1
|
Lu X, Tang L, Wu Y, Hilton T, Chen X, Houck K, Fulton S, Han C, Romero S, Wang Y, Kozar R, Cardenas JC, Fu X, Zhang J, Zhao Z, Dong JF. Metabolically aberrant extracellular mitochondria induce oxidative endotheliopathy in traumatic brain injury. Blood Adv 2025; 9:2079-2090. [PMID: 39913776 PMCID: PMC12052699 DOI: 10.1182/bloodadvances.2024015296] [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: 11/07/2024] [Accepted: 01/23/2025] [Indexed: 04/24/2025] Open
Abstract
ABSTRACT Traumatic brain injury (TBI) causes endothelial injuries both at the site of injury and in remote regions of the brain. TBI-induced endotheliopathy also disseminates to extracranial organs, such as the lungs. This TBI-induced systemic endotheliopathy has been reported extensively in clinical studies, but its underlying mechanism remains poorly understood. Here, we report results from a study designed to investigate the role of extracellular mitochondria (exMt) in TBI-induced endotheliopathy. We show that exMt are released from injured brains into the circulation, reaching a maximal level of 1.8 × 104/μL at 3 hours after mice were subjected to severe TBI. These exMt express peroxidized cardiolipin on their surface and generate significant amounts of reactive oxygen species, but they produce less adenosine triphosphate compared with intracellular mitochondria of normal cells. When infused into noninjured mice, exMt induce systemic endotheliopathy defined by increased endothelial permeability, tissue edema, and perivascular bleeding. We further demonstrate that endothelial cells (ECs) endocytose exMt through the lipid-scavenging receptor CD36 and dynamin-driven clathrin-mediated pathway. The endocytosed exMt interacted with the endoplasmic reticulum (ER) of ECs through newly formed mitochondria-associated membranes, leading to ER stress and resultant apoptosis. This study delineates a new pathway of exMt-induced endotheliopathy during acute TBI. It also demonstrates a causal role of exMt in endothelial injuries associated with (poly)trauma, severe infection, and autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoxi Lu
- Bloodworks Research Institute, Seattle, WA
- Department of Pediatric Hematology/Oncology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second Hospital, Sichuan University School of Medicine, Chengdu, China
| | - Lujia Tang
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | - Yingang Wu
- Department of Neurosurgery, The First Affiliated Hospital, School of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | | | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | | | | | - Cha Han
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | | | - Yi Wang
- Bloodworks Research Institute, Seattle, WA
| | - Rosemary Kozar
- Department of Surgery, Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD
| | | | - Xiaoyun Fu
- Bloodworks Research Institute, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | - Zilong Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital and Tianjin Neurological Institute, Tianjin, China
| | - Jing-fei Dong
- Bloodworks Research Institute, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
2
|
Li K, Wang J, Gao Y, Chen X, Peng R, Li L, Wang C, Li T, Zhang S, Yang G, Zhang J. Benzbromarone improves blood hypercoagulability after TBI by reducing phosphatidylserine externalization through inhibition of TMEM16F expression. Life Sci 2025; 366-367:123501. [PMID: 39983827 DOI: 10.1016/j.lfs.2025.123501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
AIMS Traumatic brain injury-induced coagulopathy (TBI-IC) frequently occurs after TBI, exacerbating the severity of TBI and affecting patient prognosis. Benzbromarone (BBR) is commonly used to treat hyperuricemia; however, its protective effects against TBI-IC remain unknown. Therefore, we explored whether BBR could improve TBI. MATERIALS AND METHODS C57BL/6 wild-type mice were subjected to fluid percussion injury to mimic TBI, and BBR was administered intraperitoneally 30 min after TBI. Magnetic resonance imaging (MRI) and Evans blue dye extravasation were used to assess the prognosis, tail bleeding time, ELISA, and coagulation tests assess coagulation function. We further explored the potential mechanism by which BBR alleviates hypercoagulation after TBI using flow cytometry. KEY FINDINGS The intraperitoneally injected BBR group showed improved survival and neurological severity scores compared to the TBI group. Subsequently, we found that hypercoagulability developed 3 h after TBI and that the administration of BBR improved this hypercoagulability. BBR also reduced the degree of platelet phosphatidylserine (PS) exposure after TBI, platelet activation, and Ca2+ overload, in addition to inhibition of scramblase activity in procoagulant platelets. SIGNIFICANCE Our findings indicate that BBR reduces PS externalization by inhibiting TMEM16F expression, thereby improving blood hypercoagulability after TBI.
Collapse
Affiliation(s)
- Kaiji Li
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinchao Wang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yalong Gao
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Xin Chen
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruilong Peng
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Lei Li
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Cong Wang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tuo Li
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Shu Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China.
| | - Guili Yang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China.
| | - Jianning Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin, China.
| |
Collapse
|
3
|
Li L, Peng R, Wang C, Chen X, Gheyret D, Guan S, Chen B, Liu Y, Liu X, Cao Y, Han C, Xiong J, Li F, Lu T, Jia H, Li K, Wang J, Zhang X, Xu J, Wang Y, Xu X, Li T, Zhang J, Zhang S. β2 integrin regulates neutrophil trans endothelial migration following traumatic brain injury. Cell Commun Signal 2025; 23:70. [PMID: 39923080 PMCID: PMC11806581 DOI: 10.1186/s12964-025-02071-9] [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: 11/08/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
Neutrophils are the first responders among peripheral immune cells to infiltrate the central nervous system following a traumatic brain injury (TBI), triggering neuroinflammation that can exacerbate secondary tissue damage. The precise molecular controls that dictate the inflammatory behavior of neutrophils post-TBI, however, remain largely elusive. Our comprehensive analysis of the molecular landscape surrounding the trauma in TBI mice has revealed a significant alteration in the abundance of β2 integrin (ITGB2), predominantly expressed by neutrophils and closely associated with immune responses. Using the fluid percussion injury (FPI) mouse model, we investigated the therapeutic efficacy of Rovelizumab, an agent that blocks ITGB2. The treatment has demonstrated significant improvements in neurologic function in TBI mice, attenuating blood-brain barrier permeability, mitigating oxidative stress and inflammatory mediator release, and enhancing cerebral perfusion. Moreover, ITGB2 blockade has effectively limited the adherence, migration, and infiltration of neutrophils, and has impeded the formation of neutrophil extracellular traps (NETs) upon their activation. Finally, it was demonstrated that ITGB2 mediates these effects mainly through its interaction with intercellular adhesion molecule-1 (ICAM 1) of endotheliocyte. These findings collectively illuminate ITGB2 as a crucial molecular switch that governs the adverse effects of neutrophils post-TBI and could be targeted to improve clinical outcome in patients.
Collapse
Affiliation(s)
- Lei Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Ruilong Peng
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300200, China
| | - Cong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Dilmurat Gheyret
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Siyu Guan
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Bo Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Yafan Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Xilei Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yiyao Cao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jianhua Xiong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Fanjian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Taoyuan Lu
- Xuanwu Jinan Hospital, 5106 Jingshi Road, Jinan, 250000, Shandong, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Haoran Jia
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Kaiji Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Jinchao Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Xu Zhang
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jianye Xu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
| | - Yajuan Wang
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xin Xu
- Xuanwu Jinan Hospital, 5106 Jingshi Road, Jinan, 250000, Shandong, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
| | - Tuo Li
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300200, China.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China.
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin State Key Laboratory of Experimental Hematology, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, China.
| |
Collapse
|
4
|
Ma JQ, Wang L, Zhang Y, Bian YQ, Qu XP, Song LJ, Wang C, Gao L, Fang QX, Zhao DC, Shen LL, Liu B. Single-nucleus RNA sequencing-based construction of a hippocampal neuron atlas in mice with epileptic cognitive impairment. iScience 2024; 27:111065. [PMID: 39635132 PMCID: PMC11615225 DOI: 10.1016/j.isci.2024.111065] [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: 05/28/2024] [Revised: 07/13/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024] Open
Abstract
The hippocampus plays a critical role in learning and memory, and mice with epileptic cognitive impairment exhibit hippocampal atrophy. However, there is still a lack of research on the hippocampal cell atlas related to these disorders. Here, we utilized snRNA-seq to characterize the transcriptomic changes in hippocampal neurons of drug-resistant epilepsy (DRE) cognitive-impaired mice. The intercellular heterogeneity of 20 subpopulations of neurons was analyzed, focusing on aspects such as cell communication, gene expressions, GO and KEGG enrichment analysis, and module gene set analysis. Based on the degree of relevance to synaptic biological functions, the subpopulations associated with cognitive impairment (ExN1, 3, 8 and InN1, 6) were preliminarily identified. We also identified some key biomarkers in DRE cognitive-impaired mice, such as Ptprz1 and Calb1. Finally, we integrate and validate our dataset using identified well-annotated marker genes in the hippocampal region, further supporting the functional annotation of neuronal subpopulations.
Collapse
Affiliation(s)
- Jia-Qi Ma
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Lu Wang
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
- College of Life Sciences, Northwest University, Xi’an, Shaanxi 710069, China
| | - Yue Zhang
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Yong-Qian Bian
- Department of Plastic and Burn Surgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Xiao-Peng Qu
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Li-Jia Song
- Department of Pediatrics, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Chao Wang
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Qi-Xing Fang
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - De-Chang Zhao
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| | - Liang-Liang Shen
- Department of Biochemistry and Molecular Biology, Airforce Military Medical University, Xi’an, China
| | - Bei Liu
- Department of Neurosurgery, Tangdu Hospital, Airforce Military Medical University, Xi’an, China
| |
Collapse
|
5
|
Li D, Cho MS, Gonzalez‐Delgado R, Liang X, Dong J, Cruz MA, Ma Q, Afshar‐Kharghan V. The effect of ADAMTS13 on graft-versus-host disease. J Cell Mol Med 2024; 28:e18457. [PMID: 38963011 PMCID: PMC11222974 DOI: 10.1111/jcmm.18457] [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: 12/03/2023] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 07/05/2024] Open
Abstract
Allogeneic haematopoietic stem cell transplantation (allo-HSCT) can potentially cure malignant blood disorders and benign conditions such as haemoglobinopathies and immunologic diseases. However, allo-HSCT is associated with significant complications. The most common and debilitating among them is graft-versus-host disease (GVHD). In GVHD, donor-derived T cells mount an alloimmune response against the recipient. The alloimmune response involves several steps, including recognition of recipient antigens, activation and proliferation of T cells in secondary lymphoid organs, and homing into GVHD-targeted organs. Adhesion molecules on T cells and endothelial cells mediate homing of T cells into lymphoid and non-lymphoid tissues. In this study, we showed that Von Willebrand factor (VWF), an adhesion molecule secreted by activated endothelial cells, plays an important role in mouse models of GVHD. We investigated the effect of the VWF-cleaving protease ADAMTS13 on GVHD. We found that ADAMTS13 reduced the severity of GVHD after bone marrow transplantation from C57BL6 donor to BALB/C recipient mice. A recombinant VWF-A2 domain peptide also reduced GVHD in mice. We showed that ADAMTS13 and recombinant VWF-A2 reduced the binding of T cells to endothelial cells and VWF in vitro, and reduced the number of T cells in lymph nodes, Peyer's patches and GVHD-targeted organs in vivo. We identified LFA-1 (αLβ2) as the binding site of VWF on T cells. Our results showed that blocking T-cell homing by ADAMTS13 or VWF-A2 peptide reduced the severity of the GVHD after allo-HSCT, a potentially novel method for treating and preventing GVHD.
Collapse
Affiliation(s)
- Dan Li
- Department of Hematopoietic Biology & MalignancyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Min Soon Cho
- Section of Benign HematologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Xiaowen Liang
- Department of Integrative Biology and PharmacologyMcGovern Medical School, The University of Texas Health Science CenterHoustonTexasUSA
| | - Jing‐Fei Dong
- Bloodworks Research Institute and Hematology Division, Department of MedicineUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Miguel A. Cruz
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical CenterHoustonTexasUSA
- Baylor College of MedicineHoustonTexasUSA
- Department of MedicineBaylor College of MedicineHoustonTexasUSA
| | - Qing Ma
- Department of Hematopoietic Biology & MalignancyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Vahid Afshar‐Kharghan
- Section of Benign HematologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
6
|
Ho JW, Dawood ZS, Taylor ME, Liggett MR, Jin G, Jaishankar D, Nadig SN, Bharat A, Alam HB. THE NEUROENDOTHELIAL AXIS IN TRAUMATIC BRAIN INJURY: MECHANISMS OF MULTIORGAN DYSFUNCTION, NOVEL THERAPIES, AND FUTURE DIRECTIONS. Shock 2024; 61:346-359. [PMID: 38517237 DOI: 10.1097/shk.0000000000002307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
ABSTRACT Severe traumatic brain injury (TBI) often initiates a systemic inflammatory response syndrome, which can potentially culminate into multiorgan dysfunction. A central player in this cascade is endotheliopathy, caused by perturbations in homeostatic mechanisms governed by endothelial cells due to injury-induced coagulopathy, heightened sympathoadrenal response, complement activation, and proinflammatory cytokine release. Unique to TBI is the potential disruption of the blood-brain barrier, which may expose neuronal antigens to the peripheral immune system and permit neuroinflammatory mediators to enter systemic circulation, propagating endotheliopathy systemically. This review aims to provide comprehensive insights into the "neuroendothelial axis" underlying endothelial dysfunction after TBI, identify potential diagnostic and prognostic biomarkers, and explore therapeutic strategies targeting these interactions, with the ultimate goal of improving patient outcomes after severe TBI.
Collapse
Affiliation(s)
- Jessie W Ho
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Zaiba Shafik Dawood
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Meredith E Taylor
- Department of Surgery, Division of Organ Transplant, and Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Marjorie R Liggett
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Guang Jin
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Dinesh Jaishankar
- Department of Surgery, Division of Organ Transplant, and Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Satish N Nadig
- Department of Surgery, Division of Organ Transplant, and Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University Chicago, Illinois
| | - Ankit Bharat
- Department of Surgery, Division of Thoracic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hasan B Alam
- Department of Surgery, Division of Trauma Surgery and Critical Care, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
7
|
Liu W, Patel K, Wang Y, Nodzenski M, Nguyen A, Teramura G, Higgins HA, Hoogeveen RC, Couper D, Fu X, Konkle BA, Loop MS, Dong JF. Dynamic and functional linkage between von Willebrand factor and ADAMTS-13 with aging: an Atherosclerosis Risk in Community study. J Thromb Haemost 2023; 21:3371-3382. [PMID: 37574196 DOI: 10.1016/j.jtha.2023.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/10/2023] [Accepted: 07/16/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND von Willebrand factor (VWF) is a multimeric glycoprotein critically involved in hemostasis, thrombosis, and inflammation. VWF function is regulated by its antigen levels, multimeric structures, and the state of enzymatic cleavage. Population studies in the past have focused almost exclusively on VWF antigen levels in cross-sectional study designs. OBJECTIVE To identify subjects in the Atherosclerosis Risk in Community study who had persistently low and high VWF antigen over 10 years and to quantify longitudinal changes in the biological activities and cleavage of VWF in these subjects. METHODS We measured VWF antigen, propeptide, adhesive activities, and cleavage by ADAMTS-13 quantified using a mass spectrometry method that detected the cleaved VWF peptide EQAPNLVY, as well as coagulation factor VIII activity. RESULTS We determined the mean subject-specific increase in VWF to be 22.0 International Units (IU)/dL over 10 years, with 95% between -0.3 and 59.7 IU/dL. This aging-related increase was also detected in VWF propeptide levels, ristocetin cofactor activity, and VWF binding to collagen. We identified 4.1% and 25.0% of subjects as having persistently low (<50 IU/dL) and high (>200 IU/dL) VWF antigen, respectively. Subjects with persistently low VWF had enhanced ristocetin cofactor activity, whereas those with persistently high VWF had elevated levels of ADAMTS-13, resulting in a comparable rate of VWF cleavage between the 2 groups. CONCLUSIONS These results provide new information about the effects of aging on VWF antigens and adhesive activity and identify a functional coordination between VWF and the rate of its cleavage by ADAMTS-13.
Collapse
Affiliation(s)
- Wei Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China; Bloodworks Research Institute, Seattle, WA, USA
| | | | - Yi Wang
- Bloodworks Research Institute, Seattle, WA, USA
| | - Michael Nodzenski
- Department of Biostatistics, Collaborative Studies Coordinating Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | - Ron C Hoogeveen
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - David Couper
- Department of Biostatistics, Collaborative Studies Coordinating Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiaoyun Fu
- Bloodworks Research Institute, Seattle, WA, USA
| | - Barbara A Konkle
- Washington Center for Bleeding Disorders, Seattle, WA, USA; Division of Hematology, University of Washington School of Medicine, Seattle, WA, USA.
| | - Matthew Shane Loop
- Department of Health Outcomes Organization and Policy, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA, USA; Division of Hematology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
8
|
Dong X, Dong JF, Zhang J. Roles and therapeutic potential of different extracellular vesicle subtypes on traumatic brain injury. Cell Commun Signal 2023; 21:211. [PMID: 37596642 PMCID: PMC10436659 DOI: 10.1186/s12964-023-01165-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/13/2023] [Indexed: 08/20/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of injury-related disability and death around the world, but the clinical stratification, diagnosis, and treatment of complex TBI are limited. Due to their unique properties, extracellular vesicles (EVs) are emerging candidates for being biomarkers of traumatic brain injury as well as serving as potential therapeutic targets. However, the effects of different extracellular vesicle subtypes on the pathophysiology of traumatic brain injury are very different, or potentially even opposite. Before extracellular vesicles can be used as targets for TBI therapy, it is necessary to classify different extracellular vesicle subtypes according to their functions to clarify different strategies for EV-based TBI therapy. The purpose of this review is to discuss contradictory effects of different EV subtypes on TBI, and to propose treatment ideas based on different EV subtypes to maximize their benefits for the recovery of TBI patients. Video Abstract.
Collapse
Affiliation(s)
- Xinlong Dong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, Nansihuan West Road, Fengtai District, Beijing, China.
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA, USA
- Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
9
|
Li L, Li F, Bai X, Jia H, Wang C, Li P, Zhang Q, Guan S, Peng R, Zhang S, Dong JF, Zhang J, Xu X. Circulating extracellular vesicles from patients with traumatic brain injury induce cerebrovascular endothelial dysfunction. Pharmacol Res 2023; 192:106791. [PMID: 37156450 DOI: 10.1016/j.phrs.2023.106791] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Endothelial dysfunction is a key proponent of pathophysiological process of traumatic brain injury (TBI). We previously demonstrated that extracellular vesicles (EVs) released from injured brains led to endothelial barrier disruption and vascular leakage. However, the molecular mechanisms of this EV-induced endothelial dysfunction (endotheliopathy) remain unclear. Here, we enriched plasma EVs from TBI patients (TEVs), and detected high mobility group box 1 (HMGB1) exposure to 50.33 ± 10.17% of TEVs and the number of HMGB1+TEVs correlated with injury severity. We then investigated for the first time the impact of TEVs on endothelial function using adoptive transfer models. We found that TEVs induced dysfunction of cultured human umbilical vein endothelial cells and mediated endothelial dysfunction in both normal and TBI mice, which were propagated through the HMGB1-activated receptor for advanced glycation end products (RAGE)/Cathepsin B signaling, and the resultant NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and canonical caspase-1/gasdermin D (GSDMD)-dependent pyroptosis. Finally, von Willebrand factor (VWF) was detected on the surface of 77.01 ± 7.51% of HMGB1+TEVs. The TEV-mediated endotheliopathy was reversed by a polyclonal VWF antibody, indicating that VWF might serve a coupling factor that tethered TEVs to ECs, thus facilitating HMGB1-induced endotheliopathy. These results suggest that circulating EVs isolated from patients with TBI alone are sufficient to induce endothelial dysfunction and contribute to secondary brain injury that are dependent on immunologically active HMGB1 exposed on their surface. This finding provided new insight for the development of potential therapeutic targets and diagnostic biomarkers for TBI.
Collapse
Affiliation(s)
- Lei Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Fanjian Li
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Xuesong Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China
| | - Haoran Jia
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Cong Wang
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Peng Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; Department of Neurosurgery, Beijing Fengtai You'anmen Hospital, 199 You'anmen Outer Street, Beijing, China
| | - Qiaoling Zhang
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Siyu Guan
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Ruilong Peng
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Shu Zhang
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China
| | - Jing-Fei Dong
- Bloodworks Research Institute and Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, WA, USA
| | - Jianning Zhang
- Tianjin Neurological Institute; Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, China.
| | - Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China.
| |
Collapse
|
10
|
Cai W, Wang M, Wang CY, Zhao CY, Zhang XY, Zhou Q, Zhao WJ, Yang F, Zhang CL, Yang AJ, Dong JF, Li M. Extracellular vesicles, hyperadhesive von willebrand factor, and outcomes of gastric cancer: a clinical observational study. Med Oncol 2023; 40:140. [PMID: 37031314 DOI: 10.1007/s12032-023-01950-w] [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: 08/23/2022] [Accepted: 01/12/2023] [Indexed: 04/10/2023]
Abstract
Von Willebrand factor (VWF) is an adhesive ligand critical for maintaining hemostasis. However, it has also been increasingly recognized for its role in cancer development because it has been shown to mediate the adhesion of cancer cells to endothelial cells, promote the epithelial-mesenchymal transition, and enhance angiogenesis. We have previously shown that gastric cancer cells synthesize VWF, which mediates the interaction between the cancer and endothelial cells to promote cancer growth. Here, we report results from a clinical observational study that demonstrate the association of VWF in plasma and on the surface of extracellular vesicles (EVs) with the pathological characteristics of gastric cancer. We found that patients with gastric cancer had elevated and intrinsically hyperadhesive VWF in their peripheral blood samples. VWF was detected on the surface of EVs from cancer cells, platelets, and endothelial cells. Higher levels of these VWF-bound EVs were associated with cancer aggression and poor clinical outcomes for patients. These findings suggest that VWF+ EVs from different cell types serve collectively as a new class of biomarkers for the outcome assessment of gastric cancer patients.
Collapse
Affiliation(s)
- Wei Cai
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
- Gansu Provincial Hospital, Lanzhou, China
| | - Min Wang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
- School of Basic Medical Sciences, Institute of Integrated Traditional Chinese and Western Medicine, Lanzhou University, Lanzhou, China
| | - Chen-Yu Wang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Chan-Yuan Zhao
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Xiao-Yu Zhang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Quan Zhou
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Wen-Jie Zhao
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Feng Yang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Chen-Li Zhang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China
| | - Ai-Jun Yang
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China.
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA, USA.
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Min Li
- School of Basic Medical Sciences, Institute of Pathology, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China.
| |
Collapse
|
11
|
Tischer A, Moon-Tasson L, Auton M. Removal of the vicinal disulfide enhances the platelet-capturing function of von Willebrand factor. Blood 2023; 141:1469-1473. [PMID: 36603190 PMCID: PMC10082372 DOI: 10.1182/blood.2022018803] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023] Open
Abstract
A redox autoinhibitory mechanism has previously been proposed, in which the reduced state of the vicinal disulfide bond in the von Willebrand factor (VWF) A2 domain allows A2 to bind to A1 and inhibit platelet adhesion to the A1 domain. The VWF A1A2A3 tridomain was expressed with and without the vicinal disulfide in A2 (C1669S/C1670S) via the atomic replacement of sulfur for oxygen to test the relevance of the vicinal disulfide to the physiological platelet function of VWF under shear flow. A comparative study of the shear-dependent platelet translocation dynamics on these tridomain variants reveals that the reduction of the vicinal disulfide moderately increases the platelet-capturing function of A1, an observation counter to the proposed hypothesis. Surface plasmon resonance spectroscopy confirms that C1669S/C1670S slightly increases the affinity of A1A2A3 binding to glycoprotein Ibα (GPIbα). Differential scanning calorimetry and hydrogen-deuterium exchange mass spectrometry demonstrate that reduction of the vicinal disulfide destabilizes the A2 domain, which consequently disrupts interactions between the A1, A2, and A3 domains and enhances the conformational dynamics of A1-domain secondary structures known to regulate the strength of platelet adhesion to VWF. This study clarifies that the reduced state of the A2 vicinal disulfide is not inhibitory but rather slightly activating.
Collapse
Affiliation(s)
- Alexander Tischer
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Laurie Moon-Tasson
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Matthew Auton
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| |
Collapse
|
12
|
Dong JF, Zhang F, Zhang J. Detecting traumatic brain injury-induced coagulopathy: What we are testing and what we are not. J Trauma Acute Care Surg 2023; 94:S50-S55. [PMID: 35838367 PMCID: PMC9805481 DOI: 10.1097/ta.0000000000003748] [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] [Indexed: 02/07/2023]
Abstract
ABSTRACT Coagulopathy after traumatic brain injury (TBI) is common and has been closely associated with poor clinical outcomes for the affected patients. Traumatic brain injury-induced coagulopathy (TBI-IC) is consumptive in nature and evolves rapidly from an injury-induced hypercoagulable state. Traumatic brain injury-induced coagulopathy defined by laboratory tests is significantly more frequent than clinical coagulopathy, which often manifests as secondary, recurrent, or delayed intracranial or intracerebral hemorrhage. This disparity between laboratory and clinical coagulopathies has hindered progress in understanding the pathogenesis of TBI-IC and developing more accurate and predictive tests for this severe TBI complication. In this review, we discuss laboratory tests used in clinical and research studies to define TBI-IC, with specific emphasis on what the tests detect and what they do not. We also offer perspective on developing more accurate and predictive tests for this severe TBI complication.
Collapse
Affiliation(s)
- Jing-fei Dong
- Bloodworks Research Institute, Seattle, WA, USA
- Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, WA, USA
| | - Fangyi Zhang
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - Jianning Zhang
- Tianjin Institute of Neurology, Tianjin, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
13
|
Li T, Li L, Peng R, Hao H, Zhang H, Gao Y, Wang C, Li F, Liu X, Chen F, Zhang S, Zhang J. Abrocitinib Attenuates Microglia-Mediated Neuroinflammation after Traumatic Brain Injury via Inhibiting the JAK1/STAT1/NF-κB Pathway. Cells 2022; 11:cells11223588. [PMID: 36429017 PMCID: PMC9688110 DOI: 10.3390/cells11223588] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Neuroinflammation has been shown to play a critical role in secondary craniocerebral injury, leading to poor outcomes for TBI patients. Abrocitinib, a Janus kinase1 (JAK1) selective inhibitor approved to treat atopic dermatitis (AD) by the Food and Drug Administration (FDA), possesses a novel anti-inflammatory effect. In this study, we investigated whether abrocitinib could ameliorate neuroinflammation and exert a neuroprotective effect in traumatic brain injury (TBI) models. METHODS First, next-generation sequencing (NGS) was used to select genes closely related to neuroinflammation after TBI. Then, magnetic resonance imaging (MRI) was used to dynamically observe the changes in traumatic focus on the 1st, 3rd, and 7th days after the induction of fluid percussion injury (FPI). Moreover, abrocitinib's effects on neurobehaviors were evaluated. A routine peripheral blood test was carried out and Evans blue dye extravasation, cerebral cortical blood flow, the levels of inflammatory cytokines, and changes in the numbers of inflammatory cells were evaluated to investigate the function of abrocitinib on the 1st day post-injury. Furthermore, the JAK1/signal transducer and activator of transcription1 (STAT1)/nuclear factor kappa (NF-κB) pathway was assessed. RESULTS In vivo, abrocitinib treatment was found to shrink the trauma lesions. Compared to the TBI group, the abrocitinib treatment group showed better neurological function, less blood-brain barrier (BBB) leakage, improved intracranial blood flow, relieved inflammatory cell infiltration, and reduced levels of inflammatory cytokines. In vitro, abrocitinib treatment was shown to reduce the pro-inflammatory M1 microglia phenotype and shift microglial polarization toward the anti-inflammatory M2 phenotype. The WB and IHC results showed that abrocitinib played a neuroprotective role by restraining JAK1/STAT1/NF-κB levels after TBI. CONCLUSIONS Collectively, abrocitinib treatment after TBI is accompanied by improvements in neurological function consistent with radiological, histopathological, and biochemical changes. Therefore, abrocitinib can indeed reduce excessive neuroinflammation by restraining the JAK1/STAT1/NF-κB pathway.
Collapse
Affiliation(s)
- Tuo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Department of Neurosurgery, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Lei Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Ruilong Peng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Hongying Hao
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300000, China
- Department of Neurology, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Hejun Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Department of Neurosurgery, First Hospital of Qinhuangdao, Qinhuangdao 066000, China
| | - Yalong Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Cong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Fanjian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Graduate School, Tianjin Medical University, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Xilei Liu
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Correspondence: (S.Z.); (J.Z.)
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300000, China
- Tianjin Neurological Institute, Tianjin 300000, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300000, China
- Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin 300000, China
- Correspondence: (S.Z.); (J.Z.)
| |
Collapse
|
14
|
Xu X, Feng Y, Jia Y, Zhang X, Li L, Bai X, Jiao L. Prognostic value of von Willebrand factor and ADAMTS13 in patients with COVID-19: A systematic review and meta-analysis. Thromb Res 2022; 218:83-98. [PMID: 36027630 PMCID: PMC9385270 DOI: 10.1016/j.thromres.2022.08.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/14/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Endotheliopathy and coagulopathy appear to be the main causes for critical illness and death in patients with coronavirus disease 2019 (COVID-19). The adhesive ligand von Willebrand factor (VWF) has been involved in immunothrombosis responding to endothelial injury. Here, we reviewed the current literature and performed meta-analyses on the relationship between both VWF and its cleaving protease ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13) with the prognosis of COVID-19. METHODS We searched MEDLINE, Cochrane Library, Web of Science, and EMBASE databases from inception to 4 March 2022 for studies analyzing the relationship between VWF-related variables and composite clinical outcomes of patients with COVID-19. The VWF-related variables analyzed included VWF antigen (VWF:Ag), VWF ristocetin cofactor (VWF:Rco), ADAMTS13 activity (ADAMTS13:Ac), the ratio of VWF:Ag to ADAMTS13:Ac, and coagulation factor VIII (FVIII). The unfavorable outcomes were defined as mortality, intensive care unit (ICU) admission, and severe disease course. We used random or fixed effects models to create summary estimates of risk. Risk of bias was assessed based on the principle of the Newcastle-Ottawa Scale. RESULTS A total of 3764 patients from 40 studies were included. The estimated pooled means indicated increased plasma levels of VWF:Ag, VWF:Rco, and VWF:Ag/ADAMTS13:Ac ratio, and decreased plasma levels of ADAMTS13:Ac in COVID-19 patients with unfavorable outcomes when compared to those with favorable outcomes (composite outcomes or subgroup analyses of non-survivor versus survivor, ICU versus non-ICU, and severe versus non-severe). In addition, FVIII were higher in COVID-19 patients with unfavorable outcomes. Subgroup analyses indicated that FVIII was higher in patients admitting to ICU, while there was no significant difference between non-survivors and survivors. CONCLUSIONS The imbalance of the VWF-ADAMTS13 axis (massive quantitative and qualitative increases of VWF with relative deficiency of ADAMTS13) is associated with poor prognosis of patients with COVID-19.
Collapse
Affiliation(s)
- Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China.
| | - Yao Feng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China
| | - Yitong Jia
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China
| | - Long Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China
| | - Xuesong Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China; China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, China; Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China..
| |
Collapse
|
15
|
Zhang Y, Wang L, Pan Q, Yang X, Cao Y, Yan J, Wang Y, Tao Y, Fan R, Sun X, Li L. Selective sphingosine-1-phosphate receptor 1 modulator attenuates blood-brain barrier disruption following traumatic brain injury by inhibiting vesicular transcytosis. Fluids Barriers CNS 2022; 19:57. [PMID: 35820896 PMCID: PMC9277863 DOI: 10.1186/s12987-022-00356-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) provokes secondary pathological damage, such as damage to the blood-brain barrier (BBB), ischaemia and inflammation. Major facilitator superfamily domain-containing 2a (Mfsd2a) has been demonstrated to be critical in limiting the increase in BBB vesicle transcytosis following brain injury. Recent studies suggest that a novel and selective modulator of the sphingosine-1-phosphate receptor 1 (S1P1), CYM-5442, maintains the integrity of the BBB by restricting vesicle transcytosis during acute ischaemic stroke. In the current study, we investigated whether CYM-5442, evaluated in a short-term study, could protect the brains of mice with acute-stage TBI by reversing the increase in vesicle transport due to reduced Mfsd2a expression after TBI. METHODS We used the well-characterized model of TBI caused by controlled cortical impact. CYM-5442 (0.3, 1, 3 mg/kg) was intraperitoneally injected 30 min after surgery for 7 consecutive days. To investigate the effect of CYM-5442 on vesicle transcytosis, we downregulated and upregulated Mfsd2a expression using a specific AAV prior to evaluation of the TBI model. MRI scanning, cerebral blood flow, circulating blood counts, ELISA, TEM, WB, and immunostaining evaluations were performed after brain injury. RESULTS CYM-5442 significantly attenuated neurological deficits and reduced brain oedema in TBI mice. CYM-5442 transiently suppressed lymphocyte trafficking but did not induce persistent lymphocytopenia. After TBI, the levels of Mfsd2a were decreased significantly, while the levels of CAV-1 and albumin were increased. In addition, Mfsd2a deficiency caused inadequate sphingosine-1-phosphate (S1P) transport in the brain parenchyma, and the regulation of BBB permeability by Mfsd2a after TBI was shown to be related to changes in vesicle transcytosis. Downregulation of Mfsd2a in mice markedly increased the BBB permeability, neurological deficit scores, and brain water contents after TBI. Intervention with CYM-5442 after TBI protected the BBB by significantly reducing the vesicle transcytosis of cerebrovascular endothelial cells. CONCLUSION In addition to transiently suppressing lymphocytes, CYM-5442 alleviated the neurological deficits, cerebral edema and protective BBB permeability in TBI mice by reducing the vesicle transcytosis of cerebrovascular endothelial cells.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Lin Wang
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Qiuling Pan
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaomin Yang
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yunchuan Cao
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jin Yan
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yingwen Wang
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Runjin Fan
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Xiaochuan Sun
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Lin Li
- Department of Neurosurgery, Neural Injury and Protection Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
16
|
Wang J, Xie X, Wu Y, Zhou Y, Li Q, Li Y, Xu X, Wang M, Murdiyarso L, Houck K, Hilton T, Chung D, Li M, Zhang JN, Dong J. Brain-Derived Extracellular Vesicles Induce Vasoconstriction and Reduce Cerebral Blood Flow in Mice. J Neurotrauma 2022; 39:879-890. [PMID: 35316073 DOI: 10.1089/neu.2021.0274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) impairs cerebrovascular autoregulation and reduces cerebral blood flow (CBF), leading to ischemic secondary injuries. We have shown that injured brains release brain-derived extracellular vesicles (BDEVs) into circulation, where they cause a systemic hypercoagulable state that rapidly turns into consumptive coagulopathy. BDEVs induce endothelial injury and permeability, leading to the hypothesis that they contribute to TBI-induced cerebrovascular dysregulation. In a study designed to test this hypothesis, we detected circulating BDEVs in C57BL/6J mice subjected to severe TBI, reaching peak levels of 3x104/µl at 3 hours post injury (71.2±21.5% of total annexin V-binding EVs). We further showed in an adaptive transfer model that 41.7±5.8% of non-injured mice died within 6 hours after being infused with 3x104/µl of BDEVs. BDEVs transmigrated through the vessel walls, induced rapid vasoconstriction by inducing calcium influx in vascular smooth muscle cells, and reduced CBF by 93.8±5.6% within 30 minutes after infusion. The CBF suppression was persistent in mice that eventually died but it recovered quickly in surviving mice. It was prevented by the calcium channel blocker nimodipine. When being separated, neither protein nor phospholipid components from the lethal number of BDEVs induced vasoconstriction, reduced CBF, and caused death. These results demonstrate a novel vasoconstrictive activity of BDEVs that depends on the structure of BDEVs and contributes to TBI-induced disseminated cerebral ischemia and sudden death.
Collapse
Affiliation(s)
- Jiwei Wang
- Tianjin Neurological Institute, 230967, Anshan road No.154, Tianjin, China, 300052;
| | - Xiaofeng Xie
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | - Yingang Wu
- University of Science and Technology of China, 12652, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine., Hefei, Anhui, China;
| | - Yuan Zhou
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Qifeng Li
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Ying Li
- Tianjin Neurological Institute, 230967, Tianjin, Tianjin, China;
| | - Xin Xu
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Min Wang
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | | | - Katie Houck
- Bloodworks Research institute, Seattle, United States;
| | | | - Dominic Chung
- Bloodworks Research institute, Seattle, United States;
| | - Min Li
- Lanzhou University, 12426, Lanzhou, Gansu, China;
| | - Jian-Ning Zhang
- Tianjin Neurological Institute, 230967, Tianjin Medical University General Hospital, Tianjin, Tianjin, China;
| | - Jingfei Dong
- Bloodworks Research Institute, Bloodworks Northwest, Seattle, Seattle, Washington, United States.,Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States;
| |
Collapse
|
17
|
Yang M, Houck KL, Dong X, Hernandez M, Wang Y, Nathan SS, Wu X, Afshar-Kharghan V, Fu X, Cruz MA, Zhang J, Nascimbene A, Dong JF. Hyperadhesive von Willebrand Factor Promotes Extracellular Vesicle-Induced Angiogenesis: Implication for LVAD-Induced Bleeding. JACC Basic Transl Sci 2022; 7:247-261. [PMID: 35411318 PMCID: PMC8993768 DOI: 10.1016/j.jacbts.2021.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/22/2022]
Abstract
VWF in patients on LVAD supports was hyperadhesive, activated platelets, and generated platelet-derived extracellular vesicles. Extracellular vesicles from LVAD patients and those from shear-activated platelets promoted aberrant angiogenesis in a VWF-dependent manner. The activated VWF exposed the A1 domain through the synergistic actions of oxidative stress and HSS generated in LVAD-driven circulation.
Bleeding associated with left ventricular assist device (LVAD) implantation has been attributed to the loss of large von Willebrand factor (VWF) multimers to excessive cleavage by ADAMTS-13, but this mechanism is not fully supported by the current evidence. We analyzed VWF reactivity in longitudinal samples from LVAD patients and studied normal VWF and platelets exposed to high shear stress to show that VWF became hyperadhesive in LVAD patients to induce platelet microvesiculation. Platelet microvesicles activated endothelial cells, induced vascular permeability, and promoted angiogenesis in a VWF-dependent manner. Our findings suggest that LVAD-driven high shear stress primarily activates VWF, rather than inducing cleavage in the majority of patients.
Collapse
Key Words
- ADAMTS-13:Ag, ADAMTS-13 antigen
- AVS, aortic vascular segment
- EC, endothelial cell
- EV, extracellular vesicle
- EVFP, extracellular vesicle–free plasma
- GI, gastrointestinal
- GOF, gain of function
- GP, glycoprotein
- GPM, growth factor-poor medium
- GRM, growth factor-rich medium
- HSS, high shear stress
- LVAD, left ventricular assist device
- PS, phosphatidylserine
- SIPA, shear-induced platelet aggregation
- ULVWF, ultra-large von Willebrand factor
- VEGF, vascular endothelial growth factor
- VWF, von Willebrand factor
- VWF:Ag, von Willebrand factor antigen
- VWF:CB, von Willebrand factor binding to collagen
- VWF:pp, von Willebrand factor propeptide
- aVWS, acquired von Willebrand syndrome
- angiogenesis
- extracellular vesicles
- left ventricular assist devices
- pEV, extracellular vesicle from von Willebrand factor-activated platelets
- platelets
- shear stress
- von Willebrand factor
Collapse
Affiliation(s)
- Mengchen Yang
- Bloodworks Research Institute, Seattle, Washington, USA.,Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Katie L Houck
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Xinlong Dong
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Maria Hernandez
- Center for Advanced Heart Failure, University of Texas at Houston, Houston, Texas, USA
| | - Yi Wang
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Sriram S Nathan
- Center for Advanced Heart Failure, University of Texas at Houston, Houston, Texas, USA
| | - Xiaoping Wu
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Department of Pulmonary Medicine, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Xiaoyun Fu
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Miguel A Cruz
- Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine.,Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, Texas, USA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Angelo Nascimbene
- Center for Advanced Heart Failure, University of Texas at Houston, Houston, Texas, USA
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, Washington, USA.,Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
18
|
Luo W, Yang Z, Zhang W, Zhou D, Guo X, Wang S, He F, Wang Y. Quantitative Proteomics Reveals the Dynamic Pathophysiology Across Different Stages in a Rat Model of Severe Traumatic Brain Injury. Front Mol Neurosci 2022; 14:785938. [PMID: 35145378 PMCID: PMC8821658 DOI: 10.3389/fnmol.2021.785938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/30/2021] [Indexed: 11/30/2022] Open
Abstract
Background Severe traumatic brain injury (TBI) has become a global health problem and causes a vast worldwide societal burden. However, distinct mechanisms between acute and subacute stages have not been systemically revealed. The present study aimed to identify differentially expressed proteins in severe TBI from the acute to subacute phase. Methods Sixty Sprague Dawley (SD) rats were randomly divided into sham surgery and model groups. The severe TBI models were induced by the controlled cortical impact (CCI) method. We evaluated the neurological deficits through the modified neurological severity score (NSS). Meanwhile, H&E staining and immunofluorescence were performed to assess the injured brain tissues. The protein expressions of the hippocampus on the wounded side of CCI groups and the same side of Sham groups were analyzed by the tandem mass tag-based (TMT) quantitative proteomics on the third and fourteenth days. Then, using the gene ontology (GO), Kyoto encyclopedia of genes and genomes (KEGG), and protein–protein interaction (PPI), the shared and stage-specific differentially expressed proteins (DEPs) were screened, analyzed, and visualized. Eventually, target proteins were further verified by Western blotting (WB). Results In the severe TBI, the neurological deficits always exist from the acute stage to the subacute stage, and brain parenchyma was dramatically impaired in either period. Of the significant DEPs identified, 312 were unique to the acute phase, 76 were specific to the subacute phase, and 63 were shared in both. Of the 375 DEPs between Sham-a and CCI-a, 240 and 135 proteins were up-regulated and down-regulated, respectively. Of 139 DEPs, 84 proteins were upregulated, and 55 were downregulated in the Sham-s and CCI-s. Bioinformatics analysis revealed that the differential pathophysiology across both stages. One of the most critical shared pathways is the complement and coagulation cascades. Notably, three pathways associated with gastric acid secretion, insulin secretion, and thyroid hormone synthesis were only enriched in the acute phase. Amyotrophic lateral sclerosis (ALS) was significantly enriched in the subacute stage. WB experiments confirmed the reliability of the TMT quantitative proteomics results. Conclusion Our findings highlight the same and different pathological processes in the acute and subacute phases of severe TBI at the proteomic level. The results of potential protein biomarkers might facilitate the design of novel strategies to treat TBI.
Collapse
Affiliation(s)
- Weikang Luo
- Department of Integrated Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaoyu Yang
- Department of Integrated Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Dan Zhou
- Periodical Office, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaohang Guo
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Shunshun Wang
- Postpartum Health Care Department, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Feng He
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Department of Integrated Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yang Wang,
| |
Collapse
|
19
|
Fletcher-Sandersjöö A, Tatter C, Tjerkaski J, Bartek J, Svensson M, Thelin EP, Bellander BM. Clinical Significance of Vascular Occlusive Events following Moderate-to-Severe Traumatic Brain Injury: An Observational Cohort Study. Semin Thromb Hemost 2022; 48:301-308. [PMID: 34991168 DOI: 10.1055/s-0041-1740567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Preventing hemorrhage progression is a potential therapeutic opportunity in traumatic brain injury (TBI) management, but its use has been limited by fear of provoking vascular occlusive events (VOEs). However, it is currently unclear whether VOE actually affects outcome in these patients. The aim of this study was to determine incidence, risk factors, and clinical significance of VOE in patients with moderate-to-severe TBI. A retrospective observational cohort study of adults (≥15 years) with moderate-to-severe TBI was performed. The presence of a VOE during hospitalization was noted from hospital charts and radiological reports. Functional outcome, using the Glasgow Outcome Scale (GOS), was assessed at 12 months posttrauma. Univariate and multivariate logistic regressions were used for endpoint assessment. In total, 848 patients were included, with a median admission Glasgow Coma Scale of 7. A VOE was detected in 54 (6.4%) patients, of which cerebral venous thrombosis was the most common (3.2%), followed by pulmonary embolism (1.7%) and deep vein thrombosis (1.3%). Length of ICU stay (p < 0.001), body weight (p = 0.002), and skull fracture (p = 0.004) were independent predictors of VOE. VOE development did not significantly impact 12-month GOS, even after adjusting for potential confounders using propensity score matching. In conclusion, VOE in moderate-to-severe TBI patients was relatively uncommon, and did not affect 12-month GOS. This suggests that the potential benefit of treating bleeding progression might outweigh the risks of VOE.
Collapse
Affiliation(s)
- Alexander Fletcher-Sandersjöö
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Charles Tatter
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jonathan Tjerkaski
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jiri Bartek
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Neurosurgery, Rigshospitalet, Copenhagen, Denmark
| | - Mikael Svensson
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Eric Peter Thelin
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Bo-Michael Bellander
- Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
20
|
Dong X, Liu W, Shen Y, Houck K, Yang M, Zhou Y, Zhao Z, Wu X, Blevins T, Koehne AL, Wun TC, Fu X, Li M, Zhang J, Dong JF. Anticoagulation targeting membrane-bound anionic phospholipids improves outcomes of traumatic brain injury in mice. Blood 2021; 138:2714-2726. [PMID: 34610086 PMCID: PMC8703367 DOI: 10.1182/blood.2021011310] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
Severe traumatic brain injury (TBI) often causes an acute systemic hypercoagulable state that rapidly develops into consumptive coagulopathy. We have recently demonstrated that TBI-induced coagulopathy (TBI-IC) is initiated and disseminated by brain-derived extracellular vesicles (BDEVs) and propagated by extracellular vesicles (EVs) from endothelial cells and platelets. Here, we present results from a study designed to test the hypothesis that anticoagulation targeting anionic phospholipid-expressing EVs prevents TBI-IC and improves the outcomes of mice subjected to severe TBI. We evaluated the effects of a fusion protein (ANV-6L15) for improving the outcomes of TBI in mouse models combined with in vitro experiments. ANV-6L15 combines the phosphatidylserine (PS)-binding annexin V (ANV) with a peptide anticoagulant modified to preferentially target extrinsic coagulation. We found that ANV-6L15 reduced intracranial hematoma by 70.2%, improved neurological function, and reduced death by 56.8% in mice subjected to fluid percussion injury at 1.9 atm. It protected the TBI mice by preventing vascular leakage, tissue edema, and the TBI-induced hypercoagulable state. We further showed that the extrinsic tenase complex was formed on the surfaces of circulating EVs, with the highest level found on BDEVs. The phospholipidomic analysis detected the highest levels of PS on BDEVs, as compared with EVs from endothelial cells and platelets (79.1, 15.2, and 3.5 nM/mg of protein, respectively). These findings demonstrate that TBI-IC results from a trauma-induced hypercoagulable state and may be treated by anticoagulation targeting on the anionic phospholipid-expressing membrane of EVs from the brain and other cells.
Collapse
Affiliation(s)
- Xinlong Dong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Bloodworks Research Institute, Seattle, WA
| | - Wei Liu
- Institute of Pathology, School of Medical Sciences and Gansu Provincial Key Laboratory of Preclinical Study for New Drug Development, Lanzhou University, Lanzhou, China
| | - Yu Shen
- Bloodworks Research Institute, Seattle, WA
| | | | - Mengchen Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuan Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zilong Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoping Wu
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Teri Blevins
- Department of Comparative Medicine, Fred Hutch Cancer Center, Seattle, WA
| | - Amanda L Koehne
- Department of Comparative Medicine, Fred Hutch Cancer Center, Seattle, WA
| | | | - Xiaoyun Fu
- Bloodworks Research Institute, Seattle, WA
- Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, WA
| | - Min Li
- Institute of Pathology, School of Medical Sciences and Gansu Provincial Key Laboratory of Preclinical Study for New Drug Development, Lanzhou University, Lanzhou, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA
- Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, WA
| |
Collapse
|
21
|
Choudhary S, Sharma K, Singh PK. Von Willebrand factor: A key glycoprotein involved in thrombo-inflammatory complications of COVID-19. Chem Biol Interact 2021; 348:109657. [PMID: 34516971 PMCID: PMC8432980 DOI: 10.1016/j.cbi.2021.109657] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/25/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
COVID-19 is an ongoing public health emergency that has affected millions of people worldwide and is still a threat to many more. One of the pathophysiological features of COVID-19 is associated with the activation of vascular endothelial cells (ECs) leading to the disruption of vascular integrity, coagulation and inflammation. An interlink mechanism between coagulation and inflammatory pathways has been reported in COVID-19. Multiple components are involved in these pathological pathways. Out of all, Von Willebrand Factor (VWF) is one of the primary components of coagulation pathway and also a mediator of vascular inflammation that plays an important role in thrombo-inflammation that further leads to acute respiratory distress syndrome (ARDS). The thrombo-inflammatory co-morbidities such as hyper-coagulation, thrombosis, ARDS etc. have become the major cause of mortality in the patients of COVID-19 admitted to the ICU. Thus, VWF can be explored as a potential target to manage COVID-19 associated co-morbidities. Supporting this hypothesis, there are literature reports which disclose previous attempts to target VWF for the management of thrombo-inflammation in other pathological conditions. The current report summarizes emerging insights into the pathophysiology, mechanism(s), diagnosis, management and foundations for research on this less explored clinically relevant glycoprotein as coagulation biomarker in COVID-19.
Collapse
Affiliation(s)
- Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Kajal Sharma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Pankaj Kumar Singh
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.
| |
Collapse
|
22
|
Xu X, Gao W, Li L, Hao J, Yang B, Wang T, Li L, Bai X, Li F, Ren H, Zhang M, Zhang L, Wang J, Wang D, Zhang J, Jiao L. Annexin A1 protects against cerebral ischemia-reperfusion injury by modulating microglia/macrophage polarization via FPR2/ALX-dependent AMPK-mTOR pathway. J Neuroinflammation 2021; 18:119. [PMID: 34022892 PMCID: PMC8140477 DOI: 10.1186/s12974-021-02174-3] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023] Open
Abstract
Background Cerebral ischemia–reperfusion (I/R) injury is a major cause of early complications and unfavorable outcomes after endovascular thrombectomy (EVT) therapy in patients with acute ischemic stroke (AIS). Recent studies indicate that modulating microglia/macrophage polarization and subsequent inflammatory response may be a potential adjunct therapy to recanalization. Annexin A1 (ANXA1) exerts potent anti-inflammatory and pro-resolving properties in models of cerebral I/R injury. However, whether ANXA1 modulates post-I/R-induced microglia/macrophage polarization has not yet been fully elucidated. Methods We retrospectively collected blood samples from AIS patients who underwent successful recanalization by EVT and analyzed ANXA1 levels longitudinally before and after EVT and correlation between ANXA1 levels and 3-month clinical outcomes. We also established a C57BL/6J mouse model of transient middle cerebral artery occlusion/reperfusion (tMCAO/R) and an in vitro model of oxygen–glucose deprivation and reoxygenation (OGD/R) in BV2 microglia and HT22 neurons to explore the role of Ac2-26, a pharmacophore N-terminal peptide of ANXA1, in regulating the I/R-induced microglia/macrophage activation and polarization. Results The baseline levels of ANXA1 pre-EVT were significantly lower in 23 AIS patients, as compared with those of healthy controls. They were significantly increased to the levels found in controls 2–3 days post-EVT. The increased post-EVT levels of ANXA1 were positively correlated with 3-month clinical outcomes. In the mouse model, we then found that Ac2-26 administered at the start of reperfusion shifted microglia/macrophage polarization toward anti-inflammatory M2-phenotype in ischemic penumbra, thus alleviating blood–brain barrier leakage and neuronal apoptosis and improving outcomes at 3 days post-tMCAO/R. The protection was abrogated when mice received Ac2-26 together with WRW4, which is a specific antagonist of formyl peptide receptor type 2/lipoxin A4 receptor (FPR2/ALX). Furthermore, the interaction between Ac2-26 and FPR2/ALX receptor activated the 5’ adenosine monophosphate-activated protein kinase (AMPK) and inhibited the downstream mammalian target of rapamycin (mTOR). These in vivo findings were validated through in vitro experiments. Conclusions Ac2-26 modulates microglial/macrophage polarization and alleviates subsequent cerebral inflammation by regulating the FPR2/ALX-dependent AMPK-mTOR pathway. It may be investigated as an adjunct strategy for clinical prevention and treatment of cerebral I/R injury after recanalization. Plasma ANXA1 may be a potential biomarker for outcomes of AIS patients receiving EVT. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02174-3.
Collapse
Affiliation(s)
- Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China. .,China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China.
| | - Weiwei Gao
- Department of Neurology, Tianjin Huanhu Hospital, 6 Jizhao Road, Tianjin, 300350, China.
| | - Lei Li
- Department of Neurosurgery & Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China
| | - Jiheng Hao
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, 252000, China
| | - Bin Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.,China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.,China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Long Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.,China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Xuesong Bai
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.,China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China
| | - Fanjian Li
- Department of Neurosurgery & Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China
| | - Honglei Ren
- Department of Neurosurgery & Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China
| | - Meng Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, 252000, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, 252000, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, 252000, China
| | - Dong Wang
- Department of Neurosurgery & Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China
| | - Jianning Zhang
- Department of Neurosurgery & Neurology, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China. .,China International Neuroscience Institute (China-INI), 45 Changchun Street, Beijing, 100053, China. .,Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
| |
Collapse
|
23
|
Xu X, Kozar R, Zhang J, Dong JF. Diverse activities of von Willebrand factor in traumatic brain injury and associated coagulopathy. J Thromb Haemost 2020; 18:3154-3162. [PMID: 32931638 PMCID: PMC7855263 DOI: 10.1111/jth.15096] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability. Patients with isolated TBI lose a limited amount of blood to primary injury, but they often develop secondary coagulopathy, resulting in delayed or recurrent intracranial and intracerebral hematoma. TBI-induced coagulopathy is closely associated with poor outcomes for these patients, including death. This secondary coagulopathy is consumptive in nature, involving not only brain-derived molecules, coagulation factors, and platelets, but also endothelial cells in a complex process now called blood failture. A key question is how a localized injury to the brain is rapidly disseminated to affect systemic hemostasis that is not directly affected the way it is in trauma to the body and limbs, especially with hemorrhagic shock. Increasing evidence suggests that the adhesive ligand von Willebrand factor (VWF), which is synthesized in and released from endothelial cells, plays a paradoxical role in both facilitating local hemostasis at the site of injury and also propagating TBI-induced endotheliopathy and coagulopathy systemically. This review discusses recent progress in understanding these diverse activities of VWF and the knowledge gaps in defining their roles in TBI and associated coagulopathy.
Collapse
Affiliation(s)
- Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Rosemary Kozar
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, US
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Neurology, Tianjin, China
| | - Jing-fei Dong
- Bloodworks Research Institute, Seattle, WA, US
- Hematology Division, Department of Medicine, University of Washington School of Medicine, Seattle, WA, US
| |
Collapse
|
24
|
Hubbard WB, Dong JF, Cruz MA, Rumbaut RE. Links between thrombosis and inflammation in traumatic brain injury. Thromb Res 2020; 198:62-71. [PMID: 33290884 DOI: 10.1016/j.thromres.2020.10.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/20/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) continues to be a major healthcare problem and there is much to be explored regarding the secondary pathobiology to identify early predictive markers and new therapeutic targets. While documented changes in thrombosis and inflammation in major trauma have been well described, growing evidence suggests that isolated TBI also results in systemic alterations in these mechanisms. Here, we review recent experimental and clinical findings that demonstrate how blood-brain barrier dysfunction, systemic immune response, inflammation, platelet activation, and thrombosis contribute significantly to the pathogenesis of TBI. Despite advances in the links between thrombosis and inflammation, there is a lack of treatment options aimed at both processes and this could be crucial to treating vascular injury, local and systemic inflammation, and secondary ischemic events following TBI. With emerging evidence of newly-identified roles for platelets, leukocytes, the coagulation system and extracellular vesicles in processes of inflammation and thrombosis, there is a growing need to characterize these mechanisms within the context of TBI and whether these changes persist into the chronic phase of injury. Importantly, this review defines areas in need of further research to advance the field and presents a roadmap to identify new diagnostic and treatment options for TBI.
Collapse
Affiliation(s)
- W Brad Hubbard
- Lexington VA Healthcare System, Lexington, KY, United States of America; Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY, United States of America.
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA, United States of America; Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Miguel A Cruz
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX, United States of America; Baylor College of Medicine, Houston, TX, United States of America
| | - Rolando E Rumbaut
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX, United States of America; Baylor College of Medicine, Houston, TX, United States of America
| |
Collapse
|