelt L, Gold C, Janjic A, Polewka V, Wendler F, Droste Zu Senden A, Titova A, Leunig A, Voelkl M, Engelmann B, Hernandez Petzsche MR, Boeckh-Behrens T, Liebig T, Winning S, Fandrey J, Dichgans M, Enard W, Zimmer R, Tiedt S, Massberg S, Nicolai L, Stark K. Immunothrombolytic monocyte-neutrophil axes dominate the single-cell landscape of human thrombosis and correlate with thrombus resolution. Immunity 2025; 58:1343-1358.e13. [PMID: 40280129 DOI: 10.1016/j.immuni.2025.03.020] [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: 08/30/2024] [Revised: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Thrombotic diseases remain the major cause of death and disability worldwide, and the contribution of inflammation is increasingly recognized. Thromboinflammation has been identified as a key pathomechanism, but an unsupervised map of immune-cell states, trajectories, and intercommunication at a single-cell level has been lacking. Here, we reveal innate leukocyte substates with prominent thrombolytic properties by employing single-cell omics measures on human stroke thrombi. Using in vivo and in vitro thrombosis models, we propose a pro-resolving monocyte-neutrophil axis, combining two properties: (1) NR4A1hi non-classical monocytes acquire a thrombolytic and neutrophil-chemoattractive phenotype, and (2) blood neutrophils are thereby continuously recruited to established thrombi through CXCL8-CXCR1 and CXCR2 and adopt a hypoxia-induced thrombus-resolving urokinase receptor (PLAUR)+ phenotype. This immunothrombolytic axis results in thrombus resolution. Together, with this immune landscape of thrombosis, we provide a valuable resource and introduce the concept of "immunothrombolysis" with broad mechanistic and translational implications at the crossroad of inflammation and thrombosis.
Collapse
Affiliation(s)
- Kami Pekayvaz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Badr Kilani
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luke Eivers
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Sophia Brambs
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sezer Akgöl
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Keyang Yue
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Li
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Rainer Kaiser
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Nina Meißner
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Heiko Schulz
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Larissa Belz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sven Stockhausen
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Tonina T Mueller
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Simon Millonig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lea Hartelt
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Franziska Wendler
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Anna Titova
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Leunig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Voelkl
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Moritz R Hernandez Petzsche
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boeckh-Behrens
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Liebig
- Institute for Diagnostic and Interventional Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Sandra Winning
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Joachim Fandrey
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Martin Dichgans
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Konstantin Stark
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
18
|
Yan X, Huang W, Chen Y. Serum MPO-DNA for Predicting the Risk of Venous Thromboembolism and the Effect of Statins in Patients with Spontaneous Intracerebral Hemorrhage. Thromb Haemost 2025. [PMID: 40294601 DOI: 10.1055/a-2595-1927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Patients with spontaneous intracerebral hemorrhage (ICH) are at high risk of venous thromboembolism (VTE). Recent studies have shown the involvement of neutrophil extracellular traps (NETs) in thrombogenesis.To explore the predictive value of serum MPO-DNA (a NETs surrogate) for VTE and the effect of statins on serum MPO-DNA levels and the VTE incidence in ICH patients.This prospective cohort study enrolled 117 ICH patients and 15 healthy controls. Serum MPO-DNA levels were measured via ELISA. The relationship between serum MPO-DNA levels and VTE risk was analyzed. The predictive value of MPO-DNA was evaluated by ROC curves. Effects of statin on NETs and VTE incidence were evaluated.The median MPO-DNA level in patients with VTE was 0.304 (95% CI: 0.231-0.349), significantly higher than the 0.188 (95% CI: 0.159-0.236) in non-VTE patients. Elevated MPO-DNA levels were associated with an increased VTE risk (OR 7.13, 95% CI 2.58-19.75; P < 0.001), and this association persisted after adjustment. The AUC values for MPO-DNA, CRP, and D-dimer were 0.824 (95% CI: 0.719-0.928), 0.618 (95% CI: 0.481-0.754), and 0.786 (95% CI: 0.683-0.888), respectively. Moreover, statin users exhibited reduced MPO-DNA levels (0.174 vs. 0.218; P = 0.007), though VTE incidence differences (13.8% vs. 19.3%) lacked statistical significance.Serum MPO-DNA serves as a sensitive biomarker for VTE prediction in ICH, highlighting NETs as potential therapeutic targets. Statins could attenuate NETosis, but larger trials are required to validate their clinical efficacy and safety in VTE prevention for ICH patients.
Collapse
Affiliation(s)
- Xinyan Yan
- Department of Neurology, Hunan Provincial People's Hospital and The First-Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Wenyan Huang
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital and The First-Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Yunrong Chen
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital and The First-Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
19
|
Maiocchi S, Burnham EE, Cartaya A, Lisi V, Buechler N, Pollard R, Babaki D, Bergmeier W, Pinkerton NM, Bahnson EM. Development of DNase-1 Loaded Polymeric Nanoparticles Synthesized by Inverse Flash Nanoprecipitation for Neutrophil-Mediated Drug Delivery to In Vitro Thrombi. Adv Healthc Mater 2025:e2404584. [PMID: 40341904 DOI: 10.1002/adhm.202404584] [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: 11/17/2024] [Revised: 03/31/2025] [Indexed: 05/11/2025]
Abstract
Activated neutrophils release Neutrophil Extracellular Traps (NETs), comprising decondensed chromatin, peroxidases, and serine proteases, which aid in host defense but are also implicated in thrombosis and resistance to thrombolysis. Recombinant DNase 1, which degrades NETs, may aid in thrombus dissolution synergistically with fibrinolytics. However, its short half-life and susceptibility to plasma proteases limit its therapeutic applicability. To address these limitations, DNase1 is encapsulated into polymeric nanoparticles (DNPs) using inverse Flash Nanoprecipitation (iFNP), a scalable nanoparticle synthesis technique. Previously only used with model proteins, the study demonstrates for the first time the feasibility of extending iFNP to the encapsulation of therapeutic proteins. Conditions that promote DNase1 solubility, preserve activity, and demonstrate release resulting in ex vivo NET degradation are detailed. Furthermore, the use of neutrophils, the source of NETs, as carriers for DNPs to enhance targeted delivery is investigated. These findings confirm that DNP-loaded neutrophils maintain key functionalities, including viability and oxidative burst, and associate with in vitro blood clots to deliver nanoparticles, and DNase1 protein. This study not only extends the feasibility of applying iFNP to encapsulate therapeutic proteins into polymeric nanoparticles, a promising alternative to lipid nanoparticles, but also contributes to the emerging literature on neutrophils as delivery vectors for nanocarriers.
Collapse
Affiliation(s)
- Sophie Maiocchi
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Erica E Burnham
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Ana Cartaya
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Light Microscopy Core Facility, Duke University, Durham, NC, 27710, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Veronica Lisi
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Nancy Buechler
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Rachel Pollard
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Danial Babaki
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nathalie M Pinkerton
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, NY, 11201, USA
| | - Edward M Bahnson
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Center for Nanotechnology in Drug Delivery, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
20
|
Yan X, Lu Y, Lv K, Jiang M, Fang C, Wu Y, Yang A. Endoplasmic reticulum protein 29 negatively regulates platelet functions and thrombosis in mice. Thromb J 2025; 23:44. [PMID: 40336049 PMCID: PMC12057190 DOI: 10.1186/s12959-025-00726-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Several members of protein disulfide isomerase (PDI) family with the CXYC active motif such as PDI, ERp57, ERp72, ERp46, ERp5 and TMX1 have important roles in platelet functions and thrombosis. These members contribute to the network of redox regulation of platelet activities. However, whether other PDI family members without the CXYC motif such as ERp29, have a role in these processes remains unknown. AIMS To determine the role of ERp29 in platelet functions and thrombosis. METHODS The phenotypes of platelet-specific ERp29-deficient (Pf4-Cre/ERp29fl/fl) mice were evaluated using tail bleeding assay and laser-induced and FeCl3-induced arterial injury models, as well as venous thrombosis model. In vitro, the functions of ERp29-deficient platelets were assessed in respect to aggregation, adhesion, spreading, clot retraction, granule secretion and integrin αIIbβ3 activation measured by flow cytometry. Redox state of integrin αIIbβ3 thiols was detected using 3-(N-maleimido-propionyl) biotin (MPB) labeling. RESULTS Compared with WT mice, Pf4-Cre/ERp29fl/fl mice exhibited shortened tail-bleeding times, increased platelet accumulation in the two arterial thrombosis models, and enhanced thrombogenesis in the venous thrombosis model. ERp29-deficient platelets had enhanced response in aggregation, ATP release, spreading, clot retraction, αIIbβ3 activation, fibrinogen binding and P-selectin expression. As detected by MPB labeling, the free thiol content of integrin αIIbβ3 in ERp29-deficient platelets were increased compared with WT platelets, suggesting that the role of ERp29 is associated with oxidation of the functional disulfides of integrin αIIb and/or β3 subunits. CONCLUSION(S) ERp29 is the first disulfide isomerase without the CXYC motif that negatively regulates platelet function. This study provides new insight into the redox network controlling thrombosis.
Collapse
Affiliation(s)
- Xiaofeng Yan
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yishan Lu
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Keyu Lv
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Miao Jiang
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Yi Wu
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Aizhen Yang
- Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Cyrus Tang Medical Institute, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
21
|
Kaiser R, Gold C, Stark K. Recent Advances in Immunothrombosis and Thromboinflammation. Thromb Haemost 2025. [PMID: 40311639 DOI: 10.1055/a-2523-1821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Inflammation and thrombosis are traditionally considered two separate entities of acute host responses to barrier breaks. While inciting inflammatory responses is a prerequisite to fighting invading pathogens and subsequent restoration of tissue homeostasis, thrombus formation is a crucial step of the hemostatic response to prevent blood loss following vascular injury. Though originally designed to protect the host, excessive induction of either inflammatory signaling or thrombus formation and their reciprocal activation contribute to a plethora of disorders, including cardiovascular, autoimmune, and malignant diseases. In this state-of-the-art review, we summarize recent insights into the intricate interplay of inflammation and thrombosis. We focus on the protective aspects of immunothrombosis as well as evidence of detrimental sequelae of thromboinflammation, specifically regarding recent studies that elucidate its pathophysiology beyond coronavirus disease 2019 (COVID-19). We introduce recently identified molecular aspects of key cellular players like neutrophils, monocytes, and platelets that contribute to both immunothrombosis and thromboinflammation. Further, we describe the underlying mechanisms of activation involving circulating plasma proteins and immune complexes. We then illustrate how these factors skew the inflammatory state toward detrimental thromboinflammation across cardiovascular as well as septic and autoimmune inflammatory diseases. Finally, we discuss how the advent of new technologies and the integration with clinical data have been used to investigate the mechanisms and signaling cascades underlying immunothrombosis and thromboinflammation. This review highlights open questions that will need to be addressed by the field to translate our mechanistic understanding into clinically meaningful therapeutic targeting.
Collapse
Affiliation(s)
- Rainer Kaiser
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christoph Gold
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
22
|
Liu Q, Zhu W, Wen X, Da Y. The Role of Platelet-Neutrophil Interactions in Driving Autoimmune Diseases. Immunology 2025; 175:1-15. [PMID: 39825744 DOI: 10.1111/imm.13901] [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: 09/11/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025] Open
Abstract
Platelets and neutrophils are among the most abundant cell types in peripheral blood. Beyond their traditional roles in thrombosis and haemostasis, they also play an active role in modulating immune responses. Current knowledge on the role of platelet-neutrophil interactions in the immune system has been rapidly expanding. Notably, circulating platelet-neutrophil complexes (PNCs) have been widely detected in various inflammatory diseases and infections, closely associated with inflammatory processes affecting multiple organs. These findings emphasise the critical role of platelet-neutrophil interactions in driving and sustaining inflammatory responses. In this review, we elucidate the mechanisms by which neutrophils and platelets physically interact, leading to mutual activation. Additionally, activated platelets release pro-inflammatory factors that further modulate neutrophil effector functions, enhancing their immune response capabilities. We highlight the role of platelets in promoting the formation of neutrophil extracellular traps (NETs), which, in turn, promote local platelet activation, thereby exacerbating the immune response and sustaining chronic inflammation. Furthermore, we review current evidence on the role of platelet-neutrophil interactions in common autoimmune diseases such as systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and rheumatoid arthritis (RA). Finally, we identify gaps in understanding the mechanisms of these interactions in the context of other autoimmune diseases and underscore the potential of targeting platelets and neutrophils as a therapeutic strategy for these conditions.
Collapse
Affiliation(s)
- Qinyao Liu
- Department of Neurology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Wenjia Zhu
- Department of Neurology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Xinmei Wen
- Department of Neurology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Levinson S, Pulli B, Heit JJ. Neuroinflammation and acute ischemic stroke: impact on translational research and clinical care. Front Surg 2025; 12:1501359. [PMID: 40356948 PMCID: PMC12066521 DOI: 10.3389/fsurg.2025.1501359] [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: 09/24/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Background Stroke, encompassing both ischemic and hemorrhagic subtypes, is a leading cause of mortality and disability globally and current treatments remain limited. Neuroinflammation plays a crucial role in the pathophysiology of stroke, influencing both acute injury and long-term recovery. Objective This review aims to provide a comprehensive overview of neuroinflammation in stroke, detailing the mechanisms, clinical implications, and potential therapeutic strategies. Methods A detailed literature review was conducted, focusing on recent advancements in understanding the neuroinflammatory processes in stroke, including the roles of thromboinflammation, blood-brain barrier (BBB) disruption, and the immune response. Results The initial ischemic insult triggers an inflammatory cascade involving both innate and adaptive immune responses. BBB disruption allows peripheral immune cells and neurotoxic substances to infiltrate the brain, exacerbating neuronal damage and increasing the risk of infections such as pneumonia and urinary tract infections. Thromboinflammation, characterized by platelet activation and immune cell interactions, further complicates the ischemic environment. Proteomic studies have identified key biomarkers that offer insights into neuroinflammatory mechanisms and potential therapeutic targets. Advances in imaging techniques, such as PET and MRI, enable real-time monitoring of neuroinflammation, facilitating personalized treatment approaches. Conclusion Neuroinflammation significantly impacts stroke outcomes, presenting both challenges and opportunities for treatment. Current immunologic therapeutic strategies are limited. Future research should aim to further elucidate the complex immune interactions in stroke, refine imaging biomarkers for clinical use, and develop effective interventions to mitigate neuroinflammation.
Collapse
Affiliation(s)
- Simon Levinson
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Benjamin Pulli
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jeremy J. Heit
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
24
|
Husieva SA, Osyodlo GV, Husiev AV, Savichan KV. A Clinical Case of Timely Diagnosis and Successful Treatment of Budd-Chiari Syndrome With Fulminant Cytolysis in the Setting of a First-time Diagnosed Myeloproliferative Disease. Mil Med 2025; 190:1292-1299. [PMID: 38935394 DOI: 10.1093/milmed/usae331] [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: 02/14/2024] [Revised: 05/03/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
The article presents a clinical case of peculiarities of clinical manifestations, diagnostic and therapeutic approaches of undiagnosed chronic myeloproliferative disease, on the background of which Budd-Chiari syndrome (BCS) developed. The results of clinical course, examination, and treatment of a patient with BCS as a manifestation of the hidden course of primary myelofibrosis with the presence of somatic mutation (V617F) in Janus-tyrosine kinase-2 (JAK2) gene in myeloid cells are presented. Standard clinical and laboratory examinations, and cytomorphologic and histologic examination of bone marrow were used. The diagnosis of BCS was confirmed by ultrasound (US) Doppler examination of the portal system vessels. Symptomatic therapy resulted in insignificant positive results. The analysis of this clinical case showed that the development of BCS was due to a chronic myeloproliferative disease that was not diagnosed before the development of thrombosis. Hepatic vein thrombosis was accompanied by the development of fulminant cytolytic syndrome. Along with symptomatic therapy, patient K., female, 32 years old, underwent transjugular intrahepatic portosystemic shunting 1 month after the first symptoms of BCS appeared, which contributed to a significant clinical effect. Seven years after the installation of 4 transjugular intrahepatic portosystemic shunts, the patient's condition remains satisfactory. The uniqueness of this clinical case lies in the presence of 2 serious diseases at the same time: myeloproliferative pathology (primary myelofibrosis) JAK2-positive variant and BCS. Timely diagnosis of both hematological diseases and their complication in the form of hepatic vein thrombosis with fulminant cytolytic syndrome allowed timely prescription of adequate treatment with a good clinical response.
Collapse
Affiliation(s)
- Svitlana A Husieva
- Military Therapy Department, Ukrainian Military Medical Academy, Kyiv 01015, Ukraine
| | - Galyna V Osyodlo
- Military Therapy Department, Ukrainian Military Medical Academy, Kyiv 01015, Ukraine
| | - Andrii V Husiev
- Department of Liver Surgery and Transplantation, Shalimov National Scientific Center of Surgery and Transplantation of the National Academy of Medical Sciences of Ukraine, Kyiv 03680, Ukraine
| | - Kyrylo V Savichan
- Military Therapy Department, Ukrainian Military Medical Academy, Kyiv 01015, Ukraine
| |
Collapse
|
25
|
Morrissey SM, Kirkland LG, Phillips TK, Levit RD, Hopke A, Jensen BC. Multifaceted roles of neutrophils in cardiac disease. J Leukoc Biol 2025; 117:qiaf017. [PMID: 39936506 DOI: 10.1093/jleuko/qiaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/15/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025] Open
Abstract
Neutrophils, the most abundant leukocytes in human blood, have long been recognized as critical first responders in the innate immune system's defense against pathogens. Some of the more notable innate antimicrobial properties of neutrophils include generation of superoxide free radicals like myeloperoxidase, production of proteases that reshape the extracellular matrix allowing for easier access to infected tissues, and release of neutrophil extracellular traps, extruded pieces of DNA that ensnare bacterial and fungi. These mechanisms developed to provide neutrophils with a vast array of specialized functions to provide the host defense against infection in an acute setting. However, emerging evidence over the past few decades has revealed a far more complex and nuanced role for these neutrophil-driven processes in various chronic conditions, particularly in cardiovascular diseases. The pathophysiology of cardiac diseases involves a complex interplay of hemodynamic, neurohumoral, and inflammatory factors. Neutrophils, as key mediators of inflammation, contribute significantly to this intricate network. Their involvement extends far beyond their classical role in pathogen clearance, encompassing diverse functions that can both exacerbate tissue damage and contribute to repair processes. Here, we consider the contributions of neutrophils to myocardial infarction, heart failure, cardiac arrhythmias, and nonischemic cardiomyopathies. Understanding these complex interactions is crucial for developing novel therapeutic strategies aimed at modulating neutrophil functions in these highly morbid cardiac diseases.
Collapse
Affiliation(s)
- Samantha M Morrissey
- Department of Medicine, University of North Carolina School of Medicine, 125 MacNider Hall, Chapel Hill, NC 27599-7005, United States
| | - Logan G Kirkland
- McAllister Heart Institute, University of North Carolina School of Medicine, 111 Mason Farm Rd., Chapel Hill, NC 27599-7126, United States
| | - Tasha K Phillips
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN 37614, United States
| | - Rebecca D Levit
- Division of Cardiology, Department of Medicine, Emory University, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Alex Hopke
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN 37614, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, PO Box 70300, Johnson City, TN 37614, United States
| | - Brian C Jensen
- Department of Medicine, University of North Carolina School of Medicine, 125 MacNider Hall, Chapel Hill, NC 27599-7005, United States
- McAllister Heart Institute, University of North Carolina School of Medicine, 111 Mason Farm Rd., Chapel Hill, NC 27599-7126, United States
- Department of Pharmacology, University of North Carolina School of Medicine, 120 Mason Farm Rd., Chapel Hill, NC 27599-7365, United States
| |
Collapse
|
26
|
Feng Y, Zhao W, Fang S, Zhao J, Wang W, Zhou S, Wang T, Fang X, Chen X, Awais M, Cai C, Shen C, Liu M. Low-Molecular-Weight Fucoidan Inhibits Thromboinflammation and Ameliorates Deep Vein Thrombosis via Targeting S100A8/A9. Mar Drugs 2025; 23:180. [PMID: 40422770 DOI: 10.3390/md23050180] [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: 03/25/2025] [Revised: 04/19/2025] [Accepted: 04/20/2025] [Indexed: 05/28/2025] Open
Abstract
Deep vein thrombosis (DVT) is a prevalent life-threatening complication among hospitalized patients. DVT is characterized by the hypercoagulability and thromboinflammation in which platelet activation and neutrophil extracellular trap (NET) formation are critically involved. Studies have shown that S100A8/A9 is significantly elevated in patients with DVT, and is closely associated with platelet activation and NET formation. Fucoidan, the marine polysaccharide derived from Fucus algae, has potential anti-inflammatory and cardioprotective effects. We found low-molecular-weight fucoidan (LMF) bound to S100A8/A9 with an equilibrium dissociation constant (KD) of 2.368 × 10-8 M. LMF inhibited S100A8/A9-induced platelet hyperactivity and NET formation in vitro, and ameliorated DVT without significantly perturbing hemostasis in vivo. Our results indicate that the alarmin protein S100A8/A9 is a novel target of LMF. LMF may have therapeutic potential in S100A8/A9-induced thromboinflammation in DVT.
Collapse
Affiliation(s)
- Yiting Feng
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Weiqing Zhao
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Siwen Fang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Jingwen Zhao
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Wanshuai Wang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Shaoyun Zhou
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Tianyu Wang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Xinke Fang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Xue Chen
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Muhammad Awais
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| | - Chao Cai
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Qingdao 266000, China
| | - Chuanbin Shen
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266071, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Qingdao 266000, China
| | - Ming Liu
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Chinese Ministry of Education, Qingdao 266000, China
| |
Collapse
|
27
|
Han L, Pan TW, Yang LL, Qian WY, Xu XP, Wang F, Wang WZ, Liu Y, Yang WY. Nomogram for Deep Vein Thrombosis Prediction Post-Endovascular Thrombectomy in Acute Ischemic Stroke: A Retrospective Multicenter Observational Study. J Clin Nurs 2025. [PMID: 40251701 DOI: 10.1111/jocn.17786] [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: 06/27/2024] [Revised: 10/20/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Deep vein thrombosis (DVT) is a frequent complication following endovascular thrombectomy (EVT) in patients with acute ischaemic stroke (AIS), potentially leading to fatal pulmonary embolism (PE). Identifying patients early at high risk for DVT is clinically important. This study developed and validated a nomogram combining laboratory findings and clinical characteristics to predict the risk of lower-extremity DVT after EVT in patients with AIS. METHODS This retrospective multicentre observational study was conducted in two tertiary hospitals in China, enrolling 640 patients who underwent ultrasonography for DVT diagnosis within 10 days following EVT. Data on medical history, examination and laboratory results were collected for logistic regression analyses to develop a DVT risk nomogram. RESULTS Logistic regression analyses identified critical predictors of DVT: lower limb National Institutes of Health Stroke Scale (NIHSS) score ≥ 2, elevated D-dimer levels (≥ 1.62 mg/L) and prolonged puncture-to-recanalization time (PRT ≥ 66 min). The nomogram demonstrated good discriminative ability (AUC 0.741-0.822) and clinical utility across internal and external validation cohorts. Additionally, the presence of DVT was significantly associated with reduced functional independence at 90 days post-EVT, highlighting the negative impact of DVT on patient recovery (OR = 3.85; 95% CI: 2.18-6.78; p < 0.001). CONCLUSION The study provides a practical clinical tool for early detection and intervention in patients with AIS at high risk for DVT following EVT. Early identification and intervention may help improve outcomes in patients with AIS undergoing EVT. RELEVANCE TO CLINICAL PRACTICE This nomogram helps in the early detection and proactive management of DVT in AIS patients, which can reduce severe complications and improve patient recovery outcomes. PATIENT OR PUBLIC CONTRIBUTION No patient or public contributions were involved in this study due to its retrospective design, where data were utilised from existing medical records without direct patient interaction.
Collapse
Affiliation(s)
- Li Han
- Department of Neurology, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Teng-Wei Pan
- Department of Neurology, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Li-Li Yang
- Department of Neurology, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Wei-Yang Qian
- Department of Neurosurgery, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Xiao-Ping Xu
- Department of Neurology, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Feng Wang
- Department of Neurology, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Wei-Zhen Wang
- Department of Nursing, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yang Liu
- Department of Neurology, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Department of Neurology, Saarland University, Homburg, Germany
| | - Wei-Ying Yang
- Department of Nursing, Taizhou Hospital of Zhejiang Province, Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
28
|
Yuan Z, Luozhong S, Li R, Gu W, Chen Y, Bhashyam D, Lai R, Jiang S. Transient Macrophage Depletion Circumvents Scavenging and Redirects Biodistribution of mRNA-Lipid Nanoparticles. ACS NANO 2025; 19:14422-14433. [PMID: 40167042 DOI: 10.1021/acsnano.5c02001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The mononuclear phagocytic system is recognized as a major scavenger of mRNA-lipid nanoparticles (LNPs), clearing and redirecting these particles away from their intended targets and thus diminishing their delivery efficacy. Understanding the mechanism by which mRNA-LNPs interact with phagocytes and how this interaction affects the mRNA transfection is critical to enhancing the delivery of mRNA. In this study, we temporarily depleted both circulating and resident macrophages (MF) and evaluated the transfection efficiency and biodistribution of mRNA-LNPs. We first demonstrated the enhanced liver expression using two liver-tropic formulations and the significant improvement of the in vivo gene editing efficiency of CRISPR-Cas9 in the Ai14 mouse model after MF depletion, providing a versatile strategy for enhanced mRNA delivery to the liver regardless of the formulation employed. We then extended our investigations to lung-tropic and lymphoid-tropic LNP formulations and discovered that MF depletion abolishes the targeting capacities of these non-liver-tropic formulations, providing insights into the organ targeting of LNPs. Finally, we screened and compared various clinically relevant MF depletion methods, providing the translation potential of this method on enhanced hepatic delivery of mRNA.
Collapse
Affiliation(s)
- Zhefan Yuan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Sijin Luozhong
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Ruoxin Li
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Wenchao Gu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Yu Chen
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Dani Bhashyam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Rachel Lai
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Shaoyi Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
29
|
Gagliano-Jucá T, Pencina KM, Shang YV, Travison TG, Lincoff AM, Nissen SE, Artz AS, Li X, Chan A, Patel R, Miller MG, Bhasin S. Association of testosterone-induced increase in neutrophil and monocyte counts with thromboembolic events: The TRAVERSE trial. Am Heart J 2025; 288:77-88. [PMID: 40246046 DOI: 10.1016/j.ahj.2025.04.004] [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] [Received: 01/15/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND In epidemiological studies, higher leukocyte and platelet counts are associated with increased risk of cardiovascular events. Effects of testosterone replacement therapy (TRT) on leukocyte subsets and platelets in men with hypogonadism and association of circulating leukocyte subtypes and platelets during TRT with cardiovascular events remain unknown. METHODS In the TRAVERSE Trial, 5,204 men, 45-80 years with hypogonadism and preexisting or increased risk of cardiovascular disease (CVD) were randomized to transdermal testosterone or placebo gel daily for up to 5 years. We determined the effect of TRT on neutrophils, monocytes, lymphocytes and platelets and association of changes in leukocyte subtypes and platelets with risk of major adverse cardiovascular (MACE) and venous thromboembolism (VTE) events. RESULTS TRT was associated with significantly greater increase in neutrophils and monocytes, and greater decrease in lymphocytes and platelets than placebo. Changes in neutrophil (odds ratio for 1 SD increase in cell count (OR) 1.32 [1.01, 1.73]) and monocyte (OR 1.39 [1.08, 1.79]) counts were associated with increased risk of VTE, accounting for TRT. Neutrophil and monocyte counts at baseline and on-treatment were also associated with increased risk of MACE, adjusting for treatment (baseline: neutrophils OR 1.18 [1.06,1.31], monocytes OR 1.16 [1.05,1.29]; on-treatment neutrophils: OR 1.25 [1.12, 1.40]; monocytes: OR 1.18 [1.06,1.31]). CONCLUSIONS TRT increased circulating neutrophils and monocytes and decreased lymphocytes and platelets in men with hypogonadism. Changes in monocyte and neutrophil counts were associated with increased risk of VTE. Neutrophil and monocyte counts should be considered when evaluating VTE risk in hypogonadal men treated with TRT. CLINICAL TRIAL REGISTRATION URL:https://clinicaltrials.gov/study/NCT03518034. Unique identifier: NCT03518034. The study was initially registered on March 5, 2018, and the first participant was enrolled on May 23, 2018.
Collapse
Affiliation(s)
- Thiago Gagliano-Jucá
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | - Karol M Pencina
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Yili V Shang
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - A Michael Lincoff
- Cleveland Clinic Coordinating Center for Clinical Research, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Steven E Nissen
- Cleveland Clinic Coordinating Center for Clinical Research, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Andrew S Artz
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | - Xue Li
- AbbVie, Inc, North Chicago, IL
| | | | | | | | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
30
|
Imiela AM, Kucharska J, Kukliński F, Fernandez Moreno T, Dzik K, Pruszczyk P. Advanced Research in the Pathophysiology of Venous Thromboembolism-Acute Pulmonary Embolism. Biomedicines 2025; 13:906. [PMID: 40299499 PMCID: PMC12025274 DOI: 10.3390/biomedicines13040906] [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: 03/03/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
According to the literature, cardiovascular diseases (CVDs)-including myocardial infarction, stroke, and venous thromboembolism (VTE)-are among the leading causes of mortality and morbidity worldwide. Evidence suggests that CVDs share common risk factors and pathophysiological mechanisms. Similar to the Mosaic Theory of Hypertension proposed by Irvine Page in 1949, the pathophysiology of VTE is multifactorial, involving multiple interacting processes. The concept of immunothrombosis, introduced by Engelmann and Massberg in 2009, describes the interplay between the immune system and thrombosis. Both thrombosis and hemostasis share core mechanisms, including platelet activation and fibrin formation. Additionally, immune mediators-such as monocytes, neutrophil extracellular traps (NETs), lymphocytes, selectins, and various molecular factors-play a critical role in thrombus formation. This review highlights inflammation as a key risk factor for pulmonary embolism (APE). Immunity is central to the complex interactions among the coagulation cascade, platelets, endothelium, reactive oxygen species (ROS), and genetic factors. Specifically, we examine the roles of the endothelium, immune cells, and microRNAs (miRNAs) in the pathophysiology of APE and explore potential therapeutic targets. This review aims to elucidate the roles of the endothelium, immune cells, and miRNAs in the pathophysiology of APE and explore potential future perspective.
Collapse
Affiliation(s)
- Anna M. Imiela
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| | - Joanna Kucharska
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| | - Franciszek Kukliński
- Department of Intensive Cardiac Care, Medical University of Bialystok, 15-089 Białystok, Poland
| | - Teresa Fernandez Moreno
- Department of Intensive Cardiac Care, Medical University of Bialystok, 15-089 Białystok, Poland
| | - Konrad Dzik
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| | - Piotr Pruszczyk
- Department of Internal Diseases and Cardiology, Infant Jesus Clinical Hospital, Medical University of Warsaw, Lindleya 4 Street, 02-005 Warsaw, Poland
| |
Collapse
|
31
|
Liu J, He M, Song R, Li J. The association between deep vein thrombosis at admission and the time from injury to admission in hip fractures. BMC Geriatr 2025; 25:222. [PMID: 40186130 PMCID: PMC11971877 DOI: 10.1186/s12877-025-05875-z] [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: 10/22/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This study aims to explore the association between deep vein thrombosis (DVT) of the lower limbs at admission and the time from injury to admission (TFITA), providing clinical references for the prevention of DVT at admission. PATIENTS AND METHODS Data was collected from patients who were admitted to our hospital for hip fractures between January 2017 and December 2023. Univariable and multivariable logistic regression analyses were conducted to examine the relationship between TFITA and DVT at admission, using both continuous and categorized variables based on thresholds for TFITA. Propensity score matching (PSM) and subanalyses stratified by TFITA and characteristics of DVT at admission were further employed to investigate the relationship. Additionally, restricted cubic splines (RCS) analysis was performed to determine whether a non-linear association exists between TFITA and DVT at admission. RESULTS A total of 1230 patients were included in the statistical analysis, comprising 116 patients with DVT at admission and 1114 without. Both Univariable and multivariable logistic regression analyses indicated a positive association between TFITA and DVT at admission before and after matching. Subanalyses revealed significant associations for older age, low-energy injuries, high D-dimer levels, and low platelet counts subgroup with TFITA and DVT at admission. RCS analysis indicated no non-linear relationship between TFITA and DVT at admission. CONCLUSION For patients with hip fractures, longer TFITA is positively correlated with the incidence of DVT at admission. These findings support the potential of TFITA as an intervention strategy for managing DVT at admission.
Collapse
Affiliation(s)
- Jian Liu
- Department of Orthopaedic Surgery, Chongqing Emergency Medical Center (Chongqing University Central Hospital), No. 1 Jiankang Road, Chongqing, 400010, China
| | - Miao He
- Department of Orthopaedic Surgery, Chongqing Emergency Medical Center (Chongqing University Central Hospital), No. 1 Jiankang Road, Chongqing, 400010, China
| | - Ruoyu Song
- Department of Orthopaedic Surgery, Chongqing Emergency Medical Center (Chongqing University Central Hospital), No. 1 Jiankang Road, Chongqing, 400010, China
| | - Jie Li
- Department of Orthopaedic Surgery, Chongqing Emergency Medical Center (Chongqing University Central Hospital), No. 1 Jiankang Road, Chongqing, 400010, China.
| |
Collapse
|
32
|
Zhang Y, Wang R. A mendelian randomization study on the association between 731 types of immune cells and 91 types of blood cells with venous thromboembolism. Thromb J 2025; 23:28. [PMID: 40181342 PMCID: PMC11967152 DOI: 10.1186/s12959-025-00714-y] [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: 01/14/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Venous thromboembolism (VTE) is a grave medical condition characterized by the blockage of distant blood vessels due to blood clots or detached vessel wall fragments, leading to ischemia or necrosis of the affected tissues. With the recent introduction of immunothrombosis, the significance of immune cells in the process of thrombus formation has gained prominent attention. Complex cross-talk occurs between immune cells and blood cells during infection or inflammation, with immune cells actively participating in blood clot formation by promoting platelet recruitment and thrombin activation. Nevertheless, comprehensive studies on the genetic association between immune cells phenotypes and VTE remain scarce. This article employed Mendelian randomization (MR) to investigate the association between the incidence of VTE and a range of 731 immune cell types, along with 91 blood cell perturbation phenotypes, utilizing single nucleotide polymorphisms as instrumental variables. METHODS Through the utilization of publicly available genetic data, a two-sample bi-directional MR analysis was conducted. Sensitivity analyses included Cochran's Q test, MR-Egger intercept test, MR-pleiotropy residual sum and outlier (MR-PRESSO) and leave-one-out analysis. For significant associations, replication analysis was conducted using GWAS data from deep vein thrombosis (DVT) and pulmonary embolism (PE). RESULTS We firstly investigated the causal relationship between 731 immune cells and VTE risk. All the GWAS data were obtained from European populations and from men and women. The IVW analysis revealed that CD20 on naive-mature B cell, CD20 on IgD- CD38dim B cell, and CD20 on unswitched memory B cell may increase the risk of VTE (P < 0.05). CD28- CD8dim T cell %T cell, CD64 on monocyte and CD64 on CD14 + CD16- monocyte may be protective factors against DVT (P < 0.05). Then disturbed blood cells types as exposure were analyzed to examine its association with occurrence of VTE. Initial and replication analysis both revealed that environmental KCl-impacted red blood cells and butyric acid-impacted platelet accelerated incidence of VTE (P < 0.05), while colchicine -impacted eosinophil, KCl-impacted reticulocyte and Lipopolysaccharide (LPS) -impacted neutrophil reduced VTE risk (P < 0.05). Sensitivity analyses confirmed the robustness and reliability of these positive findings. CONCLUSIONS Our study presents evidence of a causal link between six immune cell phenotypes and VTE. Additionally, we have identified two types of blood cells that are associated with both VTE and DVT, and three types of blood cells that are relevant to both VTE and PE. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Rui Wang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
33
|
Knauss EA, Guci J, Luc N, Disharoon D, Huang GH, Gupta AS, Nieman MT. Mice with reduced protease-activated receptor 4 reactivity show decreased venous thrombosis and platelet procoagulant activity. J Thromb Haemost 2025; 23:1278-1288. [PMID: 39798922 PMCID: PMC11992619 DOI: 10.1016/j.jtha.2024.12.031] [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: 10/14/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Hypercoagulation and thrombin generation are major risk factors for venous thrombosis. Sustained thrombin signaling through protease-activated receptor (PAR) 4 promotes platelet activation, phosphatidylserine exposure, and subsequent thrombin generation. A single nucleotide polymorphism in PAR4 (rs2227376) changes proline to leucine extracellular loop 3, which decreases PAR4 reactivity and is associated with a lower risk for venous thromboembolism (VTE) in a genome wide association studies meta-analysis. OBJECTIVES The goal of this study was to determine the mechanism for the association of rs2227376 with a reduced risk of VTE using mice with a homologous mutation (PAR4-P322L). METHODS Venous thrombosis was examined using our recently generated PAR4-P322L mice in the inferior vena cava stasis and stenosis models. Coagulation and clot stability were measured using rotational thromboelastometry. Thrombin-generating potential was measured in platelet-rich plasma. Phosphatidylserine surface expression and platelet-neutrophil aggregates were analyzed using flow cytometry. RESULTS Mice heterozygous (PAR4P/L) or homozygous (PAR4L/L) at position 310 had reduced sizes of venous clots at 48 hours. PAR4P/L and PAR4L/L platelets had progressively decreased phosphatidylserine in response to thrombin and convulxin, in addition to decreased thrombin generation and decreased PAR4-mediated platelet-neutrophil aggregation. CONCLUSION The leucine allele in extracellular loop 3, PAR4-322L, leads to fewer procoagulant platelets, decreased endogenous thrombin potential, and reduced platelet-neutrophil aggregation. This decreased ability to generate thrombin and bind to neutrophils offers a mechanism for PAR4's role in VTE, highlighting a key role for PAR4 signaling.
Collapse
Affiliation(s)
- Elizabeth A Knauss
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Johana Guci
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Norman Luc
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dante Disharoon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Grace H Huang
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Marvin T Nieman
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
34
|
Masset C, Drillaud N, Ternisien C, Degauque N, Gerard N, Bruneau S, Branchereau J, Blancho G, Mesnard B, Brouard S, Giral M, Cantarovich D, Dantal J. The concept of immunothrombosis in pancreas transplantation. Am J Transplant 2025; 25:650-668. [PMID: 39709128 DOI: 10.1016/j.ajt.2024.11.025] [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: 08/09/2024] [Revised: 11/06/2024] [Accepted: 11/23/2024] [Indexed: 12/23/2024]
Abstract
Early failure of a pancreatic allograft due to complete thrombosis has an incidence of approximately 10% and is the main cause of comorbidity in pancreas transplantation. Although several risk factors have been identified, the exact mechanisms leading to this serious complication are still unclear. In this review, we define the roles of the individual components involved during sterile immunothrombosis-namely endothelial cells, platelets, and innate immune cells. Further, we review the published evidence linking the main risk factors for pancreatic thrombosis to cellular activation and vascular modifications. We also explore the unique features of the pancreas itself: the vessel endothelium, specific vascularization, and relationship to other organs-notably the spleen and adipose tissue. Finally, we summarize the therapeutic possibilities for the prevention of pancreatic thrombosis depending on the different mechanisms such as anticoagulation, anti-inflammatory molecules, endothelium protectors, antagonism of damage-associated molecular patterns, and use of machine perfusion.
Collapse
Affiliation(s)
- Christophe Masset
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France.
| | - Nicolas Drillaud
- Laboratory of Hemostasis, Nantes University Hospital, Nantes, France
| | | | - Nicolas Degauque
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Nathalie Gerard
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Sarah Bruneau
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Julien Branchereau
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Gilles Blancho
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Benoit Mesnard
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Sophie Brouard
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Magali Giral
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Diego Cantarovich
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jacques Dantal
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France; Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
35
|
Zhang B, Yao Z, Li P, Niu G, Yan Z, She K, Cheng G, Yang M. Causal Relationships of Circulating Inflammatory Proteins and Portal Vein Thrombosis: A Mendelian Randomization Study. Semin Thromb Hemost 2025; 51:272-278. [PMID: 39293485 DOI: 10.1055/s-0044-1790259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Portal vein thrombosis (PVT) is commonly encountered in patients with cirrhosis, challenging our understanding of its development, particularly the ambiguous contribution of inflammation. This study utilized Mendelian randomization (MR) to explore the causal impact of circulating inflammatory markers on PVT.Employing a two-sample MR framework, we merged genome-wide association study (GWAS) meta-analysis findings of 91 inflammation-associated proteins with independent PVT data from the FinnGen consortium's R10 release. A replication analysis was performed using a distinct GWAS dataset from the UK Biobank. Inverse variance weighting, MR-Egger regression, weighted median estimator, and Mendelian Randomization Pleiotropy RESidual Sum and Outlier were used for analysis, supplemented by multivariable MR (MVMR) to adjust for cirrhosis effects.Findings indicate a significant inverse association between the genetically inferred concentration of eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and PVT risk, evidenced by an odds ratio (OR) of 0.37 (95% confidence interval [CI]: 0.21-0.67; p = 9.2 × 10-4; adjusted for multiple testing p = 0.084). This association was corroborated in the replication phase (OR = 0.39, 95% CI: 0.17-0.93; p = 0.03) and through MVMR analysis (OR = 0.34, 95% CI: 0.15-0.79; p = 0.012). Sensitivity analyses disclosed no evidence of heterogeneity or pleiotropy.Our investigation emphasizes the 4E-BP1 as a protective factor against PVT, underscoring its potential relevance in understanding PVT pathogenesis and its implications for diagnosis and therapy.
Collapse
Affiliation(s)
- Bihui Zhang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Ziping Yao
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Pengyu Li
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Guochen Niu
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Ziguang Yan
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Kang She
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Gong Cheng
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Min Yang
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
36
|
Varjú I, Tanka-Salamon A, Kolev K. Neutrophil Extracellular Traps: At the Interface of Thrombosis and Comorbidities. Semin Thromb Hemost 2025. [PMID: 40020757 DOI: 10.1055/a-2548-0805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
Since their discovery in 2004, neutrophil extracellular traps (NETs) have been at the center of multidisciplinary attention. Although a key tool in neutrophil-mediated immunity, these filamentous, enzyme-enriched DNA-histone complexes can be detrimental to tissues and have been identified as an underlying factor in a range of pathological conditions. Building on more than 20 years of research into NETs, this review places thrombosis, the pathological formation of blood clots, in the spotlight. From this point of view, we discuss the structure and formation of NETs, as well as the interaction of their components with the hemostatic system, dissecting the pathways through which NETs exert their marked effect on formation and the dissolution of thrombi. We pay distinct attention to the latest developments in the research of a key player in NET formation, peptidyl-arginine-deiminase (PAD) enzymes: their types, sources, and potential cross-play with the hemostatic machinery. Besides these molecular details, we elaborate on the link between pathological thrombosis, NETs, and widespread conditions that represent a debilitating public health burden worldwide, such as sepsis and neoplasms. Finally, future implications on the treatment of thrombosis-related conditions will be discussed.
Collapse
Affiliation(s)
- Imre Varjú
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Anna Tanka-Salamon
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Krasimir Kolev
- Department of Biochemistry, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
37
|
Li Y, Ren S, Zhou S. Advances in sepsis research: Insights into signaling pathways, organ failure, and emerging intervention strategies. Exp Mol Pathol 2025; 142:104963. [PMID: 40139086 DOI: 10.1016/j.yexmp.2025.104963] [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: 12/30/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Sepsis is a complex syndrome resulting from an aberrant host response to infection. A hallmark of sepsis is the failure of the immune system to restore balance, characterized by hyperinflammation or immunosuppression. However, the net effect of immune system imbalance and the clinical manifestations are highly heterogeneous among patients. In recent years, research interest has shifted from focusing on the pathogenicity of microorganisms to the molecular mechanisms of host responses which is also associated with biomarkers that can help early diagnose sepsis and guide treatment decisions. Despite significant advancements in medical science, sepsis remains a major challenge in healthcare, contributing to substantial morbidity and mortality worldwide. Further research is needed to improve our understanding of this condition and develop novel therapies to improve outcomes for patients with sepsis. This review explores the related signal pathways of sepsis and underscores recent advancements in understanding its mechanisms. Exploration of diverse biomarkers and the emerging concept of sepsis endotypes offer promising avenues for precision therapy in the future.
Collapse
Affiliation(s)
- Yehua Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China.
| | - Siying Ren
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Shen'ao Zhou
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, CAS. Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
38
|
Li H, Xing J, Song Z, Fan Z, Wen H, Liang S, Yan Q, Feng H, Han S, Yang N, Wang P, Zhang K. Effect of mild-to-moderate COVID‑19 on the incidence and risk factors for deep vein thrombosis in patients with hip fracture: a retrospective study. BMC Surg 2025; 25:113. [PMID: 40128691 PMCID: PMC11931773 DOI: 10.1186/s12893-025-02831-x] [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: 01/11/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
PURPOSE This retrospective study aimed to investigate the effect of mild-to-moderate COVID-19 on the risk of deep vein thrombosis (DVT) in patients with hip fractures. Hip fractures are common in the elderly, and previous research has shown that they accounted for 58.3% of traumatic fractures in older inpatients during the COVID-19 pandemic in China. Meanwhile, the relationship between COVID-19 and DVT is complex. Some studies have reported that the incidence of DVT in critically ill COVID-19 patients can be as high as 46%, and 20% in those with moderate-to-severe cases. However, the impact of mild-to-moderate COVID-19 on DVT risk in hip fracture patients remains unclear. METHODS Adult patients who underwent surgery for hip fractures between December 8, 2022, and January 9, 2023, were included in the study. All patients were tested for SARS-CoV-2 nucleic acid and were assessed for DVT preoperatively using doppler ultrasonography (DUS). Logistic regression was used to identify risk factors for DVT. RESULTS The records of 98 patients with hip fractures, were included in the analysis, of whom 63 were SARS-CoV-2 positive and 35 were SARS-CoV-2 negative. Pre-operative DUS showed that 36/98 patients (37%) had DVT, including 25/63 (40%) patients with COVID-19, and 11/35 (31%) patients without COVID-19. Multivariable logistic regression analysis showed that pre-operative leukocyte count and platelet-to-lymphocyte ratio (PLR) were independent risk factors for DVT, whereas mild-to-moderate COVID-19 was not an independent risk factor for DVT. In patients with hip fractures, COVID-19 did not significantly increase the risk of DVT. CONCLUSIONS Therefore, in patients with hip fractures, DVT prevention measures should be implemented routinely, regardless of COVID-19 status.
Collapse
Affiliation(s)
- Haoran Li
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jian Xing
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhe Song
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhiqiang Fan
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hongquan Wen
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shaobo Liang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qiang Yan
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haoxuan Feng
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shuang Han
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Na Yang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Wang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Kun Zhang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
39
|
Chauhan W, Ferdowsi S, Sudharshan SJ, Zennadi R. Rpl13a snoRNAs-regulated NADPH oxidase 1-dependent ROS generation: A novel RBC pathway mediating complement C3a deposition and triggering thrombosis in aging and venous blood clotting disorders. Free Radic Biol Med 2025; 230:138-150. [PMID: 39938620 PMCID: PMC11936428 DOI: 10.1016/j.freeradbiomed.2025.02.008] [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: 10/25/2024] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
Adults older than 45 years old are at higher risk of developing venous blood clotting known as venous thrombosis/thromboembolism than a cohort <45 years old. Complement activation, which can be mediated by oxidative stress, plays a central role in venous thrombosis. Yet, whether RBCs contribute to complement activation triggering thrombosis in aging and in patients with venous thrombosis/thromboembolism remains an open question. RBCs from healthy Mid-life stage (55-68 years old) adults and patients with venous thrombosis/thromboembolism showed higher deposition of the complement C3 and the anaphylatoxin C3a, and NADPH oxidase (Nox)1 expression than a younger cohort (21-30 years old). Increased C3/C3a deposition on RBCs from mid-life stage adults and patients with venous thrombosis/thromboembolism triggered prothrombin activation via Nox1-dependent reactive oxygen species (ROS) generation, and G protein-coupled receptor kinase 2 (GRK2) activation. Interaction of C3/C3a positive RBCs from mid-life stage adults with endothelial cells led to increased endothelial ROS production. TGF-β1-stimulated GRK2 and Nox1 activation in RBCs from the younger and older adults exacerbated RBC C3/C3a deposition and C3/C3a-mediated prothrombotic activation, which appears to result from ROS-mediated increased RBC phosphatidylserine exposure. Using human RBCs, and Rpl13a snoRNA knockout aged mice, we show that Rpl13a snoRNAs, the master regulators of ROS levels and oxidative stress response, regulate human and murine RBC C3a deposition and prothrombic activation in aging by modulating Nox1 mRNA expression. In vivo Rpl13a snoRNA knockout in aged mice decreased thrombi size by blunting RBC C3a deposition, and RBCs-triggering prothrombin activation. These findings point out to a novel role of RBC Rpl13a snoRNAs in dysregulating RBC ROS-induced C3a deposition promoting venous thrombosis in aging.
Collapse
Affiliation(s)
- Waseem Chauhan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Shirin Ferdowsi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - S J Sudharshan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Rahima Zennadi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
40
|
Zhang W, Pei B, Zhou Y, Li H, Ma W, Zhou B, Zhou C, Jiang H, Ji X. Emerging Targets, Novel Directions, and Innovative Approaches in Thrombosis Therapy. Aging Dis 2025:AD.2024.1688. [PMID: 40153578 DOI: 10.14336/ad.2024.1688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/11/2025] [Indexed: 03/30/2025] Open
Abstract
In clinical practice, antiplatelet, anticoagulant and fibrinolytic drugs are the mainstay of thrombosis treatment, but their potential bleeding side effects limit their widespread use. Therefore, modifying these existing drugs or developing new therapies that mitigate bleeding risks while maintaining their efficacy and utilization is necessary. Since the critical role of platelets in thrombosis is closely related to their cell surface receptors, intracellular signaling pathways and metabolism, current research focuses on these three major classes of platelet targets to develop new antithrombotic drugs. The coagulation cascade has always been the main target of anticoagulant drugs, but since the role of molecules of the contact system is more critical in thrombosis than in hemostasis, molecules targeting the contact system, such as FXIa and FXIIa, have become the main direction of anticoagulant drug research at present. Moreover, since the inflammatory response has been found to be significantly associated with thrombosis in recent years, the development of drugs that target inflammatory pathways, such as inflammasome, has also become a hot topic. This article provides a detailed description of these targets or drug formulations that are currently being investigated, including their mode of action and antithrombotic efficiency, and also points out their existing shortcomings. Moreover, antithrombotic nanomedicines can achieve precise release of drugs, which can greatly improve the thrombolytic efficiency and reduce side effects. In conclusion, this review focuses on summarizing the current new targets and new methods of antithrombotic drug research, hoping to provide a little reference for future related research.
Collapse
Affiliation(s)
- Weiyue Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Baoqing Pei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Hui Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| |
Collapse
|
41
|
Yang L, Guo R, Liu H, Chen B, Li C, Liu R, Liao S, Xie Q, Yin G. Mechanism of antiphospholipid antibody-mediated thrombosis in antiphospholipid syndrome. Front Immunol 2025; 16:1527554. [PMID: 40181965 PMCID: PMC11966034 DOI: 10.3389/fimmu.2025.1527554] [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: 11/13/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by the occurrence of thrombotic or obstetrical events in patients with persistent antiphospholipid antibodies (aPL). Thrombotic events, the primary pathological hallmarks and clinical manifestations, are among the leading causes of mortality in APS. Our understanding of the mechanism underlying APS-related thrombosis has significantly advanced in recent years. The presence of aPL, particularly anti-β2-glycoprotein I (anti-β2GPI) antibodies, is a major driver of thrombosis. The proposed pathophysiological mechanisms of aPL-mediated pro-thrombotic events can be broadly categorized into three types: disruption of anticoagulant reactions and fibrinolysis, interference with coagulation cascade cells, and complement activation. A triggering 'second hit' is typically necessary to initiate thrombosis. The development of animal models of APS has further refined our understanding of the role of aPL in thrombosis. In this review, we focused on the role of β2GPI-dependent aPL in thrombosis of thrombotic APS.
Collapse
Affiliation(s)
- Leiyi Yang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruibing Guo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Changpei Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiting Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyi Liao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Geng Yin
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
De Bartolo A, Angelone T, Rocca C. Elucidating emerging signaling pathways driving endothelial dysfunction in cardiovascular aging. Vascul Pharmacol 2025; 158:107462. [PMID: 39805379 DOI: 10.1016/j.vph.2025.107462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
The risk for developing cardiovascular diseases dramatically increases in older individuals, and aging vasculature plays a crucial role in determining their morbidity and mortality. Aging disrupts endothelial balance between vasodilators and vasoconstrictors, impairing function and promoting pathological vascular remodeling. In this Review, we discuss the impact of key and emerging molecular pathways that transduce aberrant inflammatory signals (i.e., chronic low-grade inflammation-inflammaging), oxidative stress, and mitochondrial dysfunction in aging vascular compartment. We focus on the interplay between these events, which contribute to generating a vicious cycle driving the progressive alterations in vascular structure and function during cardiovascular aging. We also discuss the primary role of senescent endothelial cells and vascular smooth muscle cells, and the potential link between vascular and myeloid cells, in impairing plaque stability and promoting the progression of atherosclerosis. The aim of this summary is to provide potential novel insights into targeting these processes for therapeutic benefit.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
43
|
Zalghout S, Martinod K. Therapeutic potential of DNases in immunothrombosis: promising succor or uncertain future? J Thromb Haemost 2025; 23:760-778. [PMID: 39667687 DOI: 10.1016/j.jtha.2024.11.028] [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: 09/24/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024]
Abstract
Sepsis, a life-threatening condition characterized by systemic inflammation and multiorgan dysfunction, is closely associated with the excessive formation of neutrophil extracellular traps (NETs) and the release of cell-free DNA. Both play a central role in sepsis progression, acting as major contributors to immunothrombosis and associated complications. Endogenous DNases play a pivotal role in degrading NETs and cell-free DNA, yet their activity is often dysregulated during thrombotic disease. Although exogenous DNase1 administration has shown potential in reducing NET burden and mitigating the detrimental effects of immunothrombosis, its therapeutic efficacy upon intravenous administration remains uncertain. The development of engineered DNase formulations and combination therapies may further enhance its therapeutic effectiveness by modifying its pharmacodynamic properties and avoiding the adverse effects associated with NET degradation, respectively. Although NETs are well-established targets of DNase1, it remains uncertain whether the positive effects of DNase1 on immunothrombosis are exclusively related to it's targeting of NETs or if other components contributing to immunothrombosis are also affected. This review examines the endogenous regulation of NETs in circulation and the therapeutic potential of DNases in immunothrombosis, underscoring the necessity for further investigation to optimize their clinical application.
Collapse
Affiliation(s)
- Sara Zalghout
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| |
Collapse
|
44
|
Rajput S, Malviya R, Srivastava S, Ahmad I, Rab SO, Uniyal P. Cardiovascular disease and thrombosis: Intersections with the immune system, inflammation, and the coagulation system. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:228-250. [PMID: 39159826 DOI: 10.1016/j.pharma.2024.08.005] [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: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The coagulation and immune system, both essential physiological systems in the human body, are intricately interconnected and play a critical role in determining the overall health of patients. These systems collaborate via various shared regulatory pathways, such as the Tissue Factor (TF) Pathway. Immunological cells that express TF and generate pro-inflammatory cytokines have the ability to affect coagulation. Conversely, coagulation factors and processes have a reciprocal effect on immunological responses by stimulating immune cells and regulating their functions. These interconnected pathways play a role in both preserving well-being and contributing to a range of pathological disorders. The close relationship between blood clotting and inflammation in the development of vascular disease has become a central focus of clinical study. This research specifically examines the crucial elements of this interaction within the contexts of cardiovascular disease and acute coronary syndrome. Tissue factor, the primary trigger of the extrinsic coagulation pathway, has a crucial function by inducing a proinflammatory reaction through the activation of coagulation factors. This, in turn, initiates coagulation and subsequent cellular signalling pathways. Protease-activated receptors establish the molecular connection between coagulation and inflammation by interacting with activated clotting factors II, X, and VII. Thrombosis, a condition characterised by the formation of blood clots, is the most dreaded consequence of cardiovascular disorders and a leading cause of death globally. Consequently, it poses a significant challenge to healthcare systems. Antithrombotic treatments efficiently target platelets and the coagulation cascade, but they come with the inherent danger of causing bleeding. Furthermore, antithrombotics are unable to fully eliminate thrombotic events, highlighting a treatment deficiency caused by a third mechanism that has not yet been sufficiently addressed, namely inflammation. Understanding these connections may aid in the development of novel approaches to mitigate the harmful mutual exacerbation of inflammation and coagulation. Gaining a comprehensive understanding of the intricate interaction among these systems is crucial for the management of diseases and the creation of efficacious remedies. Through the examination of these prevalent regulatory systems, we can discover novel therapeutic approaches that specifically target these complex illnesses. This paper provides a thorough examination of the reciprocal relationship between the coagulation and immune systems, emphasising its importance in maintaining health and understanding disease processes. This review examines the interplay between inflammation and thrombosis and its role in the development of thrombotic disorders.
Collapse
Affiliation(s)
- Shivam Rajput
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India.
| | - Saurabh Srivastava
- School of Pharmacy, KPJ Healthcare University College (KPJUC), Nilai, Malaysia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
45
|
Potempa M, Hart PC, Rajab IM, Potempa LA. Redefining CRP in tissue injury and repair: more than an acute pro-inflammatory mediator. Front Immunol 2025; 16:1564607. [PMID: 40093010 PMCID: PMC11906453 DOI: 10.3389/fimmu.2025.1564607] [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: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Most early studies investigating the role of C-reactive protein (CRP) in tissue damage determined it supported pro-hemostatic and pro-inflammatory activities. However, these findings were not universal, as other data suggested CRP inhibited these same processes. A potential explanation for these disparate observations finally emerged with the recognition that CRP undergoes context-dependent conformational changes in vivo, and each of its three isoforms - pentameric CRP (pCRP), modified pentameric CRP (pCRP*), and monomeric CRP (mCRP) - have different effects. In this review, we consider this new paradigm and re-evaluate the role of CRP and its isoforms in the tissue repair process. Indeed, a growing body of evidence points toward the involvement of CRP not just in hemostasis and inflammation, but also in the resolution of inflammation and in tissue regeneration. Additionally, we briefly discuss the shortcomings of the currently available diagnostic tests for CRP and highlight the need for change in how CRP is currently utilized in clinical practice.
Collapse
Affiliation(s)
| | - Peter C. Hart
- College of Science, Health, and Pharmacy, Roosevelt University, Schaumburg, IL, United States
| | - Ibraheem M. Rajab
- College of Science, Health, and Pharmacy, Roosevelt University, Schaumburg, IL, United States
| | - Lawrence A. Potempa
- Acphazin Inc., Deerfield, IL, United States
- College of Science, Health, and Pharmacy, Roosevelt University, Schaumburg, IL, United States
| |
Collapse
|
46
|
Xue M, Wang S, Li C, Wang Y, Liu M, Huang X, Wang G, Yin Q, Xiao D, Yang S, Yan M, Niu L, Awais M, Shen C, Wang J, Lai R, Ni H, Tang X. Deficiency of neutrophil gelatinase-associated lipocalin elicits a hemophilia-like bleeding and clotting disorder in mice. Blood 2025; 145:975-987. [PMID: 39693621 DOI: 10.1182/blood.2024026476] [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: 08/08/2024] [Revised: 10/15/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
ABSTRACT Coagulation is related to inflammation, but the key pathway, especially innate immune system and coagulation regulation, is not well understood and need to be further explored. Here, we demonstrated that neutrophil gelatinase-associated lipocalin (NGAL), an innate immune inflammatory mediator, is upregulated in patients with thrombosis. Furthermore, it contributes to the initiation and amplification of coagulation, hemostasis, and thrombosis. This occurs by enhancing tissue factor expression on the cell surface, potentiating various clotting factors such as thrombin, kallikrein, factor XIa (FXIa), and FVIIa, promoting thrombin-induced platelet aggregation, and inhibiting antithrombin. NGAL knockout led to strikingly prolonged clot reaction time and kinetic time in thromboelastography analysis, along with reduced thrombus generation angle and lower thrombus maximum amplitude, which were in line with remarkably prolonged activated partial thromboplastin time and prothrombin time. In several mouse hemostasis and thrombosis models, NGAL overexpression or IV administration promoted coagulation and hemostasis and aggravated thrombosis, whereas NGAL knockout or treatment with anti-NGAL monoclonal antibody significantly prolonged bleeding time and alleviated thrombus formation. Notably, NGAL knockout prolonged mouse tail bleeding time or artery occlusion time to over 40 or 60 minutes, respectively, resembling uncontrollable bleeding and clotting disorder seen in hemophilic mice. Furthermore, anti-NGAL monoclonal antibody treatment markedly reduced the formation of blood clots in inflammation-induced thrombosis models. Collectively, these findings unveil a previously unidentified role of NGAL in the processes of coagulation, hemostasis, and thrombosis, as well as the cross talk between innate immunity, inflammation, and coagulation. Thus, modulating NGAL levels could potentially help balance thrombotic and hemorrhagic risks.
Collapse
Affiliation(s)
- Min Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shaoying Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Changjiang Li
- Department of Emergency, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Center Medical Group), Qingdao, China
| | - Yuewei Wang
- Department of Vascular Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ming Liu
- Department of Pharmacology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiaoshan Huang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Gan Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Qikai Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dandan Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shuo Yang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Musan Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Liyuan Niu
- Department of Vascular Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Muhammad Awais
- Department of Pharmacology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Chuanbin Shen
- Department of Pharmacology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jianxun Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ren Lai
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital and Toronto Platelet Immunobiology Group, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Xiaopeng Tang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
47
|
Li H, Shan W, Zhao X, Sun W. Neutrophils: Linking Inflammation to Thrombosis and Unlocking New Treatment Horizons. Int J Mol Sci 2025; 26:1965. [PMID: 40076593 PMCID: PMC11901051 DOI: 10.3390/ijms26051965] [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: 01/02/2025] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Neutrophils play a key role in inflammatory responses and thrombosis, but their complex interactions in disease pathogenesis are not fully understood. This review examines the multifaceted roles of neutrophils, focusing on their activation, cytokine release, and formation of neutrophil extracellular traps (NETs), which contribute to host defense and thrombosis. We discuss the interaction between inflammation and coagulation, the direct effect of neutrophils on thrombus stability, and their involvement in pathological thrombotic diseases. The therapeutic potential of neutrophil drug loading in the treatment of thrombosis, as well as the clinical implications and future research directions, are highlighted. The aim of this review is to gain insight into the critical neutrophil-inflammation-thrombus axis and its potential as a therapeutic target for thrombotic diseases and to suggest possible directions for neutrophil-loaded drug therapy for thrombosis.
Collapse
Affiliation(s)
| | | | | | - Wei Sun
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (H.L.); (W.S.); (X.Z.)
| |
Collapse
|
48
|
Zhang Y, Ye J, Sun S, Li R, Tang S, Wang M, Sun G. Role of platelets and NETs in arterial thrombosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03921-6. [PMID: 39992420 DOI: 10.1007/s00210-025-03921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Arterial thrombosis is one of the main causes of mortality and mortality worldwide. Platelets are effectively targeted by antithrombotic strategies. However, current antiplatelet agents are often associated with a bleeding risk and single antiplatelet agent may not completely prevent thrombosis. Platelets, neutrophils, and neutrophil extracellular traps (NETs) have been found to play crucial synergistic roles in the pathological process of arterial thrombosis in recent years. Platelets play a key regulatory role in the formation of NETs, and NETs can enhance platelet aggregation and activation, further aggravating the process of arterial thrombosis. Targeting the interaction mechanisms of platelets and NETs may provide a promising approach to better prevent arterial thrombosis. This review highlights the current insight in the interaction of platelets and neutrophil-forming NETs and their mechanisms involved in the process of arterial thrombosis. Finally, we discuss the potential of interventions targeting platelets and NETs to treat arterial thrombosis.
Collapse
Affiliation(s)
- Yaqi Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jingxue Ye
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shiyi Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ruoyun Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shuang Tang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
49
|
Bryzek D, Gasiorek A, Kowalczyk D, Santocki M, Ciaston I, Dobosz E, Kolaczkowska E, Kjøge K, Kantyka T, Lech M, Potempa B, Enghild JJ, Potempa J, Koziel J. Non-classical neutrophil extracellular traps induced by PAR2-signaling proteases. Cell Death Dis 2025; 16:109. [PMID: 39971938 PMCID: PMC11840154 DOI: 10.1038/s41419-025-07428-z] [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: 09/09/2024] [Revised: 01/21/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
Neutrophil extracellular traps (NETs) are associated with diseases linked to aberrant coagulation. The blood clotting cascade involves a series of proteases, some of which induce NET formation via a yet unknown mechanism. We hypothesized that this formation involves signaling via a factor Xa (FXa) activation of the protease-activated receptor 2 (PAR2). Our findings revealed that NETs can be triggered in vitro by enzymatically active proteases and PAR2 agonists. Intravital microscopy of the liver vasculature revealed that both FXa infusion and activation of endogenous FX promoted NET formation, effects that were prevented by the FXa inhibitor, apixaban. Unlike classical NETs, these protease-induced NETs lacked bactericidal activity and their proteomic signature indicates their role in inflammatory disorders, including autoimmune diseases and carcinogenesis. Our findings suggest a novel mechanism of NET formation under aseptic conditions, potentially contributing to a self-amplifying clotting and NET formation cycle. This mechanism may underlie the pathogenesis of disseminated intravascular coagulation and other aseptic conditions.
Collapse
Affiliation(s)
- Danuta Bryzek
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Anna Gasiorek
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Dominik Kowalczyk
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Michal Santocki
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Izabela Ciaston
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewelina Dobosz
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Elzbieta Kolaczkowska
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Katarzyna Kjøge
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Maciej Lech
- LMU Hospital, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians University, Munich, Germany
| | - Barbara Potempa
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan Potempa
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - Joanna Koziel
- Microbiology Department, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
50
|
Zhou Q, Zhou X, Li J, Wang R, Xie F. Research progress on the relationship between neutrophil extra-cellular traps and autogenous arteriovenous fistula thrombosis. J Vasc Access 2025:11297298251317298. [PMID: 39935409 DOI: 10.1177/11297298251317298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Autogenous arteriovenous fistula (AVF) is the preferred vascular access for long-term hemodialysis, and thrombosis is one of the most common complications. In recent years, it has been found that neutrophil extra-cellular traps (NETs) play an important role in thrombosis. NETs are a kind of network structure with DNA as a skeleton and intercalated with a variety of granule proteins, proteolytic enzymes, antimicrobial peptides and histone proteins, which are released into the extracellular space by neutrophils after stimulation. In this paper, the NETs in the role of AVF thrombus formation and NETs in the value of prevention and cure of AVF thrombus complications were reviewed.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Nephrology, The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xuhua Zhou
- Department of Nephrology, The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junlin Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Runxiu Wang
- Department of Nephrology, The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Fuhua Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|