1
|
Zhu H, Liu F, Liao Y, Li H, Gao K, Liang X, Jiang H, Chen F, Wu J, Wang Q, Wang Y, Shuai X, Yi X. Biomimetic nanostructural materials based on placental amniotic membrane-derived nanofibers for self-healing and anti-adhesion during cesarean section. Biomaterials 2025; 317:123081. [PMID: 39787897 DOI: 10.1016/j.biomaterials.2024.123081] [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: 09/23/2024] [Revised: 12/05/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
Cesarean section (CS) is highly prevalent surgery among females. However, current absorbable anti-adhesion membranes used clinically can partially prevent postoperative adhesions but show limited efficacy in tissue regeneration, leaving post-cesarean women at risk for severe complications including cesarean scar pregnancy, placenta previa, and uterine rupture. Herein, we designed a fully amniotic membrane (AM)-derived biomimetic nanostructural materials (AM-BNMs) as an anti-adhesion barrier, and validated its therapeutic efficacy in a rat CS model. The biomaterial consisted of AM-extracellular matrix (ECM) nanofibers, enriched with hemostatic proteins (collagen, S100A8, S100A9, etc.), carrying AM mesenchymal stem cells (MSCs)-secretome that exhibited significantly elevated levels of pro-regenerative factors (miR-302a-3p, angiogenin, VEGF, etc.) compared to endogenous secretion. The reconstituted AM-BNMs demonstrated synergistic effects at CS wounds, effectively preventing adhesion formation while promoting hemostasis and tissue regeneration. In summary, this readily accessible human-derived biomaterial shows promising potential in preventing adhesion-related complications and enhancing uterine wound healing, thereby promoting female reproductive health.
Collapse
Affiliation(s)
- Honglei Zhu
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Fenghua Liu
- Center for Reproductive Medicine, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Yuru Liao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huayan Li
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kunjie Gao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaomei Liang
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Haoyuan Jiang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Feng Chen
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jianwei Wu
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qian Wang
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yifeng Wang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Xiao Yi
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511462, China.
| |
Collapse
|
2
|
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
|
3
|
Lei XX, Liu ML, Lu CF, Han LL, Jia JZ, Li Z, Xu N, Li JF, Fu XJ, Jin YB, Tong RK, Yu YL, Luo GX, Chen Y. A self-hygroscopic, rapidly self-gelling polysaccharide-based sponge with robust wet adhesion for non-compressible hemorrhage control and infected wounds healing. Bioact Mater 2025; 46:311-330. [PMID: 39811462 PMCID: PMC11732608 DOI: 10.1016/j.bioactmat.2024.12.016] [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: 09/11/2024] [Revised: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Uncontrollable non-compressible hemorrhage and traumatic infection have been major causes of mortality and disability in both civilian and military populations. A dressing designed for point-of-care control of non-compressible hemorrhage and prevention of traumatic infections represents an urgent medical need. Here, a novel self-gelling sponge OHN@ε-pL is developed, integrating N-succinimidyl ester oxidized hyaluronic acid (OHN) and ε-poly-L-lysine (ε-pL). Upon application to the wound site, the sponge can rapidly absorb interfacial fluids and undergo a phase transition from sponge to gel. The transformed gel facilitates robust tissue adhesion and achieves synergistic hemostasis by enriching coagulation factors within the sponge phase and providing a barrier effect in the gel phase. The in vitro and in vivo studies revealed that the optimized OHN@ε-pL3 sponge possesses self-gelling capability, tissue adhesion, enhanced coagulation ability, and exhibits excellent biocompatibility and antibacterial efficacy. In hemostasis, OHN@ε-pL3 sponges exhibited reduced blood loss and decreased hemostatic time compared to commercial hemostatic agents, as demonstrated in rat liver, femoral vein, and tail truncation bleeding models. Furthermore, the OHN@ε-pL3 sponge exhibited superior performance in accelerating wound closure and healing of S. aureus-infected wounds. Collectively, OHN@ε-pL sponges represent a promising candidate for medical dressings, specifically for managing uncontrollable non-compressible hemorrhage and traumatic infections.
Collapse
Affiliation(s)
- Xiong-Xin Lei
- Department of Orthopedic Surgery, First People's Hospital of Foshan, Foshan, Guangdong, 528000, PR China
| | - Meng-Long Liu
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Chao-Feng Lu
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Li-Li Han
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Jie-Zhi Jia
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Zheng Li
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Na Xu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Jiang-Feng Li
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Xuan-Jian Fu
- Department of Orthopedic Surgery, First People's Hospital of Foshan, Foshan, Guangdong, 528000, PR China
| | - Ya-Bin Jin
- Department of Orthopedic Surgery, First People's Hospital of Foshan, Foshan, Guangdong, 528000, PR China
- School of Computer and Communication Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Ri-Kuan Tong
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Yun-Long Yu
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Gao-Xing Luo
- Institute of Burn Research, Southwest Hospital & State Key Lab of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Yang Chen
- Department of Orthopedic Surgery, First People's Hospital of Foshan, Foshan, Guangdong, 528000, PR China
| |
Collapse
|
4
|
Shi G, Cao Y, Xu J, Chen B, Zhang X, Zhu Y, Liu L, Liu X, Zhang L, Zhou Y, Li S, Yang G, Liu X, Chen F, Chen X, Zhang J, Zhang S. Inhibition of S100A8/A9 ameliorates neuroinflammation by blocking NET formation following traumatic brain injury. Redox Biol 2025; 81:103532. [PMID: 39929053 PMCID: PMC11849670 DOI: 10.1016/j.redox.2025.103532] [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: 12/13/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Traumatic brain injury (TBI) triggers a robust inflammatory response that is closely linked to worsened clinical outcomes. S100A8/A9, also known as calprotectin or myeloid-related protein-8/14 (MRP8/14), is an alarmin primarily secreted by activated neutrophils with potent pro-inflammatory property. In this study, we explored the roles of S100A8/A9 in modulating neuroinflammation and influencing TBI outcomes, delving into the underlying mechanisms. S100A8/A9-enriched neutrophils were present in the injured brain tissue of TBI patients, and elevated plasma levels of S100A8/A9 were correlated with poorer neurological function. Furthermore, using a TBI mouse model, we demonstrated that treatment with the selective S100A8/A9 inhibitor Paquinimod significantly mitigated neuroinflammation and neuronal death, thereby improving the prognosis of TBI mice. Mechanistically, we found that S100A8/A9, in conjunction with neutrophil activation and infiltration into the brain, enhances reactive oxygen species (ROS) production within neutrophils, accelerating PAD4-mediated neutrophil extracellular trap (NET) formation, which in turn exacerbates neuroinflammation. These findings suggest that S100A8/A9 amplifies neuroinflammatory responses by promoting NET formation in neutrophils. Inhibition of S100A8/A9 effectively attenuated NET-mediated neuroinflammation; however, when PAD4 was overexpressed in the brain using adenovirus, leading to an increased formation of NET in the brain, the anti-inflammatory effects of S100A8/A9 inhibition were markedly diminished. Further experiments with PAD4 knockout mice confirmed that the reduction of NETs could substantially alleviate S100A8/A9-driven neuroinflammation. Finally, we established that the suppression of NET formation by S100A8/A9 inhibition is primarily mediated through the AMPK/Nrf2/HO-1 signaling pathway. These findings underscore the critical pathological role of S100A8/A9 in TBI and emphasize the need for further exploration of S100A8/A9 inhibitor Paquinimod as a potential therapeutic strategy for TBI.
Collapse
Affiliation(s)
- Guihong Shi
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Yiyao Cao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Jianye Xu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Bo Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xu Zhang
- School of Medicine, Nankai University, Tianjin, 300052, China
| | - Yanlin Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Liang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xilei Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Luyuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Yuan Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Shenghui Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Guili Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xiao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China.
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, 300052, China.
| |
Collapse
|
5
|
Lei B, Mu J, Xu G, Yang X, Huang W, Hu L, Liu D, Cheng T, Ma Y, Xu L, Liang Q, Lin Y, Zhou L, Zhou C, Zhang W, Zheng Y. Jing-Yin-Gu-Biao formula protects mice from postinfluenza Staphylococcus aureus infection by ameliorating acute lung injury and improving hypercoagulable state via inhibiting NETosis. Front Immunol 2025; 16:1567522. [PMID: 40134435 PMCID: PMC11933027 DOI: 10.3389/fimmu.2025.1567522] [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: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Background Jing-Yin-Gu-Biao formula (JYGBF) is a Chinese medicine derived from Yupingfeng power, Huoxiangzhengqi powder and Yinqiao powder, and has been widely used to treat acute respiratory infections. This study aims to observe the effects of JYGBF against postinfluenza Staphylococcus aureus (S. aureus) infection. Purpose and study design A mouse model of secondary S. aureus infection following PR8 infection was established to evaluate the protective effects of JYGBF against postinfluenza Staphylococcus aureus (S. aureus) infection and related mechanisms were validated in vivo and in vitro. Results The administration of JYGBF significantly ameliorated acute lung injury (ALI) and inhibited overactivated inflammatory response (MIP-2, IL-6, etc.) in mice with postinfluenza S. aureus infection. Single cell RNA-sequencing (scRNA-seq) data indicated that neutrophils had the highest cytokine score in lungs and JYGBF inhibited neutrophil chemotaxis, reactive oxygen species (ROS) biosynthesis and ERK1/2 cascades in neutrophils. Meanwhile, JYGBF inhibited the formation of neutrophil extracellular traps (NETs) in lungs, which is characterized by the production of ROS, peptidyl arginine deiminase 4 (PAD4), citrullinated histone H3 (CitH3), myeloperoxidase (MPO), neutrophil elastase (NE), S100A8/A9 and MPO-CitH3 colocalization. Moreover, JYGBF decreased platelet counts and the expression of its activated markers (CD62P and αIIbβ3) accompanied by the drop of fibrinogen (FIB) and fibrin degradation product (FDP), accounting for alleviating hypercoagulable state. JYGBF inhibited ERK1/2 phosphorylation in neutrophils and in lungs of infected mice. Acacetin, a critical compound from JYGBF, inhibited NET formation via downregulating ERK/ROS axis. Conclusions These results indicated that JYGBF inhibited NET formation and overactivated inflammatory response by suppressing ERK/ROS axis in neutrophils, thereby mitigating ALI and improving the hypercoagulable state during postinfluenza S. aureus infection. JYGBF could be considered a potent therapeutic agent for the prevention and treatment of postinfluenza bacterial infection.
Collapse
Affiliation(s)
- Biao Lei
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingwen Mu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guihua Xu
- Department of Pulmonary Diseases, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaodong Yang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenbo Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liang Hu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Liu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Cheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuhe Ma
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lirong Xu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiankun Liang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Lin
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linqiong Zhou
- Shuguang Hospital, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine Epidemic Research Center, Shanghai, China
- Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Respiratory and Critical Care Medicine, Shanghai, China
| | - Chunxian Zhou
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Shuguang Hospital, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine Epidemic Research Center, Shanghai, China
- Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Respiratory and Critical Care Medicine, Shanghai, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Ma L, Xie L, Wu Q, Jin L, Li J, Tang L, Zhang L, Chen L, Qiu Z. Targeting the S100A9/P38 MAPK/HSPB1 axis as a novel approach for aortic dissection therapy. Int Immunopharmacol 2025; 149:114225. [PMID: 39904041 DOI: 10.1016/j.intimp.2025.114225] [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: 11/28/2024] [Revised: 01/02/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Aortic dissection (AD) is caused by inflammatory responses and extracellular matrix (ECM) degradation processes, in which S100A9, a proinflammatory protein, may play a role. This study explored the role S100A9/P38 MAPK/HSPB1 signaling axis in AD pathogenesis and the therapeutic potential of targeting this pathway. METHODS S100A9 expression in the aortic tissues of patients with AD/healthy controls were analyzed using bioinformatics, ELISA, qPCR, western blotting, and immunohistochemistry. In an AD mouse model induced by β-aminopropionitrile and angiotensin II (Ang-II), S100A9 expression was inhibited using specific inhibitors to assess its relationship with AD, and proteomics were performed to explore the pathways related to S100A9 expression. Human aortic vascular smooth muscle cells (HVSMC) were treated with Ang-II, S100A9 knockdown, P38 MAPK inhibitors, and HSPB1 knockdown, and experimental methods were used to assess changes in inflammatory cytokines, ECM remodeling, cell proliferation, and apoptosis. Rescue experiments validated the role of the S100A9/P38 MAPK/HSPB1 axis. RESULTS S100A9 was significantly upregulated in patients with AD, while levels of inflammatory cytokines and matrix metalloproteinases (MMPs) were elevated. S100a9 inhibition reduced the incidence of AD, improved survival, and stabilized the aortic structure in mice, with reduced collagen deposition and SMC apoptosis in vitro. S100A9 knockdown reduces Ang-II-induced HVSMC proliferation, apoptosis resistance, and ECM degradation. Mechanistic studies revealed that the S100A9/P38 MAPK/HSPB1 axis regulates inflammatory cytokine and MMPs release. CONCLUSION S100A9 regulates inflammation and ECM degradation through the P38 MAPK/HSPB1 axis, influencing HVSMC proliferation and apoptosis and promoting AD development. This pathway may be a promising therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Likang Ma
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Linfeng Xie
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Qingsong Wu
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Lei Jin
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Jiakang Li
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences Fujian Medical University Fuzhou China
| | - Lele Tang
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China
| | - Li Zhang
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences Fujian Medical University Fuzhou China.
| | - Liangwan Chen
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China.
| | - Zhihuang Qiu
- Department of Cardiovascular Surgery Fujian Medical University Union Hospital Fuzhou Fujian China; Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University) Fujian Province University Fuzhou Fujian China.
| |
Collapse
|
7
|
Sennett C, Pula G. Trapped in the NETs: Multiple Roles of Platelets in the Vascular Complications Associated with Neutrophil Extracellular Traps. Cells 2025; 14:335. [PMID: 40072064 PMCID: PMC11898727 DOI: 10.3390/cells14050335] [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: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Neutrophil extracellular traps (NETs) have received significant attention in recent years for their role in both the immune response and the vascular damage associated with inflammation. Platelets have been described as critical components of NETs since the initial description of this physio-pathological response of neutrophils. Platelets have been shown to play a dual role as responders and also as stimulators of NETs. The direct interaction with DNA leads to the entrapment of platelets into NETs, a phenomenon that significantly contributes to the thrombotic complications of inflammation and neutrophil activation, while the direct and paracrine stimulation of neutrophils by platelets has been shown to initiate the process of NET formation. In this review, we provide a comprehensive description of our current understanding of the molecular mechanisms underlying the entrapping of platelets into NETs and, in parallel, the platelet-driven cellular responses promoting NET formation. We then illustrate established examples of the contribution of NETs to vascular pathologies, describe the important questions that remain to be answered regarding the contribution of platelets to NET formation and NET-dependent cardiovascular complication, and highlight the fundamental steps taken towards the application of our understanding of platelets' contribution to NETs for the development of novel cardiovascular therapies.
Collapse
Affiliation(s)
| | - Giordano Pula
- Biomedical Institute for Multimorbidity (BIM), Hull York Medical School (HYMS), University of Hull, Hull HU6 7RX, UK
| |
Collapse
|
8
|
Gong F, Zheng X, Zhao S, Liu H, Chen E, Xie R, Li R, Chen Y. Disseminated intravascular coagulation: cause, molecular mechanism, diagnosis, and therapy. MedComm (Beijing) 2025; 6:e70058. [PMID: 39822757 PMCID: PMC11733103 DOI: 10.1002/mco2.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/19/2025] Open
Abstract
Disseminated intravascular coagulation (DIC) is a complex and serious condition characterized by widespread activation of the coagulation cascade, resulting in both thrombosis and bleeding. This review aims to provide a comprehensive overview of DIC, emphasizing its clinical significance and the need for improved management strategies. We explore the primary causes of DIC, including sepsis, trauma, malignancies, and obstetric complications, which trigger an overactive coagulation response. At the molecular level, DIC is marked by excessive thrombin generation, leading to platelet and fibrinogen activation while simultaneously depleting clotting factors, creating a paradoxical bleeding tendency. Diagnosing DIC is challenging and relies on a combination of existing diagnostic criteria and laboratory tests. Treatment strategies focus on addressing the underlying causes and may involve supportive care, anticoagulation therapy, and other supportive measures. Recent advances in understanding the pathophysiology of DIC are paving the way for more targeted therapeutic approaches. This review highlights the critical need for ongoing research to enhance diagnostic accuracy and treatment efficacy, ultimately improving patient outcomes in those affected by DIC.
Collapse
Affiliation(s)
- Fangchen Gong
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiangtao Zheng
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shanzhi Zhao
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huan Liu
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Erzhen Chen
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of Aviation Medicine, Shanghai Jiao Tong University Medical School Affiliated Ruijin HospitalShanghaiChina
| | - Rongli Xie
- Department of General SurgeryRuijin Hospital Lu Wan Branch, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Ranran Li
- Department of Critical Care MedicineRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Chen
- Department of EmergencyRuijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Emergency and Critical Care MedicineRuijin Hospital Wuxi Branch, Shanghai Jiao Tong University School of MedicineWuxiChina
| |
Collapse
|
9
|
Zhao X, Liu J, Jin C, Pan K, Du Y, Zuo Z, Yu X. S100A8 regulates postoperative responses following tooth extraction in rats. Odontology 2025:10.1007/s10266-025-01056-x. [PMID: 39878930 DOI: 10.1007/s10266-025-01056-x] [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: 09/08/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
The reduction in alveolar ridge height and width after tooth extraction poses a substantial challenge for dental implant restoration. This study aimed to observe the roles of S100A8 in the inflammatory response and bone resorption following tooth extraction. Rat mandibular second molars were extracted. Western blotting, micro-computed tomography, tartrate-resistant acid phosphatase staining, immunohistochemistry, scanning electron microscopy (SEM), and immunofluorescence were applied to observe inflammatory responses and bone resorption after rat mandibular second molar extraction. S100A8 was overexpressed in RAW 246.7 cells, and transfection efficiency was confirmed using western blotting and immunofluorescence. Receptor Activator for Nuclear Factor-κB Ligand (RANKL)-induced osteoclastogenesis of RAW 246.7 cells and a lipopolysaccharide-stimulated environment were chosen to evaluate the roles and related mechanisms of S100A8. An increase in S100A8 expression was observed 2 weeks after molar extraction compared to normal gingival tissues. Subsequently, S100 expression decreased between the second and third weeks, coinciding with the progression of inflammatory response following tooth extraction. Positive S100A8 expression was detected in the infiltrating cells and osteoclasts. S100A8 overexpression in RAW 246.7 cells promoted tumor necrosis factor-α expression and osteoclast formation by activating nuclear factor (NF)-κB p65 signaling. In summary, S100A8 was involved in inflammatory responses and bone resorption following tooth extraction by activating NF-κB p65 signaling. Interventions targeted to decrease S100A8 levels may have therapeutic implications for minimizing post-extraction bone loss.
Collapse
Affiliation(s)
- Xinyu Zhao
- School of Stomatology, Jinan Hospital, Jinan, 250000, Shandong, China
- Central Laboratory, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong, China
| | - Jian Liu
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
- Central Laboratory, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong, China
| | - Chaoran Jin
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
- Central Laboratory, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong, China
| | - Kexu Pan
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China
- Central Laboratory, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong, China
| | - Yi Du
- Central Laboratory, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong, China
| | - Zhibin Zuo
- Department of Periodontology, Central Laboratory, Jinan Stomatological Hospital, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan, 250001, Shandong, China.
| | - Xijiao Yu
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, Shandong, China.
- Central Laboratory, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Key Medical and Health Laboratory of Oral Diseases and Tissue Regeneration, Jinan Stomatological Hospital, Jinan, 250001, Shandong, China.
| |
Collapse
|
10
|
Zhang D, Dai Y, Xu X, Ma F, Wang M, Qin W. S100A8-CAMKK2-AMPK axis confers the protective effects of mild hypothermia against cerebral ischemia-reperfusion injury in rats. Sci Rep 2025; 15:2793. [PMID: 39843475 PMCID: PMC11754893 DOI: 10.1038/s41598-025-87184-4] [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: 10/21/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
To investigate the neuroprotective mechanism of mild hypothermia (MH) in ameliorating cerebral ischemia reperfusion (IR) injury. The Pulsinelli's four-vessel ligation method was utilized to establish a rat model of global cerebral IR injury. To investigate the role of S100A8 in MH treatment of cerebral IR injury, hippocampus-specific S100A8 loss or gain of function was achieved using an adeno-associated virus system. We examined the effect of S100A8 over-expression or knock-down on the function of the SH-SY5Y cell line subjected to oxygen-glucose deprivation reoxygenation (OGDR) injury under MH treatment and delved into the underlying mechanisms. MH significantly ameliorates IR-induced neurological injury in the brain. Similarly to MH, knock-down of S100A8 significantly reduced neuronal oxidative stress, attenuated mitochondrial damage, inhibited apoptosis, and improved cognitive function in IR rats. Conversely, over-expression of S100A8 attenuated MH's protective effect and aggravated brain IR injury. In vitro, low expression of S100A8 significantly inhibited the decline in mitochondrial membrane potential induced by OGDR, reduced oxidative stress response, and decreased cell apoptosis, acting as a protective agent nearly equivalent to MH in SH-SY5Y cells. However, over-expression of S100A8 significantly inhibited these protective effects of MH. Mechanistically, MH down-regulated S100A8 expression, enhancing mitochondrial function via activation of the CAMKK2/AMPK signaling pathway. Moreover, with MH treatment, the administration of CAMKK2 and AMPK inhibitors STO-609 and Dorsomorphin significantly increased oxidative stress, mitochondrial damage, and cell apoptosis, thereby diminishing MH's neuroprotective effect against cerebral IR injury. Our study identified S100A8 as a master regulator that enables MH to ameliorate neurological injury during the early stage of cerebral IR injury by enhancing mitochondrial function. By targeting the S100A8-initiated CAMKK2/AMPK signaling pathway, we may unlock a novel therapeutic intervention or develop a refined MH therapeutic strategy against cerebral IR injury.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Yuting Dai
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Xiaoyan Xu
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
- School of Anesthesiology, Shandong Second Medical University, Weifang, 262700, China
| | - Fuguo Ma
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Mingshan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Weiwei Qin
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
- Department of Anesthesiology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China.
| |
Collapse
|
11
|
Kaiser R, Dewender R, Mulkers M, Stermann J, Rossaro D, Di Fina L, Li L, Gold C, Schmid M, Kääb L, Eivers L, Akgöl S, Yue K, Kammerer L, Loew Q, Anjum A, Escaig R, Akhalkatsi A, Laun L, Kranich J, Brocker T, Mueller TT, Krächan A, Gmeiner J, Pekayvaz K, Thienel M, Massberg S, Stark K, Kilani B, Nicolai L. Procoagulant platelet activation promotes venous thrombosis. Blood 2024; 144:2546-2553. [PMID: 39440970 DOI: 10.1182/blood.2024025476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT Platelets are key players in cardiovascular disease, and platelet aggregation represents a central pharmacologic target, particularly in secondary prevention. However, inhibition of adenosine diphosphate and thromboxane signaling has low efficacy in preventing venous thromboembolism, necessitating the inhibition of the plasmatic coagulation cascade in this disease entity. Anticoagulation carries a significantly higher risk of bleeding complications, highlighting the need of alternative therapeutic approaches. We hypothesized that procoagulant activation (PA) of platelets promotes venous thrombus formation and that targeting PA could alleviate venous thrombosis. Here, we found elevated levels of procoagulant platelets in the circulation and in thrombi of patients with deep vein thrombosis (DVT) and pulmonary embolism, and in mice developing DVT following inferior vena cava stenosis. Furthermore, we detected PA of recruited platelets within murine venous thrombi and human pulmonary emboli. Mice with platelet-specific deficiency in central pathways of PA-cyclophilin D and transmembrane protein 16F-were more resistant toward low flow-induced venous thrombosis. Finally, we found that a clinically approved carbonic anhydrase inhibitor, methazolamide, reduced platelet procoagulant activity and alleviated murine thrombus formation without affecting trauma-associated hemostasis. These findings identify an essential role of platelet procoagulant function in venous thrombosis and delineate novel pharmacologic strategies targeting platelets in the prevention of venous thromboembolism.
Collapse
Affiliation(s)
- Rainer Kaiser
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Robin Dewender
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Maité Mulkers
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Julia Stermann
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Dario Rossaro
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Lea Di Fina
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Lukas Li
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Michael Schmid
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Lily Kääb
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Luke Eivers
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Sezer Akgöl
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Keyang Yue
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Lisa Kammerer
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Quentin Loew
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Afra Anjum
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Raphael Escaig
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Anastassia Akhalkatsi
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Lisa Laun
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Jan Kranich
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig Maximilian University, Munich, Germany
| | - Thomas Brocker
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig Maximilian University, Munich, Germany
| | - Tonina T Mueller
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Angelina Krächan
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Jonas Gmeiner
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Kami Pekayvaz
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Manuela Thienel
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Steffen Massberg
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Konstantin Stark
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Badr Kilani
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| | - Leo Nicolai
- Department of Medicine I, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Berlin, Germany
| |
Collapse
|
12
|
Lagrange J, Ahmed MU, Arnone D, Lacolley P, Regnault V, Peyrin-Biroulet L, Denis CV. Implications of von Willebrand Factor in Inflammatory Bowel Diseases: Beyond Bleeding and Thrombosis. Inflamm Bowel Dis 2024; 30:2500-2508. [PMID: 38960879 DOI: 10.1093/ibd/izae142] [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: 03/15/2024] [Indexed: 07/05/2024]
Abstract
Inflammatory bowel disease (IBD) displays an increased venous and arterial thrombotic risk despite the common occurrence of intestinal bleeding. While some of the mechanisms leading to these thrombotic complications have been studied, other specific changes in the hemostasis profile of IBD patients have been less explored. One such example relates to von Willebrand factor (VWF) whose plasma levels have been reported to be modulated in IBD. Von Willebrand factor is a plasma glycoprotein crucial for hemostatic functions via roles both in platelet function and coagulation. High plasma VWF is a known risk factor for venous thromboembolism. In addition to its canonical roles in hemostasis, VWF is known to be directly or indirectly involved in other vascular processes such as maintenance of endothelial barrier integrity or proliferation of vascular smooth muscle cells. The purpose of this review is to recapitulate and update the existing data about VWF biology in IBD and to highlight its role both in the existing procoagulant phenotype and in vascular alterations that may occur in IBD.
Collapse
Affiliation(s)
- Jérémy Lagrange
- Université de Lorraine, INSERM, DCAC, Nancy, France
- CHRU Nancy, IHU INFINY, Vandœuvre-lès-Nancy, France
| | | | - Djésia Arnone
- Université de Lorraine, INSERM, NGERE, IHU INFINY, Nancy, France
| | | | | | - Laurent Peyrin-Biroulet
- Université de Lorraine, INSERM, NGERE, IHU INFINY, Nancy, France
- Department of Gastroenterology, CHRU Nancy, Vandœuvre-lès-Nancy, France
- Groupe Hospitalier privé Ambroise Paré - Hartmann, Paris IBD center, Neuilly sur Seine, France
| | - Cécile V Denis
- HITh, UMR_S1176, INSERM, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
13
|
Xia Y, Sun C, Zhou K, Shen J, Li J, Huang Q, Du J, Zhang S, Sun K, Hu R, Yan R, Dai K. Platelet Glycoprotein Ibα Cytoplasmic Tail Exacerbates Thrombosis During Bacterial Sepsis. Int J Mol Sci 2024; 25:11548. [PMID: 39519103 PMCID: PMC11546206 DOI: 10.3390/ijms252111548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Septic patients, coupling severe disseminated intravascular coagulation (DIC) and thrombocytopenia, have poor prognoses and higher mortality. The platelet glycoprotein Ibα (GPIbα) is involved in thrombosis, hemostasis, and inflammation response. However, it remains unclear whether the GPIbα cytoplasmic tail regulates sepsis-mediated platelet activation and inflammation, especially in Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) infections. Using a mouse model of S. aureus-induced bacteremia, we found that both 10 amino acids of GPIbα C-terminal sequence deficiency and pharmacologic inhibition of protein kinase C (PKC) alleviated pathogenesis by diminishing platelet activation and aggregate formation. Furthermore, the GPIbα cytoplasmic tail promoted the phagocytosis of platelets by Kupffer cells in vivo. The genetically deficient GPIbα cytoplasmic tail also downregulated inflammatory cytokines and reduced liver damage, ultimately improving the survival rate of the septic mice. Our results illustrate that the platelet GPIbα cytoplasmic domain exacerbates excessive platelet activation and inflammation associated with sepsis through a PKC-dependent pathway. Thus, our findings provide insights for the development of effective therapeutic strategies using PKC inhibitor treatment against bacterial infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Rong Yan
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, Suzhou Medical College of Soochow University, NHC Key Laboratory of Thrombosis and Hemostasis, National Clinical Research Center for Hematological Diseases, Suzhou 215123, China
| | - Kesheng Dai
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, Suzhou Medical College of Soochow University, NHC Key Laboratory of Thrombosis and Hemostasis, National Clinical Research Center for Hematological Diseases, Suzhou 215123, China
| |
Collapse
|
14
|
Wang Z, Wang Y, Yan Q, Cai C, Feng Y, Huang Q, Li T, Yuan S, Huang J, Luo ZH, Zhou J. FPR1 signaling aberrantly regulates S100A8/A9 production by CD14 +FCN1 hi macrophages and aggravates pulmonary pathology in severe COVID-19. Commun Biol 2024; 7:1321. [PMID: 39402337 PMCID: PMC11473795 DOI: 10.1038/s42003-024-07025-4] [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: 01/30/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Excessive alarmins S100A8/A9 escalate the inflammation and even exacerbate immune-driven thrombosis and multi-organ damage. However, the regulatory mechanisms of S100A8/A9 expression in infectious diseases remain unclear. In this study, high-dimensional transcriptomic data analyses revealed a high proportion of CD14+FCN1hi macrophages within the pulmonary niche post-severe SARS-CoV-2 infection. By constructing the S100-coexpression gene list and supervised module scoring, we found that CD14+FCN1hi macrophages presented the highest scores of alarmin S100, and possibly served as the trigger and amplifier of inflammation in severe COVID-19. These CD14+FCN1hi cells lacked the positive regulatory activity of transcription factor PPARγ, and lost their differentiation ability towards mature macrophages. Ex vivo experiments further validated that the epithelial cells with high ORF-3a expression promoted the expression and secretion of S100A8/A9 through ANXA1/SAA1-FPR1 signaling. S100A8/A9 heterodimers, as well as the co-localization of S100A8/A9 with microtubules, were both diminished by the FPR1 inhibitor. Phospho-kinase protein array indicated that STAT3 promoted transcription, and PLC-γ and ERK1/2 pathways were involved in the hetero-dimerization and unconventional secretion of S100A8/A9. Our study highlights the pivotal role of FPR1 signaling in the excessive production of S100A8/A9 and provides a promising target for the prevention and control of severe COVID-19 and post-acute COVID-19 sequelae.
Collapse
Affiliation(s)
- Zhongyi Wang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yi Wang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Qing Yan
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Changlin Cai
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Ying Feng
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Qinghan Huang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Shenzhen Yuan
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Juan Huang
- Department of Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Hui Luo
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China.
| | - Jingjiao Zhou
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
Bai H, Zhang S, Huang J, Diao K, Li C, Wang M. Unraveling the pathogenic interplay between SARS-CoV-2 and polycystic ovary syndrome using bioinformatics and experimental validation. Sci Rep 2024; 14:22934. [PMID: 39358491 PMCID: PMC11448505 DOI: 10.1038/s41598-024-74347-y] [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: 05/04/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
The prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) among polycystic ovary syndrome (PCOS) is significantly higher than in the general population. However, the mechanisms underlying this remain obscure. This study aimed to explore the mechanisms by identifying the genetic signature of SARS-CoV-2 infection in PCOS. In the present study, a total of 27 common differentially expressed genes (DEGs) were selected for subsequent analyses. Functional analyses showed that immunity and hormone-related pathways collectively participated in the development and progression of PCOS and SARS-CoV-2 infection. Under these, 7 significant hub genes were identified, including S100A9, MMP9, TLR2, THBD, ITGB2, ICAM1, and CD86 by using the algorithm in Cytoscape. Furthermore, hub gene expression was confirmed in the validation set, PCOS clinical samples, and mouse model. Immune microenvironment analysis with the CIBERSORTx database demonstrated that the hub genes were significantly correlated with T cells, dendritic cells, mast cells, B cells, NK cells, and eosinophils and positively correlated with immune scores. Among the hub genes, S100A9, MMP9, THBD, ITGB2, CD86, and ICAM1 demonstrated potential as possible diagnostic markers for COVID-19 and PCOS. In addition, we established the interaction networks of ovary-specific genes, transcription factors, miRNAs, drugs, and chemical compounds with hub genes with NetworkAnalyst. This work uncovered the common pathogenesis and genetic signature of PCOS and SARS-CoV-2 infection, which might provide a theoretical basis and innovative ideas for further mechanistic research and drug discovery of the comorbidity of the two diseases.
Collapse
Affiliation(s)
- Hai Bai
- Institute of Applied Biotechnology, College of Agronomy and Life Science, Shanxi Datong University, Datong, 037009, Shanxi, PR China
| | - Shanshan Zhang
- School of Biological Science, Jining Medical University, Rizhao, 276826, Shandong, PR China
| | - Jing Huang
- Department of Medical Informatics Engineering, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China
| | - Kangyang Diao
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China
| | - Cui Li
- Department of Physiology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
- National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
| | - Mingming Wang
- Department of Physiology, School of Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
- National Experimental Teaching Demonstration Center for Basic Medicine, Xuzhou Medical University, Xuzhou, 221009, Jiangsu, PR China.
| |
Collapse
|
16
|
Suski M, Olszanecka A, Stachowicz A, Kiepura A, Terlecki M, Madej J, Rajzer M, Olszanecki R. Alterations in plasma proteome during acute COVID-19 and recovery. Mol Med 2024; 30:131. [PMID: 39183264 PMCID: PMC11346252 DOI: 10.1186/s10020-024-00898-5] [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: 06/19/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The severe course of COVID-19 causes cardiovascular injuries, although the mechanisms involved are still not fully recognized, linked, and understood. Their characterization is of great importance with the establishment of the conception of post-acute sequelae of COVID-19, referred to as long COVID, where blood clotting and endothelial abnormalities are believed to be the key pathomechanisms driving circulatory system impairment. METHODS The presented study investigates temporal changes in plasma proteins in COVID-19 patients during hospitalization due to SARS-CoV-2 infection and six months after recovery by targeted SureQuant acquisition using PQ500 panel. RESULTS In total, we identified 167 proteins that were differentially regulated between follow-up and hospitalization, which functionally aggregated into immune system activation, complement and coagulation cascades, interleukins signalling, platelet activation, and extracellular matrix organization. Furthermore, we found that temporal quantitative changes in acute phase proteins correlate with selected clinical characteristics of COVID-19 patients. CONCLUSIONS In-depth targeted proteome investigation evidenced substantial changes in plasma protein composition of patients during and recovering from COVID-19, evidencing a wide range of functional pathways induced by SARS-CoV-2 infection. In addition, we show that a subset of acute phase proteins, clotting cascade regulators and lipoproteins could have clinical value as potential predictors of long-term cardiovascular events in COVID-19 convalescents.
Collapse
Affiliation(s)
- Maciej Suski
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka str, Kraków, 31 531, Poland.
| | - Agnieszka Olszanecka
- Department of Cardiology, Interventional Electrocardiology and Arterial Hypertension, Jagiellonian University Medical College, 2 Jakubowskiego str, Kraków, 30-688, Poland
- University Hospital in Kraków, 2 Jakubowskiego str, Kraków, 30-688, Poland
| | - Aneta Stachowicz
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka str, Kraków, 31 531, Poland
| | - Anna Kiepura
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka str, Kraków, 31 531, Poland
| | - Michał Terlecki
- Department of Cardiology, Interventional Electrocardiology and Arterial Hypertension, Jagiellonian University Medical College, 2 Jakubowskiego str, Kraków, 30-688, Poland
- University Hospital in Kraków, 2 Jakubowskiego str, Kraków, 30-688, Poland
| | - Józef Madej
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka str, Kraków, 31 531, Poland
| | - Marek Rajzer
- Department of Cardiology, Interventional Electrocardiology and Arterial Hypertension, Jagiellonian University Medical College, 2 Jakubowskiego str, Kraków, 30-688, Poland
- University Hospital in Kraków, 2 Jakubowskiego str, Kraków, 30-688, Poland
| | - Rafał Olszanecki
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka str, Kraków, 31 531, Poland
| |
Collapse
|
17
|
Chen J, Chen C, Wang L, Feng X, Chen Y, Zhang R, Cheng Y, Liu Z, Chen Q. Identification of S100A8/A9 involved in thromboangiitis obliterans development using tandem mass tags-labeled quantitative proteomics analysis. Cell Signal 2024; 120:111199. [PMID: 38697446 DOI: 10.1016/j.cellsig.2024.111199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Thromboangiitis obliterans (TAO) is characterized by inflammation and obstruction of small-and medium-sized distal arteries, with limited pharmacotherapies and surgical interventions. The precise pathogenesis of TAO remains elusive. By utilizing the technology of tandem mass tags (TMT) for quantitative proteomics and leveraging bioinformatics tools, a comparative analysis of protein profiles was conducted between normal and TAO rats to identify key proteins driving TAO development. The results unveiled 1385 differentially expressed proteins (DEPs) in the TAO compared with the normal group-comprising 365 proteins with upregulated expression and 1020 proteins with downregulated expression. Function annotation through gene ontology indicated these DEPs mainly involved in cell adhesion, positive regulation of cell migration, and cytosol. The principal signaling pathways involved regulation of the actin cytoskeleton, vascular smooth contraction, and focal adhesion. The roles of these DEPs and associated signaling pathways serve as a fundamental framework for comprehending the mechanisms underpinning the onset and progression of TAO. Furthermore, we conducted a comprehensive evaluation of the effects of S100A8/A9 and its inhibitor, paquinimod, on smooth muscle cells (SMCs) and in TAO rats. We observed that paquinimod reduces SMCs proliferation and migration, promotes phenotype switching and alleviates vascular stenosis in TAO rats. In conclusion, our study revealed that the early activation of S100A8/A9 in the femoral artery is implicated in TAO development, targeting S100A8/A9 signaling may provide a novel approach for TAO prevention and treatment.
Collapse
Affiliation(s)
- Jing Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chunfang Chen
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lili Wang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyi Feng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinru Chen
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanyuan Cheng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Qi Chen
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
18
|
Rayes J, Brill A. Hot under the clot: venous thrombogenesis is an inflammatory process. Blood 2024; 144:477-489. [PMID: 38728383 DOI: 10.1182/blood.2023022522] [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: 12/27/2023] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
ABSTRACT Venous thrombosis (VT) is a serious medical condition in which a blood clot forms in deep veins, often causing limb swelling and pain. Current antithrombotic therapies carry significant bleeding risks resulting from targeting essential coagulation factors. Recent advances in this field have revealed that the cross talk between the innate immune system and coagulation cascade is a key driver of VT pathogenesis, offering new opportunities for potential therapeutic interventions without inducing bleeding complications. This review summarizes and discusses recent evidence from preclinical models on the role of inflammation in VT development. We highlight the major mechanisms by which endothelial cell activation, Weibel-Palade body release, hypoxia, reactive oxygen species, inflammasome, neutrophil extracellular traps, and other immune factors cooperate to initiate and propagate VT. We also review emerging clinical data describing anti-inflammatory approaches as adjuncts to anticoagulation in VT treatment. Finally, we identify key knowledge gaps and future directions that could maximize the benefit of anti-inflammatory therapies in VT. Identifying and targeting the inflammatory factors driving VT, either at the endothelial cell level or within the clot, may pave the way for new therapeutic possibilities for improving VT treatment and reducing thromboembolic complications without increasing bleeding risk.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
19
|
Yong J, Toh CH. The convergent model of coagulation. J Thromb Haemost 2024; 22:2140-2146. [PMID: 38815754 DOI: 10.1016/j.jtha.2024.05.014] [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: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
It is increasingly apparent that the pathologic interplay between coagulation and innate immunity, ie, immunothrombosis, forms the common basis of many challenges across the boundaries of specialized medicine and cannot be fully explained by the conventional concepts of cascade and cell-based coagulation. To improve our understanding of coagulation, we propose a model of coagulation that converges with inflammation and innate immune activation as a unified response toward vascular injury. Evolutionarily integral to the convergent response are damage-associated molecular patterns, which are released as a consequence of injury. Damage-associated molecular patterns facilitate diverse interactions within and between systems, not only to complement and reinforce cell-based clot formation but also to steer the response toward clot resolution and wound healing. By extending coagulation beyond its current boundaries, the convergent model aims to deliver novel diagnostics and therapeutics for contemporary and unexpected challenges across medicine, as exposed by COVID-19 and vaccine-induced immune thrombotic thrombocytopenia.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
20
|
Nunes M, Vlok M, Proal A, Kell DB, Pretorius E. Data-independent LC-MS/MS analysis of ME/CFS plasma reveals a dysregulated coagulation system, endothelial dysfunction, downregulation of complement machinery. Cardiovasc Diabetol 2024; 23:254. [PMID: 39014464 PMCID: PMC11253362 DOI: 10.1186/s12933-024-02315-x] [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: 03/22/2024] [Accepted: 06/16/2024] [Indexed: 07/18/2024] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic condition that is characterized by unresolved fatigue, post-exertion symptom exacerbation (PESE), cognitive dysfunction, orthostatic intolerance, and other symptoms. ME/CFS lacks established clinical biomarkers and requires further elucidation of disease mechanisms. A growing number of studies demonstrate signs of hematological and cardiovascular pathology in ME/CFS cohorts, including hyperactivated platelets, endothelial dysfunction, vascular dysregulation, and anomalous clotting processes. To build on these findings, and to identify potential biomarkers that can be related to pathophysiology, we measured differences in protein expression in platelet-poor plasma (PPP) samples from 15 ME/CFS study participants and 10 controls not previously infected with SARS-CoV-2, using DIA LC-MS/MS. We identified 24 proteins that are significantly increased in the ME/CFS group compared to the controls, and 21 proteins that are significantly downregulated. Proteins related to clotting processes - thrombospondin-1 (important in platelet activation), platelet factor 4, and protein S - were differentially expressed in the ME/CFS group, suggestive of a dysregulated coagulation system and abnormal endothelial function. Complement machinery was also significantly downregulated, including C9 which forms part of the membrane attack complex. Additionally, we identified a significant upregulation of lactotransferrin, protein S100-A9, and an immunoglobulin variant. The findings from this experiment further implicate the coagulation and immune system in ME/CFS, and bring to attention the pathology of or imposed on the endothelium. This study highlights potential systems and proteins that require further research with regards to their contribution to the pathogenesis of ME/CFS, symptom manifestation, and biomarker potential, and also gives insight into the hematological and cardiovascular risk for ME/CFS individuals affected by diabetes mellitus.
Collapse
Affiliation(s)
- Massimo Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa
| | - Mare Vlok
- Central Analytical Facility: Mass Spectrometry, Stellenbosch University, Tygerberg Campus, Room 6054, Clinical Building, Francie Van Zijl Drive Tygerberg, Cape Town, 7505, South Africa
| | - Amy Proal
- PolyBio Research Foundation, 7900 SE 28th ST, Suite 412, Mercer Island, DC, 98040, USA
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Chemitorvet 200, 2800, Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| |
Collapse
|
21
|
Zeng W, Gao Y, Wang Q, Chi J, Zhu Z, Diao Q, Li X, Wang Z, Qu M, Shi Y. Preliminary clinical analysis and pathway study of S100A8 as a biomarker for the diagnosis of acute deep vein thrombosis. Sci Rep 2024; 14:13298. [PMID: 38858401 PMCID: PMC11164926 DOI: 10.1038/s41598-024-61728-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024] Open
Abstract
Herein, we aimed to identify blood biomarkers that compensate for the poor specificity of D-dimer in the diagnosis of deep vein thrombosis (DVT). S100A8 was identified by conducting protein microarray analysis of blood samples from patients with and without DVT. We used ELISA to detect S100A8, VCAM-1, and ICAM-1 expression levels in human blood and evaluated their correlations. Additionally, we employed human recombinant protein S100A8 to induce human umbilical vein endothelial cells and examined the role of the TLR4/MAPK/VCAM-1 and ICAM-1 signaling axes in the pathogenic mechanism of S100A8. Simultaneously, we constructed a rat model of thrombosis induced by inferior vena cava stenosis and detected levels of S100A8, VCAM-1, and ICAM-1 in the blood of DVT rats using ELISA. The associations of thrombus tissue, neutrophils, and CD68-positive cells with S100A8 and p38MAPK, TLR4, and VCAM-1 expression levels in vein walls were explored. The results revealed that blood S100A8 was significantly upregulated during the acute phase of DVT and activated p38MAPK expression by combining with TLR4 to enhance the expression and secretion of VCAM-1 and ICAM-1, thereby affecting the occurrence and development of DVT. Therefore, S100A8 could be a potential biomarker for early diagnosis and screening of DVT.
Collapse
Affiliation(s)
- Wenjie Zeng
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Yangyang Gao
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Qitao Wang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Junyu Chi
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ziyan Zhu
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Qingfei Diao
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Xin Li
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhen Wang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ming Qu
- Vascular Gland Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Yongquan Shi
- Department of Clinical Laboratory Center, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| |
Collapse
|
22
|
Boucher J, Gilbert C, Bose S, Tessier PA. S100A9: The Unusual Suspect Connecting Viral Infection and Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1523-1529. [PMID: 38709994 PMCID: PMC11076006 DOI: 10.4049/jimmunol.2300640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/20/2024] [Indexed: 05/08/2024]
Abstract
The study of S100A9 in viral infections has seen increased interest since the COVID-19 pandemic. S100A8/A9 levels were found to be correlated with the severity of COVID-19 disease, cytokine storm, and changes in myeloid cell subsets. These data led to the hypothesis that S100A8/A9 proteins might play an active role in COVID-19 pathogenesis. This review explores the structures and functions of S100A8/9 and the current knowledge on the involvement of S100A8/A9 and its constituents in viral infections. The potential roles of S100A9 in SARS-CoV-2 infections are also discussed.
Collapse
Affiliation(s)
- Julien Boucher
- Axe de recherche sur les maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Caroline Gilbert
- Axe de recherche sur les maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Philippe A. Tessier
- Axe de recherche sur les maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
23
|
Nicolai L, Pekayvaz K, Massberg S. Platelets: Orchestrators of immunity in host defense and beyond. Immunity 2024; 57:957-972. [PMID: 38749398 DOI: 10.1016/j.immuni.2024.04.008] [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: 12/31/2023] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 06/05/2024]
Abstract
Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.
Collapse
Affiliation(s)
- Leo Nicolai
- 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.
| | - Kami Pekayvaz
- 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
| | - Steffen Massberg
- 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
|
24
|
Paramasivam S, Perumal SS, Ekambaram SP. Computational Deciphering of the Role of S100A8 and S100A9 Proteins and Their Changes in the Structure Assembly Influences Their Interaction with TLR4, RAGE, and CD36. Protein J 2024; 43:243-258. [PMID: 38431537 DOI: 10.1007/s10930-024-10186-0] [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] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
S100A8 and S100A9 belong to the calcium-binding, damage associated molecular pattern (DAMP) proteins shown to aggravate the pathogenesis of rheumatoid arthritis (RA) through their interaction with the TLR4, RAGE and CD36 receptors. S100A8 and S100A9 proteins tend to exist in monomeric, homo and heterodimeric forms, which have been implicated in the pathogenesis of RA, via interacting with Pattern Recognition receptors (PRRs). The study aims to assess the influence of changes in the structure and biological assembly of S100A8 and S100A9 proteins as well as their interaction with significant receptors in RA through computational methods and surface plasmon resonance (SPR) analysis. Molecular docking analysis revealed that the S100A9 homodimer and S100A8/A9 heterodimer showed higher binding affinity towards the target receptors. Most S100 proteins showed good binding affinity towards TLR4 compared to other receptors. Based on the 50 ns MD simulations, TLR4, RAGE, and CD36 formed stable complexes with the monomeric and dimeric forms of S100A8 and S100A9 proteins. However, SPR analysis showed that the S100A8/A9 heterodimers formed stable complexes and exhibited high binding affinity towards the receptors. SPR data also indicated that TLR4 and its interactions with S100A8/A9 proteins may play a primary role in the pathogenesis of RA, with additional contributions from CD36 and RAGE interactions. Subsequent in vitro and in vivo investigations are warranted to corroborate the involvement of S100A8/A9 and the expression of TLR4, RAGE, and CD36 in the pathophysiology of RA.
Collapse
Affiliation(s)
- Sivasakthi Paramasivam
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Senthamil Selvan Perumal
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Sanmuga Priya Ekambaram
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
25
|
Tu X, Chen L, Zheng Y, Mu C, Zhang Z, Wang F, Ren Y, Duan Y, Zhang H, Tong Z, Liu L, Sun X, Zhao P, Wang L, Feng X, Fang W, Liu X. S100A9 +CD14 + monocytes contribute to anti-PD-1 immunotherapy resistance in advanced hepatocellular carcinoma by attenuating T cell-mediated antitumor function. J Exp Clin Cancer Res 2024; 43:72. [PMID: 38454445 PMCID: PMC10921725 DOI: 10.1186/s13046-024-02985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/14/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The paucity of reliable biomarkers for predicting immunotherapy efficacy in patients with advanced hepatocellular carcinoma (HCC) has emerged as a burgeoning concern with the expanding use of immunotherapy. This study endeavors to delve into the potential peripheral biomarkers capable of prognosticating efficacy in HCC patients who are poised to receive anti-PD-1 monotherapy within the phase III clinical trial, KEYNOTE394. Additionally, we sought to elucidate the underlying molecular mechanisms for resistance to immune checkpoint blockade (ICB) and propose innovative combination immunotherapy strategies for future clinical application. METHODS Patient blood samples were collected for single-cell RNA sequencing to evaluate the immune cell signature before receiving ICB therapy. Subsequently, in vitro assays and in vivo murine model experiments were conducted to validate the mechanism that S100A9+CD14+ monocytes play a role in ICB resistance. RESULTS Our study demonstrates a notable enrichment of S100A9+CD14+ monocytes in the peripheral blood of patients exhibiting suboptimal responses to anti-PD-1 therapy. Moreover, we identified the Mono_S100A9 signature as a predictive biomarker, indicative of reduced efficacy in immunotherapy and decreased survival benefits across various tumor types. Mechanistically, S100A9 activates PD-L1 transcription by directly binding to the CD274 (PD-L1) gene promoter, thereby suppressing T-cell proliferation and cytotoxicity via the PD-1/PD-L1 axis, consequently diminishing the therapeutic effectiveness of subsequent anti-PD-1 treatments. Furthermore, our in vivo studies revealed that inhibiting S100A9 can synergistically enhance the efficacy of anti-PD-1 drugs in the eradication of hepatocellular carcinoma. CONCLUSIONS Our study underscores the significance of S100A9+CD14+ monocytes in predicting inadequate response to ICB treatment and provides insights into the monocyte cell-intrinsic mechanisms of resistance to ICB therapy. We also propose a combined therapeutic approach to enhance ICB efficacy by targeting S100A9.
Collapse
Affiliation(s)
- Xiaoxuan Tu
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, People's Republic of China
| | - Longxian Chen
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, People's Republic of China
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Yi Zheng
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Chenglin Mu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, People's Republic of China
| | - Zhiwei Zhang
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, People's Republic of China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Feiyu Wang
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, People's Republic of China
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Yiqing Ren
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Yingxin Duan
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Hangyu Zhang
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Zhou Tong
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Lulu Liu
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Xunqi Sun
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Peng Zhao
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Lie Wang
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 310058, People's Republic of China
| | - Xinhua Feng
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, People's Republic of China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, People's Republic of China.
| | - Weijia Fang
- Department of Medical Oncology, & Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China.
| | - Xia Liu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, People's Republic of China.
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
26
|
Vulliamy P, Armstrong PC. Platelets in Hemostasis, Thrombosis, and Inflammation After Major Trauma. Arterioscler Thromb Vasc Biol 2024; 44:545-557. [PMID: 38235557 DOI: 10.1161/atvbaha.123.318801] [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: 01/19/2024]
Abstract
Trauma currently accounts for 10% of the total global burden of disease and over 5 million deaths per year, making it a leading cause of morbidity and mortality worldwide. Although recent advances in early resuscitation have improved early survival from critical injury, the mortality rate in patients with major hemorrhage approaches 50% even in mature trauma systems. A major determinant of clinical outcomes from a major injury is a complex, dynamic hemostatic landscape. Critically injured patients frequently present to the emergency department with an acute traumatic coagulopathy that increases mortality from bleeding, yet, within 48 to 72 hours after injury will switch from a hypocoagulable to a hypercoagulable state with increased risk of venous thromboembolism and multiple organ dysfunction. This review will focus on the role of platelets in these processes. As effectors of hemostasis and thrombosis, they are central to each phase of recovery from injury, and our understanding of postinjury platelet biology has dramatically advanced over the past decade. This review describes our current knowledge of the changes in platelet behavior that occur following major trauma, the mechanisms by which these changes develop, and the implications for clinical outcomes. Importantly, supported by research in other disease settings, this review also reflects the emerging role of thromboinflammation in trauma including cross talk between platelets, innate immune cells, and coagulation. We also address the unresolved questions and significant knowledge gaps that remain, and finally highlight areas that with the further study will help deliver further improvements in trauma care.
Collapse
Affiliation(s)
- Paul Vulliamy
- Centre for Trauma Sciences (P.V.), Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Paul C Armstrong
- Centre for Immunobiology (P.C.A.), Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| |
Collapse
|
27
|
Jiang H, Zhao Y, Su M, Sun L, Chen M, Zhang Z, Ilyas I, Wang Z, Little PJ, Wang L, Weng J, Ge J, Xu S. A proteome-wide screen identifies the calcium binding proteins, S100A8/S100A9, as clinically relevant therapeutic targets in aortic dissection. Pharmacol Res 2024; 199:107029. [PMID: 38056513 DOI: 10.1016/j.phrs.2023.107029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/01/2023] [Accepted: 12/03/2023] [Indexed: 12/08/2023]
Abstract
Aortic dissection (AD) is a fatal cardiovascular disease with limited pharmacotherapies. To discover novel therapeutic targets for AD, the present study was conducted on ascending aorta samples from AD patients versus those from control subjects using proteomic analysis. Integrated proteomic data analysis identified S100 calcium-binding proteins A8 and A9 (S100A8/A9) as new therapeutic targets for AD. As assessed by ELISA, the circulating levels of S100A8/A9 were elevated in AD patients. In addition, we validated the upregulation of S100A8/A9 in a mouse model of AD. In vitro and in vivo studies substantiated that S100A8/A9, as danger-associated molecular pattern molecules, promotes the smooth muscle cells phenotypic switch by inhibiting serum response factor (SRF) activity but elevating NF-κB dependent inflammatory response. Depletion of S100A8/A9 attenuates the occurrence and development of AD. As a proof of concept, we tested the safety and efficacy of pharmacological inhibition of S100A8/A9 by ABR-25757 (paquinimod) in a mouse model of AD. We observed that ABR-25757 ameliorated the incidence of rupture and improved elastin morphology associated with AD. Further single-cell RNA sequencing disclosed that the phenotypic switch of vascular smooth muscle cells (VSMCs) and inflammatory response pathways were responsible for ABR-25757-mediated protection against AD. Thus, this study reveals the regulatory mechanism of S100A8/A9 in AD and offers a potential therapeutic avenue to treat AD by targeting S100A8/A9.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yaping Zhao
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Meiming Su
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Lu Sun
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Meijie Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Zhidan Zhang
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Iqra Ilyas
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Zhihua Wang
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, China
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| | - Jianjun Ge
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China.
| |
Collapse
|
28
|
Yong J, Toh CH. Rethinking coagulation: from enzymatic cascade and cell-based reactions to a convergent model involving innate immune activation. Blood 2023; 142:2133-2145. [PMID: 37890148 DOI: 10.1182/blood.2023021166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
ABSTRACT Advancements in the conceptual thinking of hemostasis and thrombosis have been catalyzed by major developments within health research over several decades. The cascade model of coagulation was first described in the 1960s, when biochemistry gained prominence through innovative experimentation and technical developments. This was followed by the cell-based model, which integrated cellular coordination to the enzymology of clot formation and was conceptualized during the growth period in cell biology at the turn of the millennium. Each step forward has heralded a revolution in clinical therapeutics, both in procoagulant and anticoagulant treatments to improve patient care. In current times, the COVID-19 pandemic may also prove to be a catalyst: thrombotic challenges including the mixed responses to anticoagulant treatment and the vaccine-induced immune thrombotic thrombocytopenia have exposed limitations in our preexisting concepts while simultaneously demanding novel therapeutic approaches. It is increasingly clear that innate immune activation as part of the host response to injury is not separate but integrated into adaptive clot formation. Our review summarizes current understanding of the major molecules facilitating such a cross talk between immunity, inflammation and coagulation. We demonstrate how such effects can be layered upon the cascade and cell-based models to evolve conceptual understanding of the physiology of immunohemostasis and the pathology of immunothrombosis.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
29
|
Panteleev MA, Sveshnikova AN, Shakhidzhanov SS, Zamaraev AV, Ataullakhanov FI, Rumyantsev AG. The Ways of the Virus: Interactions of Platelets and Red Blood Cells with SARS-CoV-2, and Their Potential Pathophysiological Significance in COVID-19. Int J Mol Sci 2023; 24:17291. [PMID: 38139118 PMCID: PMC10743882 DOI: 10.3390/ijms242417291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The hematological effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are important in COVID-19 pathophysiology. However, the interactions of SARS-CoV-2 with platelets and red blood cells are still poorly understood. There are conflicting data regarding the mechanisms and significance of these interactions. The aim of this review is to put together available data and discuss hypotheses, the known and suspected effects of the virus on these blood cells, their pathophysiological and diagnostic significance, and the potential role of platelets and red blood cells in the virus's transport, propagation, and clearance by the immune system. We pay particular attention to the mutual activation of platelets, the immune system, the endothelium, and blood coagulation and how this changes with the evolution of SARS-CoV-2. There is now convincing evidence that platelets, along with platelet and erythroid precursors (but not mature erythrocytes), are frequently infected by SARS-CoV-2 and functionally changed. The mechanisms of infection of these cells and their role are not yet entirely clear. Still, the changes in platelets and red blood cells in COVID-19 are significantly associated with disease severity and are likely to have prognostic and pathophysiological significance in the development of thrombotic and pulmonary complications.
Collapse
Affiliation(s)
- Mikhail A. Panteleev
- Department of Medical Physics, Physics Faculty, Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, 1 Samory Mashela, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya Str., 109029 Moscow, Russia
| | - Anastasia N. Sveshnikova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, 1 Samory Mashela, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya Str., 109029 Moscow, Russia
- Faculty of Fundamental Physics and Chemical Engineering, Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia
| | - Soslan S. Shakhidzhanov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, 1 Samory Mashela, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya Str., 109029 Moscow, Russia
| | - Alexey V. Zamaraev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Ulitsa Vavilova, 119991 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 1 Leninskie Gory, 119991 Moscow, Russia
| | - Fazoil I. Ataullakhanov
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, 1 Samory Mashela, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya Str., 109029 Moscow, Russia
- Moscow Institute of Physics and Technology, National Research University, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA 19104, USA
| | - Aleksandr G. Rumyantsev
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, 1 Samory Mashela, 117198 Moscow, Russia
| |
Collapse
|
30
|
Zhao Y, Qi W, Huang C, Zhou Y, Wang Q, Tian X, Li M, Zhao Y, Zeng X, Zhao J. Serum Calprotectin as a Potential Predictor of Microvascular Manifestations in Patients with Antiphospholipid Syndrome. Rheumatol Ther 2023; 10:1769-1783. [PMID: 37906398 PMCID: PMC10654303 DOI: 10.1007/s40744-023-00610-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023] Open
Abstract
INTRODUCTION Microvascular manifestations constitute a subtype of antiphospholipid syndrome, and those patients have relatively poor prognoses, so it is important to find markers for microvascular manifestations. This study was conducted to explore whether serum calprotectin could be a predictor of microvascular manifestations in antiphospholipid antibody (aPL)-positive patients. METHODS Consecutive patients with persistent aPL positivity referred to Peking Union Medical College Hospital and age- and sex-matched health controls (HCs) were included. Microvascular manifestations included antiphospholipid syndrome (APS) nephropathy, livedo reticularis, valvular lesions, non-stroke central nervous system manifestations, myocarditis, catastrophic APS, and other microvascular manifestations confirmed by pathology, imaging, or clinical diagnosis. Calprotectin was measured by an enzyme-linked immunosorbent assay (ELISA). The cutoff value was defined as mean + 2 standard deviations of HCs. Multivariable logistic regression analysis was used to analyze risk factors. Pearson correlation analysis was used to detect the correlation between calprotectin and other laboratory data. RESULTS Of the 466 patients included in the study, 281 (60.3%) patients met the 2006 Sydney Revised Classification Criteria; among the latter, 77.2% were patients with primary APS. The mean age was 39.10 ± 13.05 years old, and 77.0% were female. Thirty-eight age- and sex-matched HCs were included in the study. Serum calprotectin levels were increased in aPL-positive patients compared with HCs (649.66 ± 240.79 vs 484.62 ± 149.37 ng/ml, p < 0.001), and were increased in patients with microvascular manifestations compared with patients without (693.03 ± 271.90 vs 639.43 ± 232.06 ng/ml, p = 0.044). The cutoff value was 783.36 ng/ml. Ninety-three patients (20.0%) were positive for calprotectin. Calprotectin positivity was independently associated with microvascular manifestations (odds ratio [OR] 1.90, 95% confidence interval [CI] 1.07-3.36) and platelet count (PLT) < 100 (OR 2.04, 95% CI 1.08-3.88). Age (OR 0.98, 95% CI 0.96-1.00), systemic lupus erythematosus (OR 2.08, 95% CI 1.15-3.75), calprotectin positivity (OR 1.83, 95% CI 1.02-3.26), hypertension (OR 2.73, 95% CI 1.36-5.45), hemolytic anemia (OR 2.66, 95% CI 1.13-6.23), and anti-β2GPI antibodies (OR 2.06, 95% CI 1.11-3.83) could independently predict microvascular manifestations in aPL-positive patients. Serum calprotectin negatively correlated with PLT (R = - 0.101, p = 0.031). CONCLUSION Serum calprotectin levels are increased in aPL-positive patients and could be a potential predictor of microvascular manifestations.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wanting Qi
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Can Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yangzhong Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China.
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| |
Collapse
|
31
|
Abstract
Proteomics tools provide a powerful means to identify, detect, and quantify protein-related details in studies of platelet phenotype and function. Here, we consider how historical and recent advances in proteomics approaches have informed our understanding of platelet biology, and, how proteomics tools can be used going forward to advance studies of platelets. It is now apparent that the platelet proteome is comprised of thousands of different proteins, where specific changes in platelet protein systems can accompany alterations in platelet function in health and disease. Going forward, many challenges remain in how to best carry out, validate and interpret platelet proteomics experiments. Future studies of platelet protein post-translational modifications such as glycosylation, or studies that take advantage of single cell proteomics and top-down proteomics methods all represent areas of interest to profiling and more richly understanding platelets in human wellness and disease.
Collapse
Affiliation(s)
- Joseph E. Aslan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
32
|
Wang Q, Long G, Luo H, Zhu X, Han Y, Shang Y, Zhang D, Gong R. S100A8/A9: An emerging player in sepsis and sepsis-induced organ injury. Biomed Pharmacother 2023; 168:115674. [PMID: 37812889 DOI: 10.1016/j.biopha.2023.115674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/11/2023] Open
Abstract
Sepsis, the foremost contributor to mortality in intensive care unit patients, arises from an uncontrolled systemic response to invading infections, resulting in extensive harm across multiple organs and systems. Recently, S100A8/A9 has emerged as a promising biomarker for sepsis and sepsis-induced organ injury, and targeting S100A8/A9 appeared to ameliorate inflammation-induced tissue damage and improve adverse outcomes. S100A8/A9, a calcium-binding heterodimer mainly found in neutrophils and monocytes, serves as a causative molecule with pro-inflammatory and immunosuppressive properties, which are vital in the pathogenesis of sepsis. Therefore, improving our comprehension of how S100A8/A9 acts as a pathological player in the development of sepsis is imperative for advancing research on sepsis. Our review is the first-to the best of our knowledge-to discuss the biology of S100A8/A9 and its release mechanisms, summarize recent advances concerning the vital roles of S100A8/A9 in sepsis and the consequential organ damage, and underscore its potential as a promising diagnostic biomarker and therapeutic target for sepsis.
Collapse
Affiliation(s)
- Qian Wang
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China
| | - Gangyu Long
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China
| | - Hong Luo
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China
| | - Xiqun Zhu
- Hubei Cancer Hospital, Tongji Medical College, HUST, Wuhan 430079, China
| | - Yang Han
- Center for Translational Medicine, Wuhan Jinyintan Hospital, Wuhan 430023, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan 430030, China.
| | - Dingyu Zhang
- Wuhan Jinyintan Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430023, China; Hubei Clinical Research Center for Infectious Diseases, Wuhan 430023, China; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan 430023, China; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan 430023, China.
| | - Rui Gong
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
33
|
Sun S, Campello E, Zou J, Konings J, Huskens D, Wan J, Fernández DI, Reutelingsperger CPM, ten Cate H, Toffanin S, Bulato C, de Groot PG, de Laat B, Simioni P, Heemskerk JWM, Roest M. Crucial roles of red blood cells and platelets in whole blood thrombin generation. Blood Adv 2023; 7:6717-6731. [PMID: 37648671 PMCID: PMC10651426 DOI: 10.1182/bloodadvances.2023010027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
Red blood cells (RBCs) and platelets contribute to the coagulation capacity in bleeding and thrombotic disorders. The thrombin generation (TG) process is considered to reflect the interactions between plasma coagulation and the various blood cells. Using a new high-throughput method capturing the complete TG curve, we were able to compare TG in whole blood and autologous platelet-rich and platelet-poor plasma to redefine the blood cell contributions to the clotting process. We report a faster and initially higher generation of thrombin and shorter coagulation time in whole blood than in platelet-rich plasma upon low concentrations of coagulant triggers, including tissue factor, Russell viper venom factor X, factor Xa, factor XIa, and thrombin. The TG was accelerated with increased hematocrit and delayed after prior treatment of RBC with phosphatidylserine-blocking annexin A5. RBC treatment with ionomycin increased phosphatidylserine exposure, confirmed by flow cytometry, and increased the TG process. In reconstituted blood samples, the prior selective blockage of phosphatidylserine on RBC with annexin A5 enhanced glycoprotein VI-induced platelet procoagulant activity. For patients with anemia or erythrocytosis, cluster analysis revealed high or low whole-blood TG profiles in specific cases of anemia. The TG profiles lowered upon annexin A5 addition in the presence of RBCs and thus were determined by the extent of phosphatidylserine exposure of blood cells. Profiles for patients with polycythemia vera undergoing treatment were similar to that of control subjects. We concluded that RBC and platelets, in a phosphatidylserine-dependent way, contribute to the TG process. Determination of the whole-blood hypo- or hyper-coagulant activity may help to characterize a bleeding or thrombosis risk.
Collapse
Affiliation(s)
- Siyu Sun
- Synapse Research Institute, Maastricht, The Netherlands
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Elena Campello
- Department of Medicine, University of Padua, Padova, Italy
| | - Jinmi Zou
- Synapse Research Institute, Maastricht, The Netherlands
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Joke Konings
- Synapse Research Institute, Maastricht, The Netherlands
| | - Dana Huskens
- Synapse Research Institute, Maastricht, The Netherlands
| | - Jun Wan
- Synapse Research Institute, Maastricht, The Netherlands
| | - Delia I. Fernández
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Chris P. M. Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Hugo ten Cate
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
- Department of Internal Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | | | | | - Bas de Laat
- Synapse Research Institute, Maastricht, The Netherlands
| | - Paolo Simioni
- Department of Medicine, University of Padua, Padova, Italy
| | - Johan W. M. Heemskerk
- Synapse Research Institute, Maastricht, The Netherlands
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Mark Roest
- Synapse Research Institute, Maastricht, The Netherlands
| |
Collapse
|
34
|
Maiorca F, Lombardi L, Marrapodi R, Pallucci D, Sabetta A, Zingaropoli MA, Perri V, Flego D, Romiti GF, Corica B, Miglionico M, Russo G, Pasculli P, Ciardi MR, Mastroianni CM, Ruberto F, Pugliese F, Pulcinelli F, Raparelli V, Cangemi R, Visentini M, Basili S, Stefanini L. Breakthrough infections after COVID-19 vaccinations do not elicit platelet hyperactivation and are associated with high platelet-lymphocyte and low platelet-neutrophil aggregates. Res Pract Thromb Haemost 2023; 7:102262. [PMID: 38193050 PMCID: PMC10772876 DOI: 10.1016/j.rpth.2023.102262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 01/10/2024] Open
Abstract
Background Severe COVID-19 is associated with an excessive immunothrombotic response and thromboinflammatory complications. Vaccinations effectively reduce the risk of severe clinical outcomes in patients with COVID-19, but their impact on platelet activation and immunothrombosis during breakthrough infections is not known. Objectives To investigate how preemptive vaccinations modify the platelet-immune crosstalk during COVID-19 infections. Methods Cross-sectional flow cytometry study of the phenotype and interactions of platelets circulating in vaccinated (n = 21) and unvaccinated patients with COVID-19, either admitted to the intensive care unit (ICU, n = 36) or not (non-ICU, n = 38), in comparison to matched SARS-CoV-2-negative patients (n = 48), was performed. Results In the circulation of unvaccinated non-ICU patients with COVID-19, we detected hyperactive and hyperresponsive platelets and platelet aggregates with adaptive and innate immune cells. In unvaccinated ICU patients with COVID-19, most of whom had severe acute respiratory distress syndrome, platelets had high P-selectin and phosphatidylserine exposure but low capacity to activate integrin αIIbβ3, dysfunctional mitochondria, and reduced surface glycoproteins. In addition, in the circulation of ICU patients, we detected microthrombi and platelet aggregates with innate, but not with adaptive, immune cells. In vaccinated patients with COVID-19, who had no acute respiratory distress syndrome, platelets had surface receptor levels comparable to those in controls and did not form microthrombi or platelet-granulocyte aggregates but aggregated avidly with adaptive immune cells. Conclusion Our study provides evidence that vaccinated patients with COVID-19 are not associated with platelet hyperactivation and are characterized by platelet-leukocyte aggregates that foster immune protection but not excessive immunothrombosis. These findings advocate for the importance of vaccination in preventing severe COVID-19.
Collapse
Affiliation(s)
- Francesca Maiorca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ludovica Lombardi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ramona Marrapodi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Davide Pallucci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Annamaria Sabetta
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Valentina Perri
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Davide Flego
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giulio Francesco Romiti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Bernadette Corica
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Miglionico
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Gianluca Russo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Patrizia Pasculli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Claudio M. Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Franco Ruberto
- Department of Specialist Surgery and Organ Transplantation “Paride Stefanini,” Sapienza University of Rome, Rome, Italy
| | - Francesco Pugliese
- Department of Specialist Surgery and Organ Transplantation “Paride Stefanini,” Sapienza University of Rome, Rome, Italy
| | - Fabio Pulcinelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Valeria Raparelli
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Faculty of Nursing, University of Alberta, Edmonton, Alberta, Canada
- University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Roberto Cangemi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marcella Visentini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
35
|
Kaiser R, Escaig R, Nicolai L. Hemostasis without clot formation: how platelets guard the vasculature in inflammation, infection, and malignancy. Blood 2023; 142:1413-1425. [PMID: 37683182 DOI: 10.1182/blood.2023020535] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Platelets are key vascular effectors in hemostasis, with activation signals leading to fast recruitment, aggregation, and clot formation. The canonical process of hemostasis is well-characterized and shares many similarities with pathological thrombus formation. However, platelets are also crucially involved in the maintenance of vascular integrity under both steady-state and inflammatory conditions by ensuring blood vessel homeostasis and preventing microbleeds. In these settings, platelets use distinct receptors, signaling pathways, and ensuing effector functions to carry out their deeds. Instead of simply forming clots, they mainly act as individual sentinels that swiftly adapt their behavior to the local microenvironment. In this review, we summarize previously recognized and more recent studies that have elucidated how anucleate, small platelets manage to maintain vascular integrity when faced with challenges of infection, sterile inflammation, and even malignancy. We dissect how platelets are recruited to the vascular wall, how they identify sites of injury, and how they prevent hemorrhage as single cells. Furthermore, we discuss mechanisms and consequences of platelets' interaction with leukocytes and endothelial cells, the relevance of adhesion as well as signaling receptors, in particular immunoreceptor tyrosine-based activation motif receptors, and cross talk with the coagulation system. Finally, we outline how recent insights into inflammatory hemostasis and vascular integrity may aid in the development of novel therapeutic strategies to prevent hemorrhagic events and vascular dysfunction in patients who are critically ill.
Collapse
Affiliation(s)
- Rainer Kaiser
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig Maximilian University, Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Raphael Escaig
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig Maximilian University, Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig Maximilian University, Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
36
|
Whyte CS, Morrow GB, Gauer JS, Montague SJ, Nicolson PLR. Novel therapeutics and emerging technology in haemostasis and thrombosis: highlights from the British society for haemostasis and thrombosis annual meeting. Front Cardiovasc Med 2023; 10:1225243. [PMID: 37745127 PMCID: PMC10512947 DOI: 10.3389/fcvm.2023.1225243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
The 2023 annual meeting of the British Society for Haemostasis and Thrombosis (BSHT) was held in Birmingham, United Kingdom. The theme of this year's meeting was novel therapeutics and emerging technology. Here, the exciting research presented at the meeting is discussed.
Collapse
Affiliation(s)
- Claire S. Whyte
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gael B. Morrow
- Aberdeen Cardiovascular and Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julia S. Gauer
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Samantha J. Montague
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Philip L. R. Nicolson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- West Midlands Haemophilia Comprehensive Care Centre, University Hospitals Birmingham Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
37
|
Zhou H, Zhao C, Shao R, Xu Y, Zhao W. The functions and regulatory pathways of S100A8/A9 and its receptors in cancers. Front Pharmacol 2023; 14:1187741. [PMID: 37701037 PMCID: PMC10493297 DOI: 10.3389/fphar.2023.1187741] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Inflammation primarily influences the initiation, progression, and deterioration of many human diseases, and immune cells are the principal forces that modulate the balance of inflammation by generating cytokines and chemokines to maintain physiological homeostasis or accelerate disease development. S100A8/A9, a heterodimer protein mainly generated by neutrophils, triggers many signal transduction pathways to mediate microtubule constitution and pathogen defense, as well as intricate procedures of cancer growth, metastasis, drug resistance, and prognosis. Its paired receptors, such as receptor for advanced glycation ends (RAGEs) and toll-like receptor 4 (TLR4), also have roles and effects within tumor cells, mainly involved with mitogen-activated protein kinases (MAPKs), NF-κB, phosphoinositide 3-kinase (PI3K)/Akt, mammalian target of rapamycin (mTOR) and protein kinase C (PKC) activation. In the clinical setting, S100A8/A9 and its receptors can be used complementarily as efficient biomarkers for cancer diagnosis and treatment. This review comprehensively summarizes the biological functions of S100A8/A9 and its various receptors in tumor cells, in order to provide new insights and strategies targeting S100A8/A9 to promote novel diagnostic and therapeutic methods in cancers.
Collapse
Affiliation(s)
- Huimin Zhou
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Zhao
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rongguang Shao
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanni Xu
- NHC Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wuli Zhao
- State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Scherlinger M, Richez C, Tsokos GC, Boilard E, Blanco P. The role of platelets in immune-mediated inflammatory diseases. Nat Rev Immunol 2023; 23:495-510. [PMID: 36707719 PMCID: PMC9882748 DOI: 10.1038/s41577-023-00834-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2022] [Indexed: 01/28/2023]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are characterized by excessive and uncontrolled inflammation and thrombosis, both of which are responsible for organ damage, morbidity and death. Platelets have long been known for their role in primary haemostasis, but they are now also considered to be components of the immune system and to have a central role in the pathogenesis of IMIDs. In patients with IMIDs, platelets are activated by disease-specific factors, and their activation often reflects disease activity. Here we summarize the evidence showing that activated platelets have an active role in the pathogenesis and the progression of IMIDs. Activated platelets produce soluble factors and directly interact with immune cells, thereby promoting an inflammatory phenotype. Furthermore, platelets participate in tissue injury and promote abnormal tissue healing, leading to fibrosis. Targeting platelet activation and targeting the interaction of platelets with the immune system are novel and promising therapeutic strategies in IMIDs.
Collapse
Affiliation(s)
- Marc Scherlinger
- Service de Rhumatologie, Centre de référence des maladies auto-immunes systémiques rares RESO, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Laboratoire d'ImmunoRhumatologie Moléculaire UMR_S 1109, Institut National de la Santé et de la Recherche Médicale (INSERM), Strasbourg, France.
| | - Christophe Richez
- Service de Rhumatologie, Centre de référence des maladies auto-immunes systémiques rares RESO, Hôpital Pellegrin, Centre Hospitalier Universitaire, Bordeaux, France
- CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, Bordeaux, France
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, Quebec, Canada
- Centre de Recherche ARThrite, Université Laval, Quebec City, Quebec, Canada
| | - Patrick Blanco
- CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, Bordeaux, France.
- Laboratoire d'Immunologie et Immunogénétique, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Bordeaux, France.
| |
Collapse
|
39
|
Zaid Y, Khalki L, Jalali F, Tijani Y, Zaid N, Naya A, Oudghiri M, Akarid K, Agbani EO, Guessous F. Low α-Thrombin/GPIbα Interaction Is a Potential Contributor to Platelet Hyper-reactivity in COVID-19 Patients. Thromb Haemost 2023; 123:804-807. [PMID: 37044132 PMCID: PMC10365884 DOI: 10.1055/a-2072-0366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/14/2023] [Indexed: 04/14/2023]
Affiliation(s)
- Younes Zaid
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
- Research Center of Abulcasis University of Health Sciences, Rabat, Morocco
- Immunology and Biodiversity Laboratory, Department of Biology, Ain Chock Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Loubna Khalki
- Laboratory of Neurosciences and Oncogenetics, Neurosciences and Cellular Physiology Team, Mohammed VI Center for Research & Innovation, Higher Institute of Biosciences and Biotechnology and Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Farid Jalali
- Department of Gastroenterology, Saddleback Medical Group, Laguna Hills, California, United States
| | - Youssef Tijani
- Laboratory of Neurosciences and Oncogenetics, Neurosciences and Cellular Physiology Team, Mohammed VI Center for Research & Innovation, Higher Institute of Biosciences and Biotechnology and Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Nabil Zaid
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
- Health, Care, and Environment Laboratory, Higher Institute of Nursing Professions and Health Technology of Rabat, Morocco
| | - Abdallah Naya
- Immunology and Biodiversity Laboratory, Department of Biology, Ain Chock Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Mounia Oudghiri
- Immunology and Biodiversity Laboratory, Department of Biology, Ain Chock Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - Khadija Akarid
- Health and Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca, Morocco
| | - Ejaife O. Agbani
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada
| | - Fadila Guessous
- Laboratory of Neurosciences and Oncogenetics, Neurosciences and Cellular Physiology Team, Mohammed VI Center for Research & Innovation, Higher Institute of Biosciences and Biotechnology and Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Department of Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
40
|
Hirsch J, Uzun G, Zlamal J, Singh A, Bakchoul T. Platelet-neutrophil interaction in COVID-19 and vaccine-induced thrombotic thrombocytopenia. Front Immunol 2023; 14:1186000. [PMID: 37275917 PMCID: PMC10237318 DOI: 10.3389/fimmu.2023.1186000] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/04/2023] [Indexed: 06/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is known to commonly induce a thrombotic diathesis, particularly in severely affected individuals. So far, this COVID-19-associated coagulopathy (CAC) has been partially explained by hyperactivated platelets as well as by the prothrombotic effects of neutrophil extracellular traps (NETs) released from neutrophils. However, precise insight into the bidirectional relationship between platelets and neutrophils in the pathophysiology of CAC still lags behind. Vaccine-induced thrombotic thrombocytopenia (VITT) is a rare autoimmune disorder caused by auto-antibody formation in response to immunization with adenoviral vector vaccines. VITT is associated with life-threatening thromboembolic events and thus, high fatality rates. Our concept of the thrombophilia observed in VITT is relatively new, hence a better understanding could help in the management of such patients with the potential to also prevent VITT. In this review we aim to summarize the current knowledge on platelet-neutrophil interplay in COVID-19 and VITT.
Collapse
Affiliation(s)
- Johannes Hirsch
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Günalp Uzun
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Jan Zlamal
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Anurag Singh
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Tamam Bakchoul
- Institute of Clinical and Experimental Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
- Center for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| |
Collapse
|
41
|
Abstract
COVID-19 is characterized by dysregulated thrombosis and coagulation that can increase mortality in patients. Platelets are fast responders to pathogen presence, alerting the surrounding immune cells and contributing to thrombosis and intravascular coagulation. The SARS-CoV-2 genome has been found in platelets from patients with COVID-19, and its coverage varies according to the method of detection, suggesting direct interaction of the virus with these cells. Antibodies against Spike and Nucleocapsid have confirmed this platelet-viral interaction. This review discusses the immune, prothrombotic, and procoagulant characteristics of platelets observed in patients with COVID-19. We outline the direct and indirect interaction of platelets with SARS-CoV-2, the contribution of the virus to programmed cell death pathway activation in platelets and the consequent extracellular vesicle release. We discuss platelet activation and immunothrombosis in patients with COVID-19, the effect of Spike on platelets, and possible activation of platelets by classical platelet activation triggers as well as contribution of platelets to complement activation. As COVID-19-mediated thrombosis and coagulation are still not well understood in vivo, we discuss available murine models and mouse adaptable strains.
Collapse
Affiliation(s)
- Anthony Sciaudone
- Department of Medicine, Divisions of Cardiovascular Medicine (A.S., H.C., M.K.), University of Massachusetts Chan Medical School, Worcester, MA
| | - Heather Corkrey
- Department of Medicine, Divisions of Cardiovascular Medicine (A.S., H.C., M.K.), University of Massachusetts Chan Medical School, Worcester, MA
| | - Fiachra Humphries
- Innate Immunity (F.H.). University of Massachusetts Chan Medical School, Worcester, MA
| | - Milka Koupenova
- Department of Medicine, Divisions of Cardiovascular Medicine (A.S., H.C., M.K.), University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|
42
|
Baksamawi HA, Alexiadis A, Vigolo D, Brill A. Platelet accumulation in an endothelium-coated elastic vein valve model of deep vein thrombosis is mediated by GPIb α-VWF interaction. Front Cardiovasc Med 2023; 10:1167884. [PMID: 37180784 PMCID: PMC10174463 DOI: 10.3389/fcvm.2023.1167884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/28/2023] [Indexed: 05/16/2023] Open
Abstract
Deep vein thrombosis is a life-threatening disease that takes millions of people's lives worldwide. Given both technical and ethical issues of using animals in research, it is necessary to develop an appropriate in vitro model that would recapitulate the conditions of venous thrombus development. We present here a novel microfluidics vein-on-a-chip with moving valve leaflets to mimic the hydrodynamics in a vein, and Human Umbilical Vein Endothelial Cell (HUVEC) monolayer. A pulsatile flow pattern, typical for veins, was used in the experiments. Unstimulated human platelets, reconstituted with the whole blood, accumulated at the luminal side of the leaflet tips proportionally to the leaflet flexibility. Platelet activation by thrombin induced robust platelet accrual at the leaflet tips. Inhibition of glycoprotein (GP) IIb-IIIa did not decrease but, paradoxically, slightly increased platelet accumulation. In contrast, blockade of the interaction between platelet GPIbα and A1 domain of von Willebrand factor completely abolished platelet deposition. Stimulation of the endothelium with histamine, a known secretagogue of Weibel-Palade bodies, promoted platelet accrual at the basal side of the leaflets, where human thrombi are usually observed. Thus, platelet deposition depends on the leaflet flexibility, and accumulation of activated platelets at the valve leaflets is mediated by GPIbα-VWF interaction.
Collapse
Affiliation(s)
- Hosam Alden Baksamawi
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Alessio Alexiadis
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Daniele Vigolo
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, Australia
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
43
|
Vinaiphat A, Pazhanchamy K, JebaMercy G, Ngan SC, Leow MKS, Ho HH, Gao YG, Lim KL, Richards AM, de Kleijn DPV, Chen CP, Kalaria RN, Liu J, O'Leary DD, McCarthy NE, Sze SK. Endothelial Damage Arising From High Salt Hypertension Is Elucidated by Vascular Bed Systematic Profiling. Arterioscler Thromb Vasc Biol 2023; 43:427-442. [PMID: 36700429 DOI: 10.1161/atvbaha.122.318439] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Considerable evidence links dietary salt intake with the development of hypertension, left ventricular hypertrophy, and increased risk of stroke and coronary heart disease. Despite extensive epidemiological and basic science interrogation of the relationship between high salt (HS) intake and blood pressure, it remains unclear how HS impacts endothelial cell (EC) and vascular structure in vivo. This study aims to elucidate HS-induced vascular pathology using a differential systemic decellularization in vivo approach. METHODS We performed systematic molecular characterization of the endothelial glycocalyx and EC proteomes in mice with HS (8%) diet-induced hypertension versus healthy control animals. Isolation of eGC and EC compartments was achieved using differential systemic decellularization in vivo methodology. Altered protein expression in hypertensive compared to normal mice was characterized by liquid chromatography tandem mass spectrometry. Proteomic results were validated using functional assays, microscopic imaging, and histopathologic evaluation. RESULTS Proteomic analysis revealed a significant downregulation of eGC and associated proteins in HS diet-induced hypertensive mice (among 1696 proteins identified in this group, 723 were markedly decreased in abundance, while only 168 were increased in abundance. Bioinformatic analysis indicated substantial derangement of the eGC layer, which was subsequently confirmed by fluorescent and electron microscopy assessment of vessel damage ex vivo. In the EC fraction, HS-induced hypertension significantly altered protein mediators of contractility, metabolism, mechanotransduction, renal function, and the coagulation cascade. In particular, we observed dysregulation of integrin subunits α2, α2b, and α5, which was associated with arterial wall inflammation and substantial infiltration of CD68+ monocyte-macrophages. Consequently, HS-induced hypertensive mice also displayed reduced vascular integrity of multiple organs including lungs, kidneys, and heart. CONCLUSIONS These findings provide novel molecular insight into HS-induced structural changes in eGC and EC composition that may increase cardiovascular risk and potentially guide the development of new diagnostics and therapeutic interventions.
Collapse
Affiliation(s)
- Arada Vinaiphat
- School of Biological Sciences (A.V., K.P., G.J., S.C.N., Y.-G.G., S.K.S.), Nanyang Technological University, Singapore
| | - Kalailingam Pazhanchamy
- School of Biological Sciences (A.V., K.P., G.J., S.C.N., Y.-G.G., S.K.S.), Nanyang Technological University, Singapore
| | - Gnanasekaran JebaMercy
- School of Biological Sciences (A.V., K.P., G.J., S.C.N., Y.-G.G., S.K.S.), Nanyang Technological University, Singapore
| | - SoFong Cam Ngan
- School of Biological Sciences (A.V., K.P., G.J., S.C.N., Y.-G.G., S.K.S.), Nanyang Technological University, Singapore
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada (S.C.N., J.L., D.D.O., S.K.S.)
| | - Melvin Khee-Shing Leow
- Lee Kong Chian School of Medicine (M.K.-S.L., K.L.L.), Nanyang Technological University, Singapore
- Tan Tock Seng Hospital, Singapore (M.K.-S.L., H.H.H.)
| | - Hee Hwa Ho
- Tan Tock Seng Hospital, Singapore (M.K.-S.L., H.H.H.)
| | - Yong-Gui Gao
- School of Biological Sciences (A.V., K.P., G.J., S.C.N., Y.-G.G., S.K.S.), Nanyang Technological University, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine (M.K.-S.L., K.L.L.), Nanyang Technological University, Singapore
| | - A Mark Richards
- Department of Cardiology, National University Heart Centre, Singapore (A.M.R.)
- Department of Cardiology, University of Otago, Christchurch, New Zealand (A.M.R.)
| | | | - Christopher P Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.P.C.)
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom (R.N.K.)
| | - Jian Liu
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada (S.C.N., J.L., D.D.O., S.K.S.)
| | - Deborah D O'Leary
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada (S.C.N., J.L., D.D.O., S.K.S.)
| | - Neil E McCarthy
- Centre for Immunobiology, The Blizard Institute, Bart's and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (N.E.M.)
| | - Siu Kwan Sze
- School of Biological Sciences (A.V., K.P., G.J., S.C.N., Y.-G.G., S.K.S.), Nanyang Technological University, Singapore
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada (S.C.N., J.L., D.D.O., S.K.S.)
| |
Collapse
|
44
|
Colicchia M, Perrella G, Gant P, Rayes J. Novel mechanisms of thrombo-inflammation during infection: spotlight on neutrophil extracellular trap-mediated platelet activation. Res Pract Thromb Haemost 2023; 7:100116. [PMID: 37063765 PMCID: PMC10099327 DOI: 10.1016/j.rpth.2023.100116] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 03/13/2023] Open
Abstract
A state-of-the-art lecture titled "novel mechanisms of thrombo-inflammation during infection" was presented at the ISTH Congress in 2022. Platelet, neutrophil, and endothelial cell activation coordinate the development, progression, and resolution of thrombo-inflammatory events during infection. Activated platelets and neutrophil extracellular traps (NETs) are frequently observed in patients with sepsis and COVID-19, and high levels of NET-derived damage-associated molecular patterns (DAMPs) correlate with thrombotic complications. NET-associated DAMPs induce direct and indirect platelet activation, which in return potentiates neutrophil activation and NET formation. These coordinated interactions involve multiple receptors and signaling pathways contributing to vascular and organ damage exacerbating disease severity. This state-of-the-art review describes the main mechanisms by which platelets support NETosis and the key mechanisms by which NET-derived DAMPs trigger platelet activation and the formation of procoagulant platelets leading to thrombosis. We report how these DAMPs act through multiple receptors and signaling pathways differentially regulating cell activation and disease outcome, focusing on histones and S100A8/A9 and their contribution to the pathogenesis of sepsis and COVID-19. We further discuss the complexity of platelet activation during NETosis and the potential benefit of targeting selective or multiple NET-associated DAMPs to limit thrombo-inflammation during infection. Finally, we summarize relevant new data on this topic presented during the 2022 ISTH Congress.
Collapse
Affiliation(s)
- Martina Colicchia
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
| | - Poppy Gant
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Birmingham, U.K
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, U.K
| |
Collapse
|
45
|
Platelet-Neutrophil Crosstalk in Thrombosis. Int J Mol Sci 2023; 24:ijms24021266. [PMID: 36674781 PMCID: PMC9861587 DOI: 10.3390/ijms24021266] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Platelets are essential for the formation of a haemostatic plug to prevent bleeding, while neutrophils are the guardians of our immune defences against invading pathogens. The interplay between platelets and innate immunity, and subsequent triggering of the activation of coagulation is part of the host system to prevent systemic spread of pathogen in the blood stream. Aberrant immunothrombosis and excessive inflammation can however, contribute to the thrombotic burden observed in many cardiovascular diseases. In this review, we highlight how platelets and neutrophils interact with each other and how their crosstalk is central to both arterial and venous thrombosis and in COVID-19. While targeting platelets and coagulation enables efficient antithrombotic treatments, they are often accompanied with a bleeding risk. We also discuss how novel approaches to reduce platelet-mediated recruitment of neutrophils could represent promising therapies to treat thrombosis without affecting haemostasis.
Collapse
|
46
|
Role of S100A8/A9 in Platelet-Neutrophil Complex Formation during Acute Inflammation. Cells 2022; 11:cells11233944. [PMID: 36497202 PMCID: PMC9738100 DOI: 10.3390/cells11233944] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) due to pulmonary infections is associated with high morbidity and mortality. Upon inflammation, the alarmin S100A8/A9 is released and stimulates neutrophil recruitment mainly via binding to Toll-like receptor 4 (TLR4). TLR4 is also expressed on platelets, which modulate the immune response through direct interaction with leukocytes. In a murine model of Klebsiella pneumoniae-induced pulmonary inflammation, global S100A9 deficiency resulted in diminished neutrophil recruitment into the lung alveoli and neutrophil accumulation in the intravascular space, indicating an impaired neutrophil migration. A lack of TLR4 on platelets resulted in reduced neutrophil counts in the whole lung, emphasising the impact of TLR4-mediated platelet activity on neutrophil behaviour. Flow cytometry-based analysis indicated elevated numbers of platelet-neutrophil complexes in the blood of S100A9-/- mice. Intravital microscopy of the murine cremaster muscle confirmed these findings and further indicated a significant increase in neutrophil-platelet complex formation in S100A9-/- mice, which was reversed by administration of the S100A8/A9 tetramer. An in vitro bilayer assay simulated the murine alveolar capillary barrier during inflammation and validated significant differences in transmigration behaviour between wild-type and S100A9-/- neutrophils. This study demonstrates the role of S100A8/A9 during platelet-neutrophil interactions and neutrophil recruitment during pulmonary inflammation.
Collapse
|