1
|
Hicks AJ, Carrington H, Bura L, Yang A, Pesce R, Yew B, Dams-O'Connor K. Blood-Based Protein Biomarkers in the Chronic Phase of Traumatic Brain Injury: A Systematic Review. J Neurotrauma 2025; 42:759-797. [PMID: 40176450 DOI: 10.1089/neu.2024.0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
There has been limited exploration of blood-based biomarkers in the chronic period following traumatic brain injury (TBI). Our objective was to conduct a systematic review of studies examining blood-based protein biomarkers with at least one sample collected 12 months post-TBI in adults (≥16 years). Database searches were conducted in Embase, MEDLINE, and Science Citation Index-Expanded on July 24, 2023. Risk of bias was assessed using modified Joanna Briggs Institute critical appraisal tools. Only 30 of 12,523 articles met inclusion criteria, with samples drawn from 12 months to 48 years. Higher quality evidence (low risk of bias; large samples) identified promising inflammatory biomarkers at 12 months post-injury in both moderate-severe TBI (GFAP) and mild TBI (eotaxin-1, IFN-y, IL-8, IL-9, IL-17A, MCP-1, MIP-1β, FGF-basic, and TNF-α). Studies with low risk of bias but smaller samples also suggest NSE, MME, and CRP may be informative, alongside protein variants for α-syn (10H, D5), amyloid-β (A4, C6T), TDP-43 (AD-TDP 1;2;3;9;11), and tau (D11C). Findings for NfL were inconclusive. Longitudinal data were mostly available for acute samples followed until 12 months post-injury, with limited evaluation of changes beyond 12 months. Associations of some blood-based biomarkers with cognitive, sleep, and functional outcomes were reported. The overall strength of the evidence in this review was limited by the risk of bias and small sample sizes. Replication is required within prospective longitudinal studies that move beyond 12 months post-injury. Novel efforts should be guided by promising neurodegenerative-disease markers and use panels to model polypathology.
Collapse
Affiliation(s)
- Amelia J Hicks
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Holly Carrington
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lisa Bura
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alicia Yang
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rico Pesce
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Belinda Yew
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | |
Collapse
|
2
|
Wang M, Zhao R, Su Y, Zhai D, Liang H, Zhang L, Wang W, Wang Z, Qi M, Jiang X, Ling S, Di G. 4,4'-Dimethoxychalcone Mitigates Neuroinflammation Following Traumatic Brain Injury Through Modulation of the TREM2/PI3K/AKT/NF-κB Signaling Pathway. Inflammation 2025:10.1007/s10753-025-02279-4. [PMID: 40261458 DOI: 10.1007/s10753-025-02279-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/24/2025]
Abstract
Research on 4,4'-dimethoxychalcone (DMC) in the context of traumatic brain injury (TBI) is extremely limited, and no effective clinical treatments are available to improve outcomes for individuals with TBI. Our study aims to investigate the underlying mechanisms by which DMC may alleviate neuroinflammation and neuronal damage following TBI. This study seeks to provide a theoretical foundation for the development of future pharmacological therapies for TBI. A moderate TBI model was established using the fluid percussion injury (FPI) method. The recovery of neuromotor function following TBI was evaluated using the modified neurological severity score (mNSS), the Morris water maze test, and analysis of cerebral edema. Gene and protein expression levels were quantified using cell viability assays, quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and immunofluorescence. Network pharmacology was employed to predict potential targets of DMC, and gene ontology (GO) analysis along with KEGG pathway enrichment was conducted to predict signaling pathways affected by DMC.DMC treatment significantly improved neuromotor deficits in mice after TBI. In both in vivo and in vitro experiments, DMC suppressed microglial activation and decreased the production and release of inflammatory factors. Additionally, DMC reduced neuronal lesions after TBI. DMC notably decreased the elevated expression of triggering receptor expressed on myeloid cells 2 (TREM2) following TBI. Network pharmacological analysis indicated that DMC's therapeutic effects may be mediated through the PI3K/AKT signaling cascade. These findings indicate that DMC has therapeutic potential for TBI, with significant anti-inflammatory and neuroprotective properties likely mediated by the TREM2/PI3K/AKT/NF-κB signaling cascade.
Collapse
Affiliation(s)
- Mengran Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Rui Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Yue Su
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Duhuan Zhai
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Hengyan Liang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Lingkun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Weicheng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Zhichun Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Min Qi
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China
| | - Xiaochun Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China.
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
| | - Shizhang Ling
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China.
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
| | - Guangfu Di
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
- The Institutes of Brain Science, Wannan Medical College, No. 22 Wenchang West Road, Higher Education Park, Wuhu, 241002, Anhui, China.
- Department of Neurosurgery, The Translational Research Institute for Neurological Disorders of Wannan Medical College, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan Road, Wuhu, 241001, Anhui, China.
| |
Collapse
|
3
|
Chen GH, Sia KC, Liu SW, Kao YC, Yang PC, Ho CH, Huang SC, Lee PY, Liang MZ, Chen L, Huang CC. Implantation of MSC spheroid-derived 3D decellularized ECM enriched with the MSC secretome ameliorates traumatic brain injury and promotes brain repair. Biomaterials 2025; 315:122941. [PMID: 39515193 DOI: 10.1016/j.biomaterials.2024.122941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Traumatic brain injury (TBI) presents substantial clinical challenges, as existing treatments are unable to reverse damage or effectively promote brain tissue regeneration. Although implantable biomaterials have been proposed to support tissue repair by mitigating the adverse microenvironment in injured brains, many fail to replicate the complex composition and architecture of the native extracellular matrix (ECM), resulting in only limited therapeutic outcomes. This study introduces an innovative approach by developing a mesenchymal stem cell (MSC) spheroid-derived three-dimensional (3D) decellularized ECM (dECM) that is enriched with the MSC-derived matrisome and secretome, offering a promising solution for TBI treatment and brain tissue regeneration. Proteomic and cytokine array analyses revealed that 3D dECM retained a diverse array of MSC spheroid-derived matrisome proteins and secretome components, which are crucial for replicating the complexity of native ECM and the therapeutic capabilities of MSCs. These molecules were found to underlie the observed effects of 3D dECM on immunomodulation, proneuritogenesis, and proangiogenesis in our in vitro functional assays. Implantation of 3D dECM into TBI model mice effectively mitigated postinjury tissue damage and promoted brain repair, as evidenced by a reduced brain lesion volume, decreased cell apoptosis, alleviated neuroinflammation, reduced glial scar formation, and increased of neuroblast recruitment to the lesion site. These outcomes culminated in improved motor function recovery in animals, highlighting the multifaceted therapeutic potential of 3D dECM for TBI. In summary, our study elucidates the transformative potential of MSC spheroid-derived bioactive 3D dECM as an implantable biomaterial for effectively mitigating post-TBI neurological damage, paving the way for its broader therapeutic application.
Collapse
Affiliation(s)
- Grace H Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kee-Chin Sia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ying-Chi Kao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Pei-Ching Yang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chia-Hsin Ho
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shih-Chen Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Peng-Ying Lee
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Min-Zong Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
4
|
Eslami M, Raji-Amirhasani A, Khaksari M, Keshavarzi Z, Rostamzadeh F, Sabet N, Jafari E, Soltani Z, Karamouzian S. The changes of digestive system inflammatory, oxidative stress, and histopathology factors following oral mesenchymal stem cells administration in rats with traumatic brain injury. BMC Neurosci 2025; 26:20. [PMID: 40050727 PMCID: PMC11884162 DOI: 10.1186/s12868-025-00936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/10/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND AND AIMS Mucous mesenchymal stem cells can migrate to damaged areas, and their use is proposed as a new approach to treating diseases. The present study aimed to investigate the effect of oral mesenchymal stem cells (OMSCs) on inflammatory, oxidative stress, and histopathological indices in the tissues of the stomach, intestine, and colon after traumatic brain injury (TBI). METHODS AND MATERIALS Adult male rats were randomly divided into four groups: Sham, TBI, Vehicle (Veh), and Stem cell (SC). Intravenous injection of OMSCs was performed at 1 and 24 h after injury. The inflammatory, oxidative stress, and histopathological indices of the tissues of the stomach, small intestine, and colon were evaluated 48 h after injury. RESULTS After TBI, IL-1β and IL-6 levels increased and IL-10 levels decreased in the tissues of the stomach, small intestine, and colon, but the administration of OMSCS prevented these changes to a large extent. Oxidative stress indices (MDA, PC, TAC, SOD, and CAT) showed an increase in oxidative stress after TBI, but oxidative stress was less severe in the OMSC group. The administration of OMSCs after TBI improved the histopathological outcome in the tissues of the stomach, small intestine, and colon. CONCLUSION Administration of OMSCs in rats suffering from TBI can improve inflammatory, oxidative stress, and histopathological indices in the tissues of the stomach, small intestine, and colon, which shows the beneficial effect of using OMSCs in TBI.
Collapse
Affiliation(s)
- Masoud Eslami
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Raji-Amirhasani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zakieh Keshavarzi
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Farzaneh Rostamzadeh
- Cardiovascular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Nazanin Sabet
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Jafari
- Pathology and Stem Cells Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Saeed Karamouzian
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
5
|
Fu X, Zhang Y, Chen G, Mao G, Tang J, Xu J, Han Y, Chen H, Ding L. Responsive nanoparticles synergize with Curcumin to break the "reactive oxygen Species-Neuroinflammation" vicious cycle, enhancing traumatic brain injury outcomes. J Nanobiotechnology 2025; 23:172. [PMID: 40045354 PMCID: PMC11881390 DOI: 10.1186/s12951-025-03251-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
Traumatic brain injury (TBI) disrupts oxygen homeostasis in the brain, leading to excessive reactive oxygen species (ROS) production and dysregulated antioxidant mechanisms, which fail to clear excess ROS. This ROS overload promotes the expression of pro-inflammatory genes, releasing cytokines and chemokines and creating a vicious "ROS-neuroinflammation" cycle, making it essential to break this cycle for effective TBI treatment. In this study, we developed cysteine-alanine-glutamine-lysine (CAQK) peptide-modified antioxidant nanoparticles (C-PPS/C) for co-delivery of curcumin (Cur) to modulate oxidative and neuroinflammatory disturbances after TBI. In TBI mice, C-PPS/C nanoparticles accumulated in injured brain regions, where poly (propylene sulfide)120 scavenged ROS, reducing oxidative stress, while Cur release further suppressed ROS and inflammation. C-PPS/C nanoparticles broke the "ROS-neuroinflammation" cycle, protecting the blood-brain barrier (BBB), reducing acute brain edema, and promoting long-term neurological recovery. Further investigation showed that C-PPS/C nanoparticles inhibited the NF-κB pathway, reducing pro-inflammatory gene expression and mitigating inflammation, suggesting a promising approach for TBI treatment.
Collapse
Affiliation(s)
- Xianhua Fu
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Yongkang Zhang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guojie Chen
- Clinical Laboratory, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Guangyao Mao
- Clinical Laboratory, Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Jiajia Tang
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Jin Xu
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Yuhan Han
- Brain Injury Center, Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Honglin Chen
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China.
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China.
| |
Collapse
|
6
|
Wang J, Zhang H, Augenreich M, Martinez-Lemus A L, Liu Z, Kang X, Lu B, Chang HM, Yeh ETH, Cata J, Rangaraju S, Wulff H, Li DP. Microglia-Mediated Synaptic Dysfunction Contributes to Chemotherapy-Related Cognitive Impairment. J Neurochem 2025; 169:e70024. [PMID: 40019120 PMCID: PMC11927766 DOI: 10.1111/jnc.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Chemotherapy-related cognitive impairment (CRCI) significantly impacts cancer survivors. Due to unclear mechanisms, effective treatments for cognitive deficits are lacking. Here, we examined if microglia-mediated deficits in synaptic plasticity drive CRCI. Adult male mice were treated with the chemotherapeutic drugs 5-fluorouracil and leucovorin (5-Fu/LV, intraperitoneal injection, I.P.) on Days 1, 8, and 15 at a dosage of 50 mg/kg for 5-Fu and 90 mg/kg for LV for 3 weeks. Cognitive function was assessed using a novel object recognition (NOR) test 4 weeks after completion of 5-Fu/LV treatment. Compared with vehicle treatment, 5-Fu/LV treatment reduced the preference for exploring novel objects in the NOR test. Treatment with 5-Fu/LV increased the numbers of Iba1-positive microglial and CD68-positive/Iba1-positive microglia with shortened process lengths and diminished endpoints but decreased the number of phagocytotic (≤ 1 FITC-labeled beads) Iba1-positive microglia. Furthermore, 5-Fu/LV treatment reduced the long-term potentiation (LTP) recorded in the hippocampal CA1 region in response to a theta burst stimulation of the CA3-CA1 pathway and decreased the evoked N-methyl-D-aspartic acid receptor (NMDAR)-excitatory postsynaptic currents (NMDAR-EPSCs) in CA1 neurons. Cotreatment with the microglial inhibitor minocycline (33 mg/kg, daily for 3 weeks) restored cognitive deficits and microglial ramification, decreased the number of CD68-positive microglia, and reversed the reductions in LTP and the amplitude of NMDAR-EPSCs in 5-Fu/LV-treated mice. Our data suggest that microglial dysfunction and related synaptic dysfunction contribute to 5-Fu/LV-induced cognitive impairment.
Collapse
Affiliation(s)
- Jingxiong Wang
- Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Hua Zhang
- Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Marc Augenreich
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Luis Martinez-Lemus A
- Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Zhenguo Liu
- Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Xunlei Kang
- Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Bo Lu
- Department of Radiation Oncology, NextGen Precision Health, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| | - Hui-Ming Chang
- Departments of Pharmacology and Internal Medicine, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Edward T H Yeh
- Department of Internal Medicine, The University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Juan Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Srikant Rangaraju
- Department of Neurology, Yale University School of Medicine, New Heaven, Connecticut, USA
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - De-Pei Li
- Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, Missouri, USA
| |
Collapse
|
7
|
Han S, Yoo W, Carton O, Joo J, Kwon EJ. PEGylated Multimeric RNA Nanoparticles for siRNA Delivery in Traumatic Brain Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405806. [PMID: 39498752 PMCID: PMC11899522 DOI: 10.1002/smll.202405806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/27/2024] [Indexed: 11/07/2024]
Abstract
Traumatic brain injury (TBI) impacts millions of people globally, however currently there are no approved therapeutics that address long-term brain health. In order to create a technology that is relevant for siRNA delivery in TBI after systemic administration, sub-100 nm nanoparticles with rolling circle transcription (RCT) are synthesized and isolated in order improve payload delivery into the injured brain. Unlike conventional RCT-based RNA particles, in this method, sub-100 nm RNA nanoparticles (RNPs) are isolated. To enhance RNP pharmacokinetics, RNPs are synthesized with modified bases in order to graft polyethylene glycol (PEG) to the RNPs. PEGylated RNPs (PEG-RNPs) do not significantly impact their knockdown activity in vitro and lead to longer blood half-life after systemic administration and greater accumulation into the injured brain in a mouse model of TBI. In order to demonstrate RNA interference (RNAi) activity of RNPs, knockdown of the inflammatory cytokine TNF-α in injured brain tissue after systemic administration of RNPs in a mouse model of TBI is demonstrated. In summary, small sub-100 nm multimeric RNA nanoparticles are synthesized and isolated that can be modified using accessible chemistry in order to create a technology suitable for systemic RNAi therapy for TBI.
Collapse
Affiliation(s)
- Sangwoo Han
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Woojung Yoo
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
| | - Olivia Carton
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
| | - Jinmyoung Joo
- Department of Biomedical EngineeringUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Graduate School of Health Science and TechnologyUlsan National Institute of Science and Technology (UNIST)Ulsan44919Republic of Korea
- Center for Genomic IntegrityInstitute for Basic ScienceUlsan44919Republic of Korea
- Materials Research Science and Engineering CenterUniversity of California San DiegoLa JollaCA92093USA
| | - Ester J. Kwon
- Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Materials Research Science and Engineering CenterUniversity of California San DiegoLa JollaCA92093USA
- Sanford Consortium for Regenerative MedicineLa JollaCA92037USA
| |
Collapse
|
8
|
Migdady I, Gusdon AM, Everett AD, Cho SM. Blood and cerebrospinal fluid biomarkers in disorders of consciousness. HANDBOOK OF CLINICAL NEUROLOGY 2025; 207:165-181. [PMID: 39986720 DOI: 10.1016/b978-0-443-13408-1.00006-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
The study of blood and cerebrospinal fluid biomarkers is a promising and rapidly advancing field in the research of disorders of consciousness (DoC). The use of advanced biochemical and analytic techniques in biomarker research has improved our ability to identify new biomarkers that can aid in the diagnosis, prognosis, and treatment of patients with brain injury. However, the use of biomarkers in clinical practice is limited by several challenges, including the lack of standardization in test and research methodologies. Despite this, identifying the most promising biomarkers and supporting their findings with strong evidence can improve their clinical utility. This chapter discusses the most promising biomarkers for DoC, which fall into four categories: neuronal, glial, inflammatory, and metabolic biomarkers. Understanding the role of each category in DoC can provide valuable insights into the mechanisms of brain injury and inform the development of more effective diagnostic and treatment strategies. By integrating biomarker research with clinical practice, we can improve our understanding of DoC and provide better care for these patients.
Collapse
Affiliation(s)
- Ibrahim Migdady
- Departments of Neurology, Medicine and Neurosurgery, Division of Critical Care Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
| | - Aaron M Gusdon
- Department of Neurosurgery, McGovern Medical School at UTHealth Houston, Houston, TX, United States
| | - Allen D Everett
- Division of Pediatric Cardiology, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Sung-Min Cho
- Departments of Neurology, Neurosurgery, Surgery, and Anesthesia/Critical Care, Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
9
|
Goyal L, Singh S. Neurological Manifestations Following Traumatic Brain Injury: Role of Behavioral, Neuroinflammation, Excitotoxicity, Nrf-2 and Nitric Oxide. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:47-59. [PMID: 39082170 DOI: 10.2174/0118715273318552240708055413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 01/31/2025]
Abstract
Traumatic Brain Injury (TBI) is attributed to a forceful impact on the brain caused by sharp, penetrating bodies, like bullets and any sharp object. Some popular instances like falls, traffic accidents, physical assaults, and athletic injuries frequently cause TBI. TBI is the primary cause of both mortality and disability among young children and adults. Several individuals experience psychiatric problems, including cognitive dysfunction, depression, post-traumatic stress disorder, and anxiety, after primary injury. Behavioral changes post TBI include cognitive deficits and emotional instability (anxiety, depression, and post-traumatic stress disorder). These alterations are linked to neuroinflammatory processes. On the other hand, the direct impact mitigates inflammation insult by the release of pro-inflammatory cytokines, namely IL-1β, IL-6, and TNF-α, exacerbating neuronal injury and contributing to neurodegeneration. During the excitotoxic phase, activation of glutamate subunits like NMDA enhances the influx of Ca2+ and leads to mitochondrial metabolic impairment and calpain-mediated cytoskeletal disassembly. TBI pathological insult is also linked to transcriptional response suppression Nrf-2, which plays a critical role against TBI-induced oxidative stress. Activation of NRF-2 enhances the expression of anti-oxidant enzymes, providing neuroprotection. A possible explanation for the elevated levels of NO is that the stimulation of NMDA receptors by glutamate leads to the influx of calcium in the postsynaptic region, activating NOS's constitutive isoforms.
Collapse
Affiliation(s)
- Lav Goyal
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| |
Collapse
|
10
|
Liang S, Hu Z. Unveiling the predictive power of biomarkers in traumatic brain injury: A narrative review focused on clinical outcomes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2024. [PMID: 39687991 DOI: 10.5507/bp.2024.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
Traumatic brain injury (TBI) has long-term consequences, including neurodegenerative disease risk. Current diagnostic tools are limited in detecting subtle brain damage. This review explores emerging biomarkers for TBI, including those related to neuronal injury, inflammation, EVs, and ncRNAs, evaluating their potential to predict clinical outcomes like mortality, recovery, and cognitive impairment. It addresses challenges and opportunities for implementing biomarkers in clinical practice, aiming to improve TBI diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Sitao Liang
- Neurosurgery Department, Zhongshan City People's Hospital, Zhongshan, 528400, China
| | - Zihui Hu
- Neurosurgery Department, Zhongshan City People's Hospital, Zhongshan, 528400, China
| |
Collapse
|
11
|
Pasam T, Padhy HP, Dandekar MP. Lactobacillus Helveticus Improves Controlled Cortical Impact Injury-Generated Neurological Aberrations by Remodeling of Gut-Brain Axis Mediators. Neurochem Res 2024; 50:3. [PMID: 39541016 DOI: 10.1007/s11064-024-04251-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/15/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Considerable studies augured the potential of gut microbiota-based interventions in brain injury-associated complications. Based on our earlier study results, we envisaged the sex-specific neuroprotective effect of Lactobacillus helveticus by remodeling of gut-brain axis. In this study, we investigated the effect of L. helveticus on neurological complications in a mouse model of controlled cortical impact (CCI). Adult, male and female, C57BL/6 mice underwent CCI surgery and received L. helveticus treatment for six weeks. Sensorimotor function was evaluated via neurological severity score and rotarod test. Long-term effects on anxiety-like behavior and cognition were assessed using the elevated-zero maze (EZM) and novel object recognition test (NORT). Brain perilesional area, blood, colon, and fecal samples were collected post-CCI for molecular biology analysis. CCI-operated mice displayed significant neurological impairments at 1-, 3-, 5-, and 7-days post-injury (dpi) and exhibited altered behavior in EZM and NORT compared to sham-operated mice. However, these behavioral changes were ameliorated in mice receiving L. helveticus. GFAP, Iba-1, TNF-α, and IL-1β expressions and corticotrophin-releasing hormone (CRH) levels were elevated in the perilesional cortex of CCI-operated male/female mice. These elevated biomarkers and decreased BDNF levels in both male/female mice were modified by L. helveticus treatment. Additionally, L. helveticus treatment restored altered short-chain fatty acids (SCFAs) levels in fecal samples and improved intestinal integrity but did not affect decreased plasma levels of progesterone and testosterone in CCI mice. These results indicate that L. helveticus exerts beneficial effects in the CCI mouse model by mitigating inflammation and remodeling of gut microbiota-brain mediators.
Collapse
Affiliation(s)
- Tulasi Pasam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Manoj P Dandekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
12
|
Wehn AC, Khalin I, Hu S, Harapan BN, Mao X, Cheng S, Plesnila N, Terpolilli NA. Bradykinin 2 Receptors Mediate Long-Term Neurocognitive Deficits After Experimental Traumatic Brain Injury. J Neurotrauma 2024; 41:2442-2454. [PMID: 38818807 DOI: 10.1089/neu.2024.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
The kallikrein-kinin system is one of the first inflammatory pathways to be activated following traumatic brain injury (TBI) and has been shown to exacerbate brain edema formation in the acute phase through activation of bradykinin 2 receptors (B2R). However, the influence of B2R on chronic post-traumatic damage and outcome is unclear. In the current study, we assessed long-term effects of B2R-knockout (KO) after experimental TBI. B2R KO mice (heterozygous, homozygous) and wild-type (WT) littermates (n = 10/group) were subjected to controlled cortical impact (CCI) TBI. Lesion size was evaluated by magnetic resonance imaging up to 90 days after CCI. Motor and memory function were regularly assessed by Neurological Severity Score, Beam Walk, and Barnes maze test. Ninety days after TBI, brains were harvested for immunohistochemical analysis. There was no difference in cortical lesion size between B2R-deficient and WT animals 3 months after injury; however, hippocampal damage was reduced in B2R KO mice (p = 0.03). Protection of hippocampal tissue was accompanied by a significant improvement of learning and memory function 3 months after TBI (p = 0.02 WT vs. KO), whereas motor function was not influenced. Scar formation and astrogliosis were unaffected, but B2R deficiency led to a gene-dose-dependent attenuation of microglial activation and a reduction of CD45+ cells 3 months after TBI in cortex (p = 0.0003) and hippocampus (p < 0.0001). These results suggest that chronic hippocampal neurodegeneration and subsequent cognitive impairment are mediated by prolonged neuroinflammation and B2R. Inhibition of B2R may therefore represent a novel strategy to reduce long-term neurocognitive deficits after TBI.
Collapse
Affiliation(s)
- Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Rouen, France
| | - Senbin Hu
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Biyan Nathanael Harapan
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Xiang Mao
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Neurotrauma Laboratory, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shiqi Cheng
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, The Second affiliated Hospital of Nanchang University, Nanchang, China
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicole A Terpolilli
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
13
|
Calderone A, Latella D, Cardile D, Gangemi A, Corallo F, Rifici C, Quartarone A, Calabrò RS. The Role of Neuroinflammation in Shaping Neuroplasticity and Recovery Outcomes Following Traumatic Brain Injury: A Systematic Review. Int J Mol Sci 2024; 25:11708. [PMID: 39519259 PMCID: PMC11546226 DOI: 10.3390/ijms252111708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroplasticity and neuroinflammation are variables seen during recovery from traumatic brain injury (TBI), while biomarkers are useful in monitoring injury and guiding rehabilitation efforts. This systematic review examines how neuroinflammation affects neuroplasticity and recovery following TBI in animal models and humans. Studies were identified from an online search of the PubMed, Web of Science, and Embase databases without any search time range. This review has been registered on Open OSF (n) UDWQM. Recent studies highlight the critical role of biomarkers like serum amyloid A1 (SAA1) and Toll-like receptor 4 (TLR4) in predicting TBI patients' injury severity and recovery outcomes, offering the potential for personalized treatment and improved neurorehabilitation strategies. Additionally, insights from animal studies reveal how neuroinflammation affects recovery, emphasizing targets such as NOD-like receptor family pyrin domain-containing 3 (NLRP3) and microglia for enhancing therapeutic interventions. This review emphasizes the central role of neuroinflammation in TBI, and its adverse impact on neuroplasticity and recovery, and suggests that targeted anti-inflammatory treatments and biomarker-based personalized approaches hold the key to improvement. Such approaches will need further development in future research by integrating neuromodulation and pharmacological interventions, along with biomarker validation, to optimize management in TBI.
Collapse
Affiliation(s)
- Andrea Calderone
- Department of Clinical and Experimental Medicine, University of Messina, Piazza Pugliatti 1, 98122 Messina, Italy
| | - Desirèe Latella
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Davide Cardile
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Antonio Gangemi
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Francesco Corallo
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Carmela Rifici
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Angelo Quartarone
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| | - Rocco Salvatore Calabrò
- IRCCS Centro Neurolesi Bonino-Pulejo, S.S. 113 Via Palermo, C.da Casazza, 98124 Messina, Italy
| |
Collapse
|
14
|
Joseph CR. Assessing Mild Traumatic Brain Injury-Associated Blood-Brain Barrier (BBB) Damage and Restoration Using Late-Phase Perfusion Analysis by 3D ASL MRI: Implications for Predicting Progressive Brain Injury in a Focused Review. Int J Mol Sci 2024; 25:11522. [PMID: 39519073 PMCID: PMC11547134 DOI: 10.3390/ijms252111522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Mild traumatic brain injury (mTBI) is a common occurrence around the world, associated with a variety of blunt force and torsion injuries affecting all age groups. Most never reach medical attention, and the identification of acute injury and later clearance to return to usual activities is relegated to clinical evaluation-particularly in sports injuries. Advanced structural imaging is rarely performed due to the usual absence of associated acute anatomic/hemorrhagic changes. This review targets physiologic imaging techniques available to identify subtle blood-brain barrier dysfunction and white matter tract shear injury and their association with chronic traumatic encephalopathy. These techniques provide needed objective measures to assure recovery from injury in those patients with persistent cognitive/emotional symptoms and in the face of repetitive mTBI.
Collapse
Affiliation(s)
- Charles R Joseph
- Department of Neurology and Internal Medicine, College of Osteopathic Medicine, Liberty University, Lynchburg, VA 24502, USA
| |
Collapse
|
15
|
Chen Z, Ford KP, Islam MBAR, Wan H, Han H, Ramakrishnan A, Brown RJ, Villanueva V, Wang Y, Davis BT, Weiss C, Cui W, Gate D, Schwulst SJ. Anti-CD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8 + T-cell cytotoxicity after traumatic brain injury. J Neuroinflammation 2024; 21:267. [PMID: 39427160 PMCID: PMC11491007 DOI: 10.1186/s12974-024-03257-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people.
Collapse
Affiliation(s)
- Zhangying Chen
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Kacie P Ford
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mecca B A R Islam
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hanxiao Wan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hyebin Han
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Abhirami Ramakrishnan
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ryan J Brown
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Veronica Villanueva
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yidan Wang
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Booker T Davis
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Craig Weiss
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Weiguo Cui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Steven J Schwulst
- Department of Surgery, Division of Trauma and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
16
|
Baucom MR, Wallen TE, Price AD, England LG, Schuster RM, Goodman MD. Tranexamic Acid Administration Does Not Alter Inflammation After Traumatic Brain Injury, Regardless of Timing. J Surg Res 2024; 302:106-115. [PMID: 39094257 DOI: 10.1016/j.jss.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/15/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Tranexamic acid (TXA) administered early after traumatic brain injury (TBI) can decrease morbidity and mortality. The purpose of this study is to determine if the timing of TXA administration after TBI affects postinjury inflammatory markers or phosphorylated tau (p-tau) levels within the hippocampus. METHODS Male mice (9-11 wk) were split into six groups based on injury and timing of TXA administration (n = 5 per group): Sham, TBI-only, 100 mg/kg TXA-only, TBI + TXA 10 min, TBI + TXA 1 h, and TBI + TXA 6 h. Moderate concussive TBI was induced via weight drop. Serum and brain homogenates were collected at 6 and 24 h postinjury and analyzed for 14 inflammatory cytokines via multiplex enzyme-linked immunosorbent assay. Serum was analyzed for glial fibrillary acidic protein levels. Additional cohorts were survived to 30 d for hippocampal p-tau quantification using immunohistochemistry. RESULTS Serum levels of interleukin (IL) 1β (IL-1β), IL-3, IL-12, IL-17, monocyte chemoattractant protein-1, granulocyte-macrophage colony-stimulating factor, and regulated on activation, normal T-cell expressed and secreted were elevated in TBI mice compared to sham mice at 24 h. Levels of IL-1β and monocyte chemoattractant protein-1 were lower in 6-h TXA-treated mice than 1-h TXA-treated mice following TBI. IL-12 and macrophage inflammatory protein-1α levels were decreased in 6-h TXA-treated mice compared to 10-min TXA-treated mice. Administration of TXA at 10 min and 6 h but not 1 h postTBI reduced serum glial fibrillary acidic protein levels compared to TBI-only mice. Hippocampal p-tau accumulation was increased after TBI but not reduced by TXA administration. CONCLUSIONS Our results demonstrate that neither early nor delayed administration of TXA conveyed significant systemic or cerebral benefit in cytokine levels following TBI. Further research should be conducted to assess blood brain barrier integrity and neurobehavioral recovery following TXA administration postTBI.
Collapse
Affiliation(s)
- Matthew R Baucom
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Taylor E Wallen
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Adam D Price
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Lisa G England
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | | |
Collapse
|
17
|
Shaw BC, Anders VR, Tinkey RA, Habean ML, Brock OD, Frostino BJ, Williams JL. Immunity impacts cognitive deficits across neurological disorders. J Neurochem 2024; 168:3512-3535. [PMID: 37899543 PMCID: PMC11056485 DOI: 10.1111/jnc.15999] [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: 08/01/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023]
Abstract
Cognitive deficits are a common comorbidity with neurological disorders and normal aging. Inflammation is associated with multiple diseases including classical neurodegenerative dementias such as Alzheimer's disease (AD) and autoimmune disorders such as multiple sclerosis (MS), in which over half of all patients experience some form of cognitive deficits. Other degenerative diseases of the central nervous system (CNS) including frontotemporal lobe dementia (FTLD), and Parkinson's disease (PD) as well as traumatic brain injury (TBI) and psychological disorders like major depressive disorder (MDD), and even normal aging all have cytokine-associated reductions in cognitive function. Thus, there is likely commonality between these secondary cognitive deficits and inflammation. Neurological disorders are increasingly associated with substantial neuroinflammation, in which CNS-resident cells secrete cytokines and chemokines such as tumor necrosis factor (TNF)α and interleukins (ILs) including IL-1β and IL-6. CNS-resident cells also respond to a wide variety of cytokines and chemokines, which can have both direct effects on neurons by changing the expression of ion channels and perturbing electrical properties, as well as indirect effects through glia-glia and immune-glia cross-talk. There is significant overlap in these cytokine and chemokine expression profiles across diseases, with TNFα and IL-6 strongly associated with cognitive deficits in multiple disorders. Here, we review the involvement of various cytokines and chemokines in AD, MS, FTLD, PD, TBI, MDD, and normal aging in the absence of dementia. We propose that the neuropsychiatric phenotypes observed in these disorders may be at least partially attributable to a dysregulation of immunity resulting in pathological cytokine and chemokine expression from both CNS-resident and non-resident cells.
Collapse
Affiliation(s)
- Benjamin C. Shaw
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Victoria R. Anders
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Maria L. Habean
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
| | - Orion D. Brock
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Molecular Medicine, Lerner Research Institute, Cleveland Clinic and Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin J. Frostino
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- College of Science, University of Notre Dame, South Bend, IN, USA
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
- Molecular Medicine, Lerner Research Institute, Cleveland Clinic and Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
18
|
Ollen-Bittle N, Roseborough AD, Wang W, Wu JLD, Whitehead SN. Connecting cellular mechanisms and extracellular vesicle cargo in traumatic brain injury. Neural Regen Res 2024; 19:2119-2131. [PMID: 38488547 PMCID: PMC11034607 DOI: 10.4103/1673-5374.391329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 04/24/2024] Open
Abstract
Traumatic brain injury is followed by a cascade of dynamic and complex events occurring at the cellular level. These events include: diffuse axonal injury, neuronal cell death, blood-brain barrier break down, glial activation and neuroinflammation, edema, ischemia, vascular injury, energy failure, and peripheral immune cell infiltration. The timing of these events post injury has been linked to injury severity and functional outcome. Extracellular vesicles are membrane bound secretory vesicles that contain markers and cargo pertaining to their cell of origin and can cross the blood-brain barrier. These qualities make extracellular vesicles intriguing candidates for a liquid biopsy into the pathophysiologic changes occurring at the cellular level post traumatic brain injury. Herein, we review the most commonly reported cargo changes in extracellular vesicles from clinical traumatic brain injury samples. We then use knowledge from animal and in vitro models to help infer what these changes may indicate regrading cellular responses post traumatic brain injury. Future research should prioritize labeling extracellular vesicles with markers for distinct cell types across a range of timepoints post traumatic brain injury.
Collapse
Affiliation(s)
- Nikita Ollen-Bittle
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Austyn D. Roseborough
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Wenxuan Wang
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jeng-liang D. Wu
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Shawn N. Whitehead
- Department of Anatomy and Cell Biology, Western University, London, ON, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Deparment of Clinical Neurological Sciences, Western University, London, ON, Canada
| |
Collapse
|
19
|
Al-Khateeb ZF, Henson SM, Tremoleda JL, Michael-Titus AT. The Immune Response in Two Models of Traumatic Injury of the Immature Brain. Cells 2024; 13:1612. [PMID: 39404376 PMCID: PMC11475908 DOI: 10.3390/cells13191612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Traumatic brain injury (TBI) can cause major disability and increases the risk of neurodegeneration. Post-TBI, there is infiltration of peripheral myeloid and lymphoid cells; there is limited information on the peripheral immune response post-TBI in the immature brain-where injury may interfere with neurodevelopment. We carried out two injury types in juvenile mice: invasive TBI with a controlled cortical impact (CCI) and repetitive mild TBI (rmTBI) using weight drop injury and analysed the response at 5- and 35-days post-injury. In the two models, we detected the brain infiltration of immune cells (e.g., neutrophils, monocytes, dendritic cells, CD4+ T cells, and NK cells). There were increases in macrophages, neutrophils, and dendritic cells in the spleen, increases in dendritic cells in blood, and increases in CD8+ T cells and B cells in lymph nodes. These results indicate a complex peripheral immune response post-TBI in the immature brain, with differences between an invasive injury and a repetitive mild injury.
Collapse
Affiliation(s)
- Zahra F. Al-Khateeb
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Siân M. Henson
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jordi L. Tremoleda
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Adina T. Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
20
|
Chen W, Wang Z, Ye G, Zhu G, Li S, Chen P, Wang H, Zou S, Chen M. Changes of NLRP3 in serum and cerebrospinal fluid of patients after moderate to severe traumatic brain injury and their predictive values for prognosis. CNS Neurosci Ther 2024; 30:e70009. [PMID: 39302033 PMCID: PMC11413909 DOI: 10.1111/cns.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) remains a major concern for global health. Recent studies have suggested the role of NOD-like receptor pyrin domain-containing protein 3 (NLRP3), an inflammatory marker, in the cerebrospinal fluid (CSF) and serum as potential indicators of TBI prognosis. The objective of the study was to characterize NLRP3 as a clinically applicable tool for predicting the outcomes of TBI patients. METHODS A total of 270 patients with moderate to severe TBI were included in this retrospective analysis. Serum and CSF samples were collected at 1-, 3-, 7-, and 21-day post-injury to measure NLRP3 levels. The prognosis of patients was evaluated after 3 months using the Glasgow Outcome Scale (GOS). Patients were categorized into good prognosis (GOS score >3) and poor prognosis (GOS score ≤3) groups. The relationship between NLRP3 levels and prognosis was analyzed. RESULTS Patients with poor prognosis had significantly elevated NLRP3 levels in their serum on days 1 and 3 post-injury compared with those with a good prognosis. The difference was more pronounced during these early days compared with days 7 and 21. However, NLRP3 levels in CSF consistently showed a large difference between the two groups throughout the observation period. Receiver operating characteristic analysis revealed that the level of NLRP3 in the CSF on day 3 post-injury had the highest predictive value for prognosis, with an area under the curve of 0.83, followed by the level of NLRP3 in the serum on day 3 post-injury. CONCLUSIONS The levels of NLRP3, especially in the CSF on day 3 post-injury, can serve as a potential biomarker for predicting prognosis in moderate to severe TBI patients. Early measurement of NLRP3 levels can provide valuable insights into patient outcomes and guide therapeutic strategies.
Collapse
Affiliation(s)
- Wei Chen
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
- Department of Neurosurgery, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Zhigang Wang
- Department of Neurosurgery, Shanghai East Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Gengfan Ye
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Guangyao Zhu
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Shiwei Li
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Pandi Chen
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Hongcai Wang
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| | - Shufeng Zou
- Department of NeurosurgeryThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiChina
| | - Maosong Chen
- Department of NeurosurgeryThe Affiliated Lihuili Hospital of Ningbo UniversityNingboZhejiangChina
| |
Collapse
|
21
|
El-Menyar A, Asim M, Khan N, Rizoli S, Mahmood I, Al-Ani M, Kanbar A, Alaieb A, Hakim S, Younis B, Taha I, Jogol H, Siddiqui T, Hammo AA, Abdurraheim N, Alabdallat M, Bahey AAA, Ahmed K, Atique S, Chaudry IH, Prabhu KS, Uddin S, Al-Thani H. Systemic and cerebro-cardiac biomarkers following traumatic brain injury: an interim analysis of randomized controlled clinical trial of early administration of beta blockers. Sci Rep 2024; 14:19574. [PMID: 39179700 PMCID: PMC11343837 DOI: 10.1038/s41598-024-70470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
This is an interim analysis of the Beta-blocker (Propranolol) use in traumatic brain injury (TBI) based on the high-sensitive troponin status (BBTBBT) study. The BBTBBT is an ongoing double-blind placebo-controlled randomized clinical trial with a target sample size of 771 patients with TBI. We sought, after attaining 50% of the sample size, to explore the impact of early administration of beta-blockers (BBs) on the adrenergic surge, pro-inflammatory cytokines, and the TBI biomarkers linked to the status of high-sensitivity troponin T (HsTnT). Patients were stratified based on the severity of TBI using the Glasgow coma scale (GCS) and HsTnT status (positive vs negative) before randomization. Patients with positive HsTnT (non-randomized) received propranolol (Group-1; n = 110), and those with negative test were randomized to receive propranolol (Group-2; n = 129) or placebo (Group-3; n = 111). Propranolol was administered within 24 h of injury for 6 days, guided by the heart rate (> 60 bpm), systolic blood pressure (≥ 100 mmHg), or mean arterial pressure (> 70 mmHg). Luminex and ELISA-based immunoassays were used to quantify the serum levels of pro-inflammatory cytokines (Interleukin (IL)-1β, IL-6, IL-8, and IL-18), TBI biomarkers [S100B, Neuron-Specific Enolase (NSE), and epinephrine]. Three hundred and fifty patients with comparable age (mean 34.8 ± 9.9 years) and gender were enrolled in the interim analysis. Group 1 had significantly higher baseline levels of IL-6, IL-1B, S100B, lactate, and base deficit than the randomized groups (p = 0.001). Group 1 showed a significant temporal reduction in serum IL-6, IL-1β, epinephrine, and NSE levels from baseline to 48 h post-injury (p = 0.001). Patients with severe head injuries had higher baseline levels of IL-6, IL-1B, S100B, and HsTnT than mild and moderate TBI (p = 0.01). HsTnT levels significantly correlated with the Injury Severity Score (ISS) (r = 0.275, p = 0.001), GCS (r = - 0.125, p = 0.02), and serum S100B (r = 0.205, p = 0.001). Early Propranolol administration showed a significant reduction in cytokine levels and TBI biomarkers from baseline to 48 h post-injury, particularly among patients with positive HsTnT, indicating the potential role in modulating inflammation post-TBI.Trial registration: ClinicalTrials.gov NCT04508244. It was registered first on 11/08/2020. Recruitment started on 29 December 2020 and is ongoing. The study was partly presented at the 23rd European Congress of Trauma and Emergency Surgery (ECTES), April 28-30, 2024, in Estoril, Lisbon, Portugal.
Collapse
Affiliation(s)
- Ayman El-Menyar
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar.
- Department of Clinical Medicine, Weill Cornell Medicine, P.O. Box 24144, Doha, Qatar.
| | - Mohammad Asim
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Naushad Khan
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Sandro Rizoli
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ismail Mahmood
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Mushreq Al-Ani
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ahad Kanbar
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Abubaker Alaieb
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Suhail Hakim
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Basil Younis
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ibrahim Taha
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Hisham Jogol
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Tariq Siddiqui
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Abdel Aziz Hammo
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Nuri Abdurraheim
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Mohammad Alabdallat
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | | | - Khalid Ahmed
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Sajid Atique
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Irshad H Chaudry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Hassan Al-Thani
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
22
|
Hiskens MI, Schneiders AG, Fenning AS. Selective COX-2 Inhibitors as Neuroprotective Agents in Traumatic Brain Injury. Biomedicines 2024; 12:1930. [PMID: 39200394 PMCID: PMC11352079 DOI: 10.3390/biomedicines12081930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/31/2024] [Accepted: 08/21/2024] [Indexed: 09/02/2024] Open
Abstract
Traumatic brain injury (TBI) is a significant contributor to mortality and morbidity in people, both young and old. There are currently no approved therapeutic interventions for TBI. Following TBI, cyclooxygenase (COX) enzymes generate prostaglandins and reactive oxygen species that perpetuate inflammation, with COX-1 and COX-2 isoforms providing differing responses. Selective COX-2 inhibitors have shown potential as neuroprotective agents. Results from animal models of TBI suggest potential treatment through the alleviation of secondary injury mechanisms involving neuroinflammation and neuronal cell death. Additionally, early clinical trials have shown that the use of celecoxib improves patient mortality and outcomes. This review aims to summarize the therapeutic effects of COX-2 inhibitors observed in TBI animal models, highlighting pertinent studies elucidating molecular pathways and expounding upon their mechanistic actions. We then investigated the current state of evidence for the utilization of COX-2 inhibitors for TBI patients.
Collapse
Affiliation(s)
- Matthew I. Hiskens
- Mackay Institute of Research and Innovation, Mackay Hospital and Health Service, Mackay, QLD 4740, Australia
| | - Anthony G. Schneiders
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia (A.S.F.)
| | - Andrew S. Fenning
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia (A.S.F.)
| |
Collapse
|
23
|
Lampros M, Vlachodimitropoulou L, Alexiou GA, Voulgaris S. Reader Response: Utility of Acute and Subacute Blood Biomarkers to Assist Diagnosis in CT-Negative Isolated Mild Traumatic Brain Injury. Neurology 2024; 103:e209336. [PMID: 38959469 DOI: 10.1212/wnl.0000000000209336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
|
24
|
Chand Dakal T, Choudhary K, Tiwari I, Yadav V, Kumar Maurya P, Kumar Sharma N. Unraveling the Triad: Hypoxia, Oxidative Stress and Inflammation in Neurodegenerative Disorders. Neuroscience 2024; 552:126-141. [PMID: 38936458 DOI: 10.1016/j.neuroscience.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
The mammalian brain's complete dependence on oxygen for ATP production makes it highly susceptible to hypoxia, at high altitudes or in clinical scenarios including anemia or pulmonary disease. Hypoxia plays a crucial role in the development of various brain disorders, such as Alzheimer's, Parkinson's, and other age-related neurodegenerative diseases. On the other hand, a decrease in environmental oxygen levels, such as prolonged stays at high elevations, may have beneficial impacts on the process of ageing and the likelihood of death. Additionally, the utilization of controlled hypoxia exposure could potentially serve as a therapeutic approach for age-related brain diseases. Recent findings indicate that the involvement of HIF-1α and the NLRP3 inflammasome is of significant importance in the development of Alzheimer's disease. HIF-1α serves as a pivotal controller of various cellular reactions to oxygen deprivation, exerting influence on a multitude of physiological mechanisms such as energy metabolism and inflammatory responses. The NLRP3 plays a crucial role in the innate immune system by coordinating the initiation of inflammatory reactions through the assembly of the inflammasome complex. This review examines the information pertaining to the contrasting effects of hypoxia on the brain, highlighting both its positive and deleterious effects and molecular pathways that are involved in mediating these different effects. This study explores potential strategies for therapeutic intervention that focus on restoring cellular balance and reducing neuroinflammation, which are critical aspects in addressing this severe neurodegenerative condition and addresses crucial inquiries that warrant further future investigations.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Kanika Choudhary
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Isha Tiwari
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk 304022, Rajasthan, India
| | - Vikas Yadav
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk 304022, Rajasthan, India.
| |
Collapse
|
25
|
Negrin LL, Ristl R, Wollner G, Hajdu S. Differences in Eotaxin Serum Levels between Polytraumatized Patients with and without Concomitant Traumatic Brain Injury-A Matched Pair Analysis. J Clin Med 2024; 13:4218. [PMID: 39064258 PMCID: PMC11277900 DOI: 10.3390/jcm13144218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Background/Objectives: Early detection of traumatic brain injury (TBI) is crucial for minimizing secondary neurological damage. Our study aimed to assess the potential of IL-4, IL-6, IL-7, IL-8, IL-10, TNF, and eotaxin serum levels-as a single clinical tool or combined into a panel-for diagnosing TBI in multiple injured patients. Methods: Out of 110 prospectively enrolled polytrauma victims (median age, 39 years; median ISS, 33; 70.9% male) admitted to our level I trauma center over four years, we matched 41 individuals with concomitant TBI (TBI cohort) to 41 individuals without TBI (non-TBI cohort) based on age, gender, Injury Severity Score (ISS), and mortality. Patients' protein levels were measured upon admission (day 0) and on days 1, 3, 5, 7, and 10 during routine blood withdrawal using one separation gel tube each time. Results: The median serum levels of IL-4, IL-6, IL-7, IL-8, IL-10, and TNF exhibited non-similar time courses in the two cohorts and showed no significant differences on days 0, 1, 3, 5, and 7. However, the median eotaxin levels had similar trend lines in both cohorts, with consistently higher levels in the TBI cohort, reaching significance on days 0, 3, and 5. In both cohorts, the median eotaxin level significantly decreased from day 0 to day 1, then significantly increased until day 10. We also found a significant positive association between day 0 eotaxin serum levels and the presence of TBI, indicating that for every 20 pg/mL increase in eotaxin level, the odds of a prevalent TBI rose by 10.5%. ROC analysis provided a cutoff value of 154 pg/mL for the diagnostic test (sensitivity, 0.707; specificity, 0.683; AUC = 0.718). Conclusions: Our findings identified the brain as a significant source, solely of eotaxin release in humans who have suffered a TBI. Nevertheless, the eotaxin serum level assessed upon admission has limited diagnostic value. IL-4, IL-6, IL-7, IL-8, IL-10, and TNF do not indicate TBI in polytraumatized patients.
Collapse
Affiliation(s)
- Lukas L. Negrin
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (S.H.)
| | - Robin Ristl
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, 1090 Vienna, Austria;
| | - Gregor Wollner
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (S.H.)
| | - Stefan Hajdu
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (S.H.)
| |
Collapse
|
26
|
Boucher ML, Conley G, Morriss NJ, Ospina-Mora S, Qiu J, Mannix R, Meehan WP. Time-Dependent Long-Term Effect of Memantine following Repetitive Mild Traumatic Brain Injury. J Neurotrauma 2024; 41:e1736-e1758. [PMID: 38666723 DOI: 10.1089/neu.2023.0423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Repetitive mild traumatic brain injury (rmTBI, e.g., sports concussions) may be associated with both acute and chronic symptoms and neurological changes. Despite the common occurrence of these injuries, therapeutic strategies are limited. One potentially promising approach is N-methyl-D-aspartate receptor (NMDAR) blockade to alleviate the effects of post-injury glutamatergic excitotoxicity. Initial pre-clinical work using the NMDAR antagonist, memantine, suggests that immediate treatment following rmTBI improves a variety of acute outcomes. It remains unclear (1) whether acute memantine treatment has long-term benefits and (2) whether delayed treatment following rmTBI is beneficial, which are both clinically relevant concerns. To test this, animals were subjected to rmTBI via a weight drop model with rotational acceleration (five hits in 5 days) and randomized to memantine treatment immediately, 3 months, or 6 months post-injury, with a treatment duration of one month. Behavioral outcomes were assessed at 1, 4, and 7 months post-injury. Neuropathological outcomes were characterized at 7 months post-injury. We observed chronic changes in behavior (anxiety-like behavior, motor coordination, spatial learning, and memory), as well as neuroinflammation (microglia, astrocytes) and tau phosphorylation (T231). Memantine treatment, either immediately or 6 months post-injury, appears to confer greater rescue of neuroinflammatory changes (microglia) than vehicle or treatment at the 3-month time point. Although memantine is already being prescribed chronically to address persistent symptoms associated with rmTBI, this study represents the first evidence of which we are aware to suggest a small but durable effect of memantine treatment in mild, concussive injuries. This effect suggests that memantine, although potentially beneficial, is insufficient to treat all aspects of rmTBI alone and should be combined with other therapeutic agents in a multi-therapy approach, with attention given to the timing of treatment.
Collapse
Affiliation(s)
- Masen L Boucher
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Nicholas J Morriss
- University of Rochester School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Jianhua Qiu
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebekah Mannix
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - William P Meehan
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- The Micheli Center for Sports Injury Prevention, Waltham, Massachusetts, USA
| |
Collapse
|
27
|
Baudo G, Flinn H, Holcomb M, Tiwari A, Soriano S, Taraballi F, Godin B, Zinger A, Villapol S. Sex-dependent improvement in traumatic brain injury outcomes after liposomal delivery of dexamethasone in mice. Bioeng Transl Med 2024; 9:e10647. [PMID: 39036088 PMCID: PMC11256133 DOI: 10.1002/btm2.10647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/02/2023] [Accepted: 01/11/2024] [Indexed: 07/23/2024] Open
Abstract
Traumatic brain injury (TBI) can have long-lasting physical, emotional, and cognitive consequences due to the neurodegeneration caused by its robust inflammatory response. Despite advances in rehabilitation care, effective neuroprotective treatments for TBI patients are lacking. Furthermore, current drug delivery methods for TBI treatment are inefficient in targeting inflamed brain areas. To address this issue, we have developed a liposomal nanocarrier (Lipo) encapsulating dexamethasone (Dex), an agonist for the glucocorticoid receptor utilized to alleviate inflammation and swelling in various conditions. In vitro studies show that Lipo-Dex were well tolerated in human and murine neural cells. Lipo-Dex showed significant suppression of inflammatory cytokines, IL-6 and TNF-α, release after induction of neural inflammation with lipopolysaccharide. Further, the Lipo-Dex were administered to young adult male and female C57BL/6 mice immediately after controlled cortical impact injury (a TBI model). Our findings demonstrate that Lipo-Dex can selectively target the injured brain, thereby reducing lesion volume, cell death, astrogliosis, the release of pro-inflammatory cytokines, and microglial activation compared to Lipo-treated mice in a sex-dependent manner, showing a major impact only in male mice. This highlights the importance of considering sex as a crucial variable in developing and evaluating new nano-therapies for brain injury. These results suggest that Lipo-Dex administration may effectively treat acute TBI.
Collapse
Affiliation(s)
- Gherardo Baudo
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Hannah Flinn
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Morgan Holcomb
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Anjana Tiwari
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Sirena Soriano
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Francesca Taraballi
- Department of Orthopedics and Sports Medicine and Center for Musculoskeletal RegenerationHouston Methodist HospitalHoustonTexasUSA
| | - Biana Godin
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
- Department of Obstetrics and GynecologyHouston Methodist Research InstituteHoustonTexasUSA
- Department of Obstetrics and GynecologyWeill Cornell Medicine College (WCMC)New YorkNew YorkUSA
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Assaf Zinger
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexasUSA
- Department of Chemical EngineeringTechnion−Israel Institute of TechnologyHaifaIsrael
| | - Sonia Villapol
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
- Department of Neuroscience in Neurological SurgeryWeill Cornell Medicine College (WCMC)New YorkNew YorkUSA
| |
Collapse
|
28
|
Houle S, Tapp Z, Dobres S, Ahsan S, Reyes Y, Cotter C, Mitsch J, Zimomra Z, Peng J, Rowe RK, Lifshitz J, Sheridan J, Godbout J, Kokiko-Cochran ON. Sleep fragmentation after traumatic brain injury impairs behavior and conveys long-lasting impacts on neuroinflammation. Brain Behav Immun Health 2024; 38:100797. [PMID: 38803369 PMCID: PMC11128763 DOI: 10.1016/j.bbih.2024.100797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024] Open
Abstract
Traumatic brain injury (TBI) causes a prolonged inflammatory response in the central nervous system (CNS) driven by microglia. Microglial reactivity is exacerbated by stress, which often provokes sleep disturbances. We have previously shown that sleep fragmentation (SF) stress after experimental TBI increases microglial reactivity and impairs hippocampal function 30 days post-injury (DPI). The neuroimmune response is highly dynamic the first few weeks after TBI, which is also when injury induced sleep-wake deficits are detected. Therefore, we hypothesized that even a few weeks of TBI SF stress would synergize with injury induced sleep-wake deficits to promote neuroinflammation and impair outcome. Here, we investigated the effects of environmental SF in a lateral fluid percussion model of mouse TBI. Half of the mice were undisturbed, and half were exposed to 5 h of SF around the onset of the light cycle, daily, for 14 days. All mice were then undisturbed 15-30 DPI, providing a period for SF stress recovery (SF-R). Mice exposed to SF stress slept more than those in control housing 7-14 DPI and engaged in more total daily sleep bouts during the dark period. However, SF stress did not exacerbate post-TBI sleep deficits. Testing in the Morris water maze revealed sex dependent differences in spatial reference memory 9-14 DPI with males performing worse than females. Post-TBI SF stress suppressed neurogenesis-related gene expression and increased inflammatory signaling in the cortex at 14 DPI. No differences in sleep behavior were detected between groups during the SF stress recovery period 15-30 DPI. Microscopy revealed cortical and hippocampal IBA1 and CD68 percent-area increased in TBI SF-R mice 30 DPI. Additionally, neuroinflammatory gene expression was increased, and synaptogenesis-related gene expression was suppressed in TBI-SF mice 30 DPI. Finally, IPA canonical pathway analysis showed post-TBI SF impaired and delayed activation of synapse-related pathways between 14 and 30 DPI. These data show that transient SF stress after TBI impairs recovery and conveys long-lasting impacts on neuroimmune function independent of continuous sleep deficits. Together, these finding support that even limited exposure to post-TBI SF stress can have lasting impacts on cognitive recovery and regulation of the immune response to trauma.
Collapse
Affiliation(s)
- Samuel Houle
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Zoe Tapp
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
| | - Shannon Dobres
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Sakeef Ahsan
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Yvanna Reyes
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Christopher Cotter
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Jessica Mitsch
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
| | - Zachary Zimomra
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
| | - Juan Peng
- Center for Biostatistics, The Ohio State University, 320-55 Lincoln Tower, 1800 Cannon Drive, 43210, Columbus, OH, USA
| | - Rachel K. Rowe
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Jonathan Lifshitz
- Phoenix VA Health Care System and University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - John Sheridan
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
- Division of Biosciences, College of Dentistry, The Ohio State University, 305 W. 12th Ave, 43210, Columbus, OH, USA
| | - Jonathan Godbout
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, 190 North Oval Mall, 43210, Columbus, OH, USA
| | - Olga N. Kokiko-Cochran
- Dept. of Neuroscience, College of Medicine, The Ohio State University, 1858 Neil Ave, 43210, Columbus, OH, USA
- Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, 460 Medical Center Drive, 43210, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, 190 North Oval Mall, 43210, Columbus, OH, USA
| |
Collapse
|
29
|
Chen Z, Ford KP, Islam MBAR, Wan H, Han H, Ramakrishnan A, Brown RJ, Villanueva V, Wang Y, Davis BT, Weiss C, Cui W, Gate D, Schwulst SJ. antiCD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8+ T-cell cytotoxicity after traumatic brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.596673. [PMID: 38948775 PMCID: PMC11212861 DOI: 10.1101/2024.06.17.596673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people. Graphic abstract Aged brains after TBI comprise two pools of CD8 + T cells . The aged brain has long been resided by a population of CD8 + T cells that's exhaustive and dysfunctional. Post TBI, due to BBB impairment, functional CD8 + T cells primarily migrate into the brain parenchyma. Aged, injury-associated microglia with upregulated MHC class I molecules can present neoantigens such as neuronal and/or myelin debris in the injured brains to functional CD8+ T, resulting in downstream CD8+ T cell cytotoxicity. aCD49d Ab treatment exerts its function by blocking the migration of functional effector CD8 + T cell population, leading to less cytotoxicity and resulting in improved TBI outcomes in aged mice.
Collapse
|
30
|
Huang CY, Rau CS, Huang CY, Su WT, Hsu SY, Hsieh CH. The Stress Index as a Predictor of Mortality in Patients with Isolated Moderate to Severe Traumatic Brain Injury. Diagnostics (Basel) 2024; 14:1244. [PMID: 38928658 PMCID: PMC11203316 DOI: 10.3390/diagnostics14121244] [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: 05/21/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The Stress Index (SI), calculated as the ratio of blood glucose to serum potassium levels, is a promising prognostic marker in various acute care settings. This study aimed to evaluate the utility of the SI for predicting mortality in patients with isolated moderate-to-severe traumatic brain injury (TBI). METHODS This retrospective cohort study included adult trauma patients (aged ≥ 20 years) with isolated moderate to severe TBI (Abbreviated Injury Scale ≥ 3 for only head region) treated from 2009-2022. The SI was computed from the initial glucose and potassium levels upon arrival at the emergency department. Logistic regression models were used to assess the association between the SI and mortality after adjusting for relevant covariates. The most effective threshold value of the SI for predicting mortality was identified using receiver operating characteristic (ROC) analysis. RESULTS Among the 4357 patients with isolated moderate and severe TBI, 463 (10.6%) died. Deceased patients had a significantly higher SI (61.7 vs. 44.1, p < 0.001). In multivariate analysis, higher SI independently predicted greater mortality risk (odds ratio (OR) 6.70, 95% confidence interval (CI) 1.66-26.99, p = 0.007). The optimal SI cutoff for predicting mortality was 48.50 (sensitivity 62.0%, specificity 71.4%, area under the curve 0.724). Patients with SI ≥ 48.5 had nearly two-fold higher adjusted mortality odds compared to those below the threshold (adjusted OR 1.94, 95% CI 1.51-2.50, p < 0.001). CONCLUSIONS SI is a useful predictor of mortality in patients with isolated moderate-to-severe TBI. Incorporating SI with standard clinical assessments could enhance risk stratification and management approaches for this patient population.
Collapse
Affiliation(s)
- Ching-Ya Huang
- Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Chun-Ying Huang
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-Y.H.); (W.-T.S.); (S.-Y.H.)
| | - Wei-Ti Su
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-Y.H.); (W.-T.S.); (S.-Y.H.)
| | - Shiun-Yuan Hsu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-Y.H.); (W.-T.S.); (S.-Y.H.)
| | - Ching-Hua Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-Y.H.); (W.-T.S.); (S.-Y.H.)
| |
Collapse
|
31
|
Rodriguez S, Sharma S, Tiarks G, Peterson Z, Jackson K, Thedens D, Wong A, Keffala-Gerhard D, Mahajan VB, Ferguson PJ, Newell EA, Glykys J, Nickl-Jockschat T, Bassuk AG. Neuroprotective effects of naltrexone in a mouse model of post-traumatic seizures. Sci Rep 2024; 14:13507. [PMID: 38867062 PMCID: PMC11169394 DOI: 10.1038/s41598-024-63942-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
Traumatic Brain Injury (TBI) induces neuroinflammatory response that can initiate epileptogenesis, which develops into epilepsy. Recently, we identified anti-convulsive effects of naltrexone, a mu-opioid receptor (MOR) antagonist, used to treat drug addiction. While blocking opioid receptors can reduce inflammation, it is unclear if post-TBI seizures can be prevented by blocking MORs. Here, we tested if naltrexone prevents neuroinflammation and/or seizures post-TBI. TBI was induced by a modified Marmarou Weight-Drop (WD) method on 4-week-old C57BL/6J male mice. Mice were placed in two groups: non-telemetry assessing the acute effects or in telemetry monitoring for interictal events and spontaneous seizures both following TBI and naltrexone. Molecular, histological and neuroimaging techniques were used to evaluate neuroinflammation, neurodegeneration and fiber track integrity at 8 days and 3 months post-TBI. Peripheral immune responses were assessed through serum chemokine/cytokine measurements. Our results show an increase in MOR expression, nitro-oxidative stress, mRNA expression of inflammatory cytokines, microgliosis, neurodegeneration, and white matter damage in the neocortex of TBI mice. Video-EEG revealed increased interictal events in TBI mice, with 71% mice developing post-traumatic seizures (PTS). Naltrexone treatment ameliorated neuroinflammation, neurodegeneration, reduced interictal events and prevented seizures in all TBI mice, which makes naltrexone a promising candidate against PTS, TBI-associated neuroinflammation and epileptogenesis in a WD model of TBI.
Collapse
Affiliation(s)
- Saul Rodriguez
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Shaunik Sharma
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Grant Tiarks
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Zeru Peterson
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kyle Jackson
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Daniel Thedens
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Angela Wong
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - David Keffala-Gerhard
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Vinit B Mahajan
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA
| | - Polly J Ferguson
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Elizabeth A Newell
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
| | - Joseph Glykys
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| | - Thomas Nickl-Jockschat
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Alexander G Bassuk
- Stead Family Department of Pediatrics , Carver College of Medicine, University of Iowa, 25 South Grand Ave, 2040 MedLabs, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
- Department of Neurology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
32
|
Cao Q, Gu L, Wang L, Sun G, Ying T, Su H, Wang W, Sun Z. Resveratrol alleviates endoplasmic reticulum stress-induced cell death and improves functional prognosis after traumatic brain injury in mice. J Appl Biomed 2024; 22:99-106. [PMID: 38912865 DOI: 10.32725/jab.2024.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 03/27/2024] [Indexed: 06/25/2024] Open
Abstract
Resveratrol (RSV) is a polyphenol antioxidant that has been shown to have neuroprotective effects. We sought molecular mechanisms that emphasize the anti-inflammatory activity of RSV in traumatic brain injury (TBI) in mice associated with endoplasmic reticulum stress (ERS). After establishing three experimental groups (sham, TBI, and TBI+RSV), we explored the results of RSV after TBI on ERS and caspase-12 apoptotic pathways. The expression levels of C/EBP homologous protein (CHOP), glucose regulated protein 78kD (GRP78), caspase-3, and caspase-12 in cortical brain tissues were assessed by western blotting. The qPCR analysis was also performed on mRNA expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1β in cortical brain tissue. In addition, the expression of GRP78 in microglia (ionized calcium binding adaptor molecule 1; Iba-1) and neurons (neuronal nuclei; NeuN) was identified by immunofluorescence staining. The neurological function of mice was assessed by modified neurological severity scores (mNSS). After drug treatment, the expression of CHOP, GRP78, caspase-3 and caspase-12 decreased, and qPCR results showed that TNF-α and IL-1β were down-regulated. Immunofluorescence staining showed down-regulation of Iba-1+/GRP78+ and NeuN+/GRP78+ cells after RSV treatment. The mNSS analysis confirmed improvement after RSV treatment. RSV improved apoptosis by downregulating the ERS signaling pathway and improved neurological prognosis in mice with TBI.
Collapse
Affiliation(s)
- Qinghua Cao
- The First Affiliated Hospital of Ningbo University, Department of Neurology, Ningbo 315000, China
| | - Lei Gu
- Ningbo Medical Center Lihuili Hospital, Department of Rehabilitation, Ningbo 315000, China
| | - Liangzhu Wang
- The First Affiliated Hospital of Ningbo University, Department of Neurology, Ningbo 315000, China
| | - Guangling Sun
- The First Affiliated Hospital of Ningbo University, Department of Neurology, Ningbo 315000, China
| | - Tao Ying
- The First Affiliated Hospital of Ningbo University, Department of Neurology, Ningbo 315000, China
| | - Hang Su
- The First Affiliated Hospital of Ningbo University, Department of Neurology, Ningbo 315000, China
| | - Wei Wang
- The First Affiliated Hospital of Ningbo University, Department of Neurology, Ningbo 315000, China
| | - Zhezhe Sun
- The First Affiliated Hospital of Ningbo University, Department of Neurology, Ningbo 315000, China
| |
Collapse
|
33
|
Özen I, Clausen F, Flygt J, Marklund N, Paul G. Neutralization of Interleukin 1-beta is associated with preservation of thalamic capillaries after experimental traumatic brain injury. Front Neurol 2024; 15:1378203. [PMID: 38765267 PMCID: PMC11100426 DOI: 10.3389/fneur.2024.1378203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/11/2024] [Indexed: 05/21/2024] Open
Abstract
Introduction Traumatic brain injury to thalamo-cortical pathways is associated with posttraumatic morbidity. Diffuse mechanical forces to white matter tracts and deep grey matter regions induce an inflammatory response and vascular damage resulting in progressive neurodegeneration. Pro-inflammatory cytokines, including interleukin-1β (IL-1β), may contribute to the link between inflammation and the injured capillary network after TBI. This study investigates whether IL-1β is a key contributor to capillary alterations and changes in pericyte coverage in the thalamus and cortex after TBI. Methods Animals were subjected to central fluid percussion injury (cFPI), a model of TBI causing widespread axonal and vascular pathology, or sham injury and randomized to receive a neutralizing anti-IL-1β or a control, anti-cyclosporin A antibody, at 30 min post-injury. Capillary length and pericyte coverage of cortex and thalamus were analyzed by immunohistochemistry at 2- and 7-days post-injury. Results and Conclusion Our results show that early post-injury attenuation of IL-1β dependent inflammatory signaling prevents capillary damage by increasing pericyte coverage in the thalamus.
Collapse
Affiliation(s)
- Ilknur Özen
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Fredrik Clausen
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Johanna Flygt
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Niklas Marklund
- Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden
| |
Collapse
|
34
|
Pordel S, McCloskey AP, Almahmeed W, Sahebkar A. The protective effects of statins in traumatic brain injury. Pharmacol Rep 2024; 76:235-250. [PMID: 38448729 DOI: 10.1007/s43440-024-00582-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Traumatic brain injury (TBI), often referred to as the "silent epidemic", is the most common cause of mortality and morbidity worldwide among all trauma-related injuries. It is associated with considerable personal, medical, and economic consequences. Although remarkable advances in therapeutic approaches have been made, current treatments and clinical management for TBI recovery still remain to be improved. One of the factors that may contribute to this gap is that existing therapies target only a single event or pathology. However, brain injury after TBI involves various pathological mechanisms, including inflammation, oxidative stress, blood-brain barrier (BBB) disruption, ionic disturbance, excitotoxicity, mitochondrial dysfunction, neuronal necrosis, and apoptosis. Statins have several beneficial pleiotropic effects (anti-excitotoxicity, anti-inflammatory, anti-oxidant, anti-thrombotic, immunomodulatory activity, endothelial and vasoactive properties) in addition to promoting angiogenesis, neurogenesis, and synaptogenesis in TBI. Supposedly, using agents such as statins that target numerous and diverse pathological mechanisms, may be more effective than a single-target approach in TBI management. The current review was undertaken to investigate and summarize the protective mechanisms of statins against TBI. The limitations of conducted studies and directions for future research on this potential therapeutic application of statins are also discussed.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
35
|
Kim DS, Kim GW. Biofluid-based Biomarkers in Traumatic Brain Injury: A Narrative Review. BRAIN & NEUROREHABILITATION 2024; 17:e8. [PMID: 38585027 PMCID: PMC10990840 DOI: 10.12786/bn.2024.17.e8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/13/2024] [Indexed: 04/09/2024] Open
Abstract
Traumatic brain injury (TBI) is a complex condition characterized by a multifaceted pathophysiology. It presents significant diagnostic and prognostic challenges in clinical settings. This narrative review explores the evolving role of biofluid biomarkers as essential tools in the diagnosis, prognosis, and treatment of TBI. In recent times, preclinical and clinical trials utilizing these biofluid biomarkers have been actively pursued internationally. Among the biomarkers for nerve tissue proteins are neuronal biomarkers like neuronal specific enolase and ubiquitin C-terminal hydrolase L1; astroglia injury biomarkers such as S100B and glial fibrillary acidic protein; axonal injury and demyelination biomarkers, including neurofilaments and myelin basic protein; new axonal injury and neurodegeneration biomarkers like total tau and phosphorylated tau; and others such as spectrin breakdown products and microtubule-associated protein 2. The interpretation of these biomarkers can be influenced by various factors, including secretion from organs other than the injury site and systemic conditions. This review highlights the potential of these biomarkers to transform TBI management and emphasizes the need for continued research to validate their efficacy, refine testing platforms, and ultimately improve patient care and outcomes.
Collapse
Affiliation(s)
- Da-Sol Kim
- Department of Physical Medicine and Rehabilitation, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine-Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Korea
| | - Gi-Wook Kim
- Department of Physical Medicine and Rehabilitation, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine-Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
36
|
Allen J, Dames SS, Foldi CJ, Shultz SR. Psychedelics for acquired brain injury: a review of molecular mechanisms and therapeutic potential. Mol Psychiatry 2024; 29:671-685. [PMID: 38177350 DOI: 10.1038/s41380-023-02360-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Acquired brain injury (ABI), such as traumatic brain injury and stroke, is a leading cause of disability worldwide, resulting in debilitating acute and chronic symptoms, as well as an increased risk of developing neurological and neurodegenerative disorders. These symptoms can stem from various neurophysiological insults, including neuroinflammation, oxidative stress, imbalances in neurotransmission, and impaired neuroplasticity. Despite advancements in medical technology and treatment interventions, managing ABI remains a significant challenge. Emerging evidence suggests that psychedelics may rapidly improve neurobehavioral outcomes in patients with various disorders that share physiological similarities with ABI. However, research specifically focussed on psychedelics for ABI is limited. This narrative literature review explores the neurochemical properties of psychedelics as a therapeutic intervention for ABI, with a focus on serotonin receptors, sigma-1 receptors, and neurotrophic signalling associated with neuroprotection, neuroplasticity, and neuroinflammation. The promotion of neuronal growth, cell survival, and anti-inflammatory properties exhibited by psychedelics strongly supports their potential benefit in managing ABI. Further research and translational efforts are required to elucidate their therapeutic mechanisms of action and to evaluate their effectiveness in treating the acute and chronic phases of ABI.
Collapse
Affiliation(s)
- Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Shannon S Dames
- Psychedelic-Assisted Therapy Post-Graduate Program, Health Sciences and Human Services, Vancouver Island University, Nanaimo, BC, Canada
| | - Claire J Foldi
- Department of Physiology, Monash University, Clayton, VIC, Australia
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Centre for Trauma and Mental Health Research, Health Sciences and Human Services, Vancouver Island University, Nanaimo, BC, Canada.
| |
Collapse
|
37
|
Morais A, Chung JY, Wu L, Ayata C, Simon B, Whalen MJ. Non-Invasive Vagal Nerve Stimulation Pre-Treatment Reduces Neurological Dysfunction After Closed Head Injury in Mice. Neurotrauma Rep 2024; 5:150-158. [PMID: 38435077 PMCID: PMC10908330 DOI: 10.1089/neur.2023.0058] [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] [Indexed: 03/05/2024] Open
Abstract
Non-invasive vagus nerve stimulation (nVNS) has recently been suggested as a potential therapy for traumatic brain injury (TBI). We previously demonstrated that nVNS inhibits cortical spreading depolarization, the electrophysiological event underlying migraine aura, and is relevant to TBI. Our past work also suggests a role for interleukin-1 beta (IL-1β) in cognitive deficits after closed head injury (CHI) in mice. We show that nVNS pre-treatment suppresses CHI-associated spatial learning and memory impairment and prevents IL-1β activation in injured neurons, but not endothelial cells. In contrast, nVNS administered 10 min after CHI was ineffective. These data suggest that nVNS prophylaxis might ameliorate neuronal dysfunction associated with CHI in populations at high risk for concussive TBI.
Collapse
Affiliation(s)
- Andreia Morais
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Joon Yong Chung
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Limin Wu
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Bruce Simon
- ElectroCore, Inc., Basking Ridge, New Jersey, USA
| | - Michael J. Whalen
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
38
|
Lyu IJ, Han K, Park KA, Oh SY. Ocular Motor Cranial Nerve Palsies and Increased Risk of Primary Malignant Brain Tumors: South Korean National Health Insurance Data. Cancers (Basel) 2024; 16:781. [PMID: 38398172 PMCID: PMC10886462 DOI: 10.3390/cancers16040781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
The aim of this study was to investigate the association between ocular motor cranial nerve palsies (OMCNP) and the occurrence of primary malignant brain tumors in a Korean population, using the national sample cohort database from Korea National Health Insurance Service (KNHIS). KNHIS data between 2010 and 2017 were analyzed. Our sample encompassed 118,686 participants, including 19,781 from a recently diagnosed OMCNP cohort and 98,905 from a matched control cohort through a 1:5 propensity score matching based on age and gender. To counteract the issue of reverse causation, we integrated a one-year time lag in our sensitivity analysis. Study participants were followed up until 31 December 2019. Cox proportional hazard regression analysis was used to compute the adjusted hazard ratio (HR) for primary malignant brain tumors according to the OMCNP diagnosis. Additionally, we performed a subgroup analysis to discern effects of various factors on the association between OMCNP and primary malignant brain tumors. HR for primary malignant brain tumors was 3.272 (95% confidence interval [CI]: 2.294 to 4.665) in the OMCNP cohort compared to the control cohort in a fully adjusted model for age, sex, socio-economic status, smoking, drinking, regular physical exercise, hypertension, diabetes, dyslipidemia, obesity, chronic kidney disease, and human immunodeficiency virus infection. Further subgroup analysis revealed that the risk of primary malignant brain tumors was significantly increased in women with OMCNP compared to men with OMCNP (HR: 5.118 in women vs. 2.441 in men, p = 0.0440), and in those aged <65 years than in those aged ≥65 years (HR: 6.951 in age < 65 years vs. 1.899 in age ≥ 65 years, p = 0.0006). Our population-based cohort study demonstrated a significantly increased risk of subsequent primary malignant brain tumors in patients with OMCNP. Particularly, OMCNP-afflicted women aged below 65 manifested a heightened probability of developing primary malignant brain tumors compared to those devoid of OMCNP.
Collapse
Affiliation(s)
- In Jeong Lyu
- Department of Ophthalmology, Korea Cancer Center Hospital, Seoul 01812, Republic of Korea;
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul 06978, Republic of Korea;
| | - Kyung-Ah Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Sei Yeul Oh
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
39
|
Kazis D, Chatzikonstantinou S, Ciobica A, Kamal FZ, Burlui V, Calin G, Mavroudis I. Epidemiology, Risk Factors, and Biomarkers of Post-Traumatic Epilepsy: A Comprehensive Overview. Biomedicines 2024; 12:410. [PMID: 38398011 PMCID: PMC10886732 DOI: 10.3390/biomedicines12020410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
This paper presents an in-depth exploration of Post-Traumatic Epilepsy (PTE), a complex neurological disorder following traumatic brain injury (TBI), characterized by recurrent, unprovoked seizures. With TBI being a global health concern, understanding PTE is crucial for effective diagnosis, management, and prognosis. This study aims to provide a comprehensive overview of the epidemiology, risk factors, and emerging biomarkers of PTE, thereby informing clinical practice and guiding future research. The epidemiological aspect of the study reveals PTE as a significant contributor to acquired epilepsies, with varying incidence influenced by injury severity, age, and intracranial pathologies. The paper delves into the multifactorial nature of PTE risk factors, encompassing clinical, demographic, and genetic elements. Key insights include the association of injury severity, intracranial hemorrhages, and early seizures with increased PTE risk, and the roles of age, gender, and genetic predispositions. Advancements in neuroimaging, electroencephalography, and molecular biology are presented, highlighting their roles in identifying potential PTE biomarkers. These biomarkers, ranging from radiological signs to electroencephalography EEG patterns and molecular indicators, hold promise for enhancing PTE pathogenesis understanding, early diagnosis, and therapeutic guidance. The paper also discusses the critical roles of astrocytes and microglia in PTE, emphasizing the significance of neuroinflammation in PTE development. The insights from this review suggest potential therapeutic targets in neuroinflammation pathways. In conclusion, this paper synthesizes current knowledge in the field, emphasizing the need for continued research and a multidisciplinary approach to effectively manage PTE. Future research directions include longitudinal studies for a better understanding of TBI and PTE outcomes, and the development of targeted interventions based on individualized risk profiles. This research contributes significantly to the broader understanding of epilepsy and TBI.
Collapse
Affiliation(s)
- Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.)
| | - Symela Chatzikonstantinou
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 20th Carol I Avenue, 700506 Iasi, Romania;
- Center of Biomedical Research, Romanian Academy, Iasi Branch, Teodor Codrescu 2, 700481 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov, 050044 Bucharest, Romania
| | - Fatima Zahra Kamal
- Higher Institute of Nursing Professions and Health Technical (ISPITS), Marrakech 40000, Morocco
- Laboratory of Physical Chemistry of Processes and Materials, Faculty of Sciences and Techniques, Hassan First University, Settat 26000, Morocco
| | - Vasile Burlui
- Department of Biomaterials, Faculty of Dental Medicine, Apollonia University, 700511 Iasi, Romania;
| | - Gabriela Calin
- Department of Biomaterials, Faculty of Dental Medicine, Apollonia University, 700511 Iasi, Romania;
| | - Ioannis Mavroudis
- Department of Neuroscience, Leeds Teaching Hospitals, Leeds LS2 9JT, UK
- Faculty of Medicine, Leeds University, Leeds LS2 9JT, UK
| |
Collapse
|
40
|
van Amerongen S, Pulukuri SV, Tuz-Zahra F, Tripodis Y, Cherry JD, Bernick C, Geda YE, Wethe JV, Katz DI, Alosco ML, Adler CH, Balcer LJ, Ashton NJ, Blennow K, Zetterberg H, Daneshvar DH, Colasurdo EA, Iliff JJ, Li G, Peskind ER, Shenton ME, Reiman EM, Cummings JL, Stern RA. Inflammatory biomarkers for neurobehavioral dysregulation in former American football players: findings from the DIAGNOSE CTE Research Project. J Neuroinflammation 2024; 21:46. [PMID: 38336728 PMCID: PMC10854026 DOI: 10.1186/s12974-024-03034-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Traumatic encephalopathy syndrome (TES) is defined as the clinical manifestation of the neuropathological entity chronic traumatic encephalopathy (CTE). A core feature of TES is neurobehavioral dysregulation (NBD), a neuropsychiatric syndrome in repetitive head impact (RHI)-exposed individuals, characterized by a poor regulation of emotions/behavior. To discover biological correlates for NBD, we investigated the association between biomarkers of inflammation (interleukin (IL)-1β, IL-6, IL-8, IL-10, C-reactive protein (CRP), tumor necrosis factor (TNF)-α) in cerebrospinal fluid (CSF) and NBD symptoms in former American football players and unexposed individuals. METHODS Our cohort consisted of former American football players, with (n = 104) or without (n = 76) NBD diagnosis, as well as asymptomatic unexposed individuals (n = 55) from the DIAGNOSE CTE Research Project. Specific measures for NBD were derived (i.e., explosivity, emotional dyscontrol, impulsivity, affective lability, and a total NBD score) from a factor analysis of multiple self-report neuropsychiatric measures. Analyses of covariance tested differences in biomarker concentrations between the three groups. Within former football players, multivariable linear regression models assessed relationships among log-transformed inflammatory biomarkers, proxies for RHI exposure (total years of football, cumulative head impact index), and NBD factor scores, adjusted for relevant confounding variables. Sensitivity analyses tested (1) differences in age subgroups (< 60, ≥ 60 years); (2) whether associations could be identified with plasma inflammatory biomarkers; (3) associations between neurodegeneration and NBD, using plasma neurofilament light (NfL) chain protein; and (4) associations between biomarkers and cognitive performance to explore broader clinical symptoms related to TES. RESULTS CSF IL-6 was higher in former American football players with NBD diagnosis compared to players without NBD. Furthermore, elevated levels of CSF IL-6 were significantly associated with higher emotional dyscontrol, affective lability, impulsivity, and total NBD scores. In older football players, plasma NfL was associated with higher emotional dyscontrol and impulsivity, but also with worse executive function and processing speed. Proxies for RHI exposure were not significantly associated with biomarker concentrations. CONCLUSION Specific NBD symptoms in former American football players may result from multiple factors, including neuroinflammation and neurodegeneration. Future studies need to unravel the exact link between NBD and RHI exposure, including the role of other pathophysiological pathways.
Collapse
Affiliation(s)
- Suzan van Amerongen
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Surya V Pulukuri
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Fatima Tuz-Zahra
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jonathan D Cherry
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Charles Bernick
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Yonas E Geda
- Department of Neurology and the Franke Global Neuroscience Education Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Jennifer V Wethe
- Department of Psychiatry and Psychology, Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Douglas I Katz
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Brain Injury Program, Encompass Health Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Michael L Alosco
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Laura J Balcer
- Departments of Neurology, Population Health and Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Daniel H Daneshvar
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Colasurdo
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
| | - Jeffrey J Iliff
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Gail Li
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System Geriatric Research, Seattle, WA, USA
| | - Elaine R Peskind
- Veterans Affairs Northwest Mental Illness Research, Education, and Clinical Center, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Martha E Shenton
- Psychiatry Neuroimaging Laboratory, Harvard Medical School, Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, University of Arizona, Arizona State University, Translational Genomics Research Institute, and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Robert A Stern
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Departments of Neurosurgery, and Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
41
|
Taghizadehghalehjoughi A, Naldan ME, Yeni Y, Genc S, Hacimuftuoglu A, Isik M, Necip A, Bolat İ, Yildirim S, Beydemir S, Baykan M. Effect of fentanyl and remifentanil on neuron damage and oxidative stress during induction neurotoxicity. J Cell Mol Med 2024; 28:e18118. [PMID: 38332529 PMCID: PMC10853584 DOI: 10.1111/jcmm.18118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/06/2023] [Accepted: 01/02/2024] [Indexed: 02/10/2024] Open
Abstract
Opioids can be used for medical and non-medical purposes. Chronic pain such as cancer, as well as the frequent use of such drugs in places such as operating rooms and intensive care units, and in non-medical areas like drug abuse the effects and side effects of these drugs need to be examined in more detail. For this purpose, the effects of fentanyl and remifentanil drugs on neuroinflammation, oxidative stress and cholinesterase metabolism were investigated. Neuron cells (CRL-10742) were used for the evaluation of the toxicity of fentanyl and remifentanil. MTT, PON1 activity and total thiol levels for its effect on oxidative stress, AChE and BChE activities for its effect on the cholinergic system, and TNF, IL-8 and IL-10 gene levels for its neuroinflammation effect were determined. The highest neurotoxic dose of fentanyl and remifentanil was determined as 10 μg/mL. It was observed that the rate of neuron cells in this dose has decreased by up to 61.80% and 56.89%, respectively. The IL-8 gene expression level in both opioids was down-regulated while IL 10 gene level was up-regulated in a dose-dependent manner compared to the control. In our results, the TNF gene expression level differs between the two opioids. In the fentanyl group, it was seen to be up-regulated in a dose-dependent manner compared to the control. Fentanyl and remifentanil showed an inhibitory effect against PON1, while remifentanil showed an increase in total thiol levels. PON1, BChE and total thiol activities showed similarity with MTT.
Collapse
Affiliation(s)
| | - Muhammet Emin Naldan
- Department of Anesthesiology and ReanimationUniversity of Health Sciences, Hospital of CityErzurumTurkey
| | - Yesim Yeni
- Department of Medical Pharmacology, Faculty of MedicineAtaturk UniversityErzurumTurkey
| | - Sidika Genc
- Department of Medical Pharmacology, Faculty of MedicineBilecik Seyh Edebali UniversityBilecikTurkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of MedicineAtaturk UniversityErzurumTurkey
| | - Mesut Isik
- Department of Bioengineering, Faculty of EngineeringBilecik Seyh Edebali UniversityBilecikTurkey
| | - Adem Necip
- Department of Pharmacy Services, Vocational School of Health ServicesHarran UniversitySanlıurfaTurkey
| | - İsmail Bolat
- Department of Pathology, Faculty of Veterinary MedicineAtaturk UniversityErzurumTurkey
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary MedicineAtaturk UniversityErzurumTurkey
| | - Sukru Beydemir
- Department of Biochemistry, Faculty of PharmacyAnadolu UniversityEskisehirTurkey
- The Rectorate of Bilecik Seyh Edebali UniversityBilecikTurkey
| | - Mahmut Baykan
- Department of Microbiology, Faculty of MedicineBilecik Seyh Edebali UniversityBilecikTurkey
| |
Collapse
|
42
|
Meier TB, Huber DL, Goeckner BD, Gill JM, Pasquina P, Broglio SP, McAllister TW, Harezlak J, McCrea MA. Association of Blood Biomarkers of Inflammation With Acute Concussion in Collegiate Athletes and Military Service Academy Cadets. Neurology 2024; 102:e207991. [PMID: 38165315 PMCID: PMC11407501 DOI: 10.1212/wnl.0000000000207991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/20/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The objective was to characterize the acute effects of concussion (a subset of mild traumatic brain injury) on serum interleukin (IL)-6 and IL-1 receptor antagonist (RA) and 5 additional inflammatory markers in athletes and military service academy members from the Concussion Assessment, Research, and Education Consortium and to determine whether these markers aid in discrimination of concussed participants from controls. METHODS Athletes and cadets with concussion and matched controls provided blood at baseline and postinjury visits between January 2015 and March 2020. Linear models investigated changes in inflammatory markers measured using Meso Scale Discovery assays across time points (baseline and 0-12, 12-36, 36-60 hours). Subanalyses were conducted in participants split by sex and injury population. Logistic regression analyses tested whether acute levels of IL-6 and IL-1RA improved discrimination of concussed participants relative to brain injury markers (glial fibrillary acidic protein, tau, neurofilament light, ubiquitin c-terminal hydrolase-L1) or clinical data (Sport Concussion Assessment Tool-Third Edition, Standardized Assessment of Concussion, Balance Error Scoring System). RESULTS Participants with concussion (total, N = 422) had elevated IL-6 and IL-1RA at 0-12 hours vs controls (n = 345; IL-6: mean difference [MD] (standard error) = 0.701 (0.091), p < 0.0001; IL-1RA: MD = 0.283 (0.042), p < 0.0001) and relative to baseline (IL-6: MD = 0.656 (0.078), p < 0.0001; IL-1RA: MD = 0.242 (0.038), p < 0.0001), 12-36 hours (IL-6: MD = 0.609 (0.086), p < 0.0001; IL-1RA: MD = 0.322 (0.041), p < 0.0001), and 36-60 hours (IL-6: MD = 0.818 (0.084), p < 0.0001; IL-1RA: MD = 0.317 (0.040), p < 0.0001). IL-6 and IL-1RA were elevated in participants with sport (IL-6: MD = 0.748 (0.115), p < 0.0001; IL-1RA: MD = 0.304 (0.055), p < 0.0001) and combative-related concussions (IL-6: MD = 0.583 (0.178), p = 0.001; IL-1RA: MD = 0.312 (0.081), p = 0.0001). IL-6 was elevated in male (MD = 0.734 (0.105), p < 0.0001) and female participants (MD = 0.600 (0.177), p = 0.0008); IL-1RA was only elevated in male participants (MD = 0.356 (0.047), p < 0.0001). Logistic regression showed the inclusion of IL-6 and IL-1RA at 0-12 hours improved the discrimination of participants with concussion from controls relative to brain injury markers (χ2(2) = 17.855, p = 0.0001; area under the receiver operating characteristic curve [AUC] 0.73 [0.66-0.80] to 0.78 [0.71-0.84]), objective clinical measures (balance and cognition; χ2(2) = 40.661, p < 0.0001; AUC 0.81 [0.76-0.86] to 0.87 [0.83-0.91]), and objective and subjective measures combined (χ2(2) = 13.456, p = 0.001; AUC 0.97 [0.95-0.99] to 0.98 [0.96-0.99]), although improvement in AUC was only significantly relative to objective clinical measures. DISCUSSION IL-6 and IL-1RA (male participants only) are elevated in the early-acute window postconcussion and may aid in diagnostic decisions beyond traditional blood markers and common clinical measures. IL-1RA results highlight sex differences in the immune response to concussion which should be considered in future biomarker work.
Collapse
Affiliation(s)
- Timothy B Meier
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Daniel L Huber
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Bryna D Goeckner
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Jessica M Gill
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Paul Pasquina
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Steven P Broglio
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Thomas W McAllister
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Jaroslaw Harezlak
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| | - Michael A McCrea
- From the Departments of Neurosurgery (T.B.M., D.L.H., M.A.M.), Biomedical Engineering (T.B.M.), Cell Biology, Neurobiology and Anatomy (T.B.M.), Biophysics (B.D.G.), and Neurology (M.A.M.), Medical College of Wisconsin, Milwaukee; National Institute of Nursing Research (J.M.G.), NIH, Bethesda; Johns Hopkins School of Nursing and Medicine (J.M.G.), Baltimore, MD; Department of Physical Medicine and Rehabilitation (P.P.), Uniformed Services University of the Health Sciences, Bethesda, MD; Michigan Concussion Center (S.P.B.), University of Michigan, Ann Arbor; Department of Psychiatry (T.W.M.), Indiana University School of Medicine, Indianapolis; Department of Epidemiology and Biostatistics (J.H.), School of Public Health-Bloomington, Indiana University
| |
Collapse
|
43
|
Ollewagen T, Benecke R, Smith C. High species homology potentiates quantitative inflammation profiling in zebrafish using immunofluorescence. Heliyon 2024; 10:e23635. [PMID: 38187273 PMCID: PMC10770569 DOI: 10.1016/j.heliyon.2023.e23635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Due to substantial homology between the human and zebrafish genome and a high level of conservation of the innate immune system across species, zebrafish larvae have become an invaluable research tool for studying inflammation and modelling inflammatory disease. However, further microscopy techniques need to be developed for better profiling of inflammation and in particular, integrated cytokine responses to different stimuli - approaches are currently largely limited to assessment of changes in cytokine gene transcription and in vivo visualisation using transgenics, which is limited in terms of the number of cytokines that may be assessed at once. In this study, after confirming substantial homology of human vs zebrafish cytokine amino acid sequences, immunofluorescence staining using antibodies directed at human cytokines was performed. Inflammatory cytokine signalling responses to experimental tailfin transection was assessed over 24 h (1 hpi (hours post injury), 2 hpi, 4 hpi, 24 hpi) in zebrafish larvae, with experimental end point at 120 h post fertilization (hpf). When immunofluorescence results were compared to responses observed in rodent and human literature, it is clear that the cytokines follow a similar response, albeit with a condensed total time course. Notably, tumor necrosis factor-α and monocyte chemoattractant protein-1 increased and remained elevated over the 24-h period. In contrast, interleukin-1β and interleukin-6 peaked at 4 hpi and 2 hpi respectively but had both returned to baseline levels by 24 hpi. Macrophage migration inhibitory factor was lowest at 1 hpi, potentially encouraging macrophage movement into the site of injury, followed by a sharp increase. This protocol provides valuable insight into inflammation over a time course and more so, provides an affordable and accessible method to comprehensively assess inflammation in zebrafish disease models.
Collapse
Affiliation(s)
| | - R.M. Benecke
- Experimental Medicine Research Group, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - C. Smith
- Experimental Medicine Research Group, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| |
Collapse
|
44
|
To XV, Mohamed AZ, Cumming P, Nasrallah FA. Diffusion tensor imaging and plasma immunological biomarker panel in a rat traumatic brain injury (TBI) model and in human clinical TBI. Front Immunol 2024; 14:1293471. [PMID: 38259455 PMCID: PMC10800599 DOI: 10.3389/fimmu.2023.1293471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Neuroinflammatory reactions play a significant role in the pathology and long-term consequences of traumatic brain injury (TBI) and may mediate salutogenic processes that white matter integrity. This study aimed to investigate the relationship between inflammatory markers and white matter integrity following TBI in both a rat TBI model and clinical TBI cases. Methods In the rat model, blood samples were collected following a controlled cortical impact (CCI) to assess a panel of inflammatory markers; MR-based diffusion tensor imaging (DTI) was employed to evaluate white matter integrity 60 days post-injury. 15 clinical TBI patients were similarly assessed for a panel of inflammatory markers and DTI post-intensive care unit discharge. Blood samples from healthy controls were used for comparison of the inflammatory markers. Results Time-dependent elevations in immunological markers were observed in TBI rats, with a correlation to preserved fractional anisotropy (FA) in white matter. Specifically, TBI-induced increased plasma levels of IL-1β, IL-6, G-CSF, CCL3, CCL5, and TNF-α were associated with higher white matter integrity, as measured by FA. Clinical cases had similar findings: elevated inflammatory markers (relative to controls) were associated with preservation of FA in vulnerable white matter regions. Discussion Inflammatory markers in post-TBI plasma samples are ambivalent with respect to prediction of favourable outcome versus a progression to more pervasive pathology and morbidity.
Collapse
Affiliation(s)
- Xuan Vinh To
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - Abdalla Z. Mohamed
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Fatima A. Nasrallah
- The Queensland Brain Institute, The University of Queensland, Queensland, Australia
- The Centre for Advanced Imaging, The University of Queensland, Queensland, Australia
| |
Collapse
|
45
|
Leonard J, Ladner L, Harris EA, de Jager C, Theus MH. The Neuroimmune Interface: Age-Related Responses to Traumatic Brain Injury. ADVANCES IN NEUROBIOLOGY 2024; 42:241-262. [PMID: 39432046 DOI: 10.1007/978-3-031-69832-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Traumatic Brain Injury (TBI) is a significant public health issue, with diverse consequences across the lifespan. This comprehensive review explores the complex interplay between age-related responses and the immune system following TBI. TBI exhibits distinct effects in pediatric, adult, and elderly populations, with profound implications for recovery and long-term outcomes. The immune system, as a key player in the post-TBI inflammatory cascade, exerts age-dependent influences on inflammation, neuroinflammation, and tissue repair. We examine the evolving understanding of age-related neuroinflammatory responses, cytokine profiles, and the role of immune cells, such as microglia and T cells, in the context of TBI. Furthermore, we evaluate the therapeutic implications of age-specific immunomodulation strategies toward mitigating TBI-associated neuropathology. This review consolidates the current knowledge on age-related immune responses in TBI, shedding light on potential avenues for tailored therapeutic interventions across the age spectrum. Understanding these nuanced responses is crucial for optimizing patient care and enhancing recovery outcomes in the aftermath of traumatic brain injury.
Collapse
Affiliation(s)
- John Leonard
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Liliana Ladner
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Elizabeth A Harris
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Caroline de Jager
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Michelle H Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
46
|
Huang YN, Greig NH, Huang PS, Chiang YH, Hoffer A, Yang CH, Tweedie D, Chen Y, Ou JC, Wang JY. Pomalidomide Improves Motor Behavioral Deficits and Protects Cerebral Cortex and Striatum Against Neurodegeneration Through a Reduction of Oxidative/Nitrosative Damages and Neuroinflammation After Traumatic Brain Injury. Cell Transplant 2024; 33:9636897241237049. [PMID: 38483119 PMCID: PMC10943757 DOI: 10.1177/09636897241237049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 03/18/2024] Open
Abstract
Neuronal damage resulting from traumatic brain injury (TBI) causes disruption of neuronal projections and neurotransmission that contribute to behavioral deficits. Cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) is an early event following TBI. ROS often damage DNA, lipids, proteins, and carbohydrates while RNS attack proteins. The products of lipid peroxidation 4-hydroxynonenal (4-HNE) and protein nitration 3-nitrotyrosine (3-NT) are often used as indicators of oxidative and nitrosative damages, respectively. Increasing evidence has shown that striatum is vulnerable to damage from TBI with a disturbed dopamine neurotransmission. TBI results in neurodegeneration, oxidative stress, neuroinflammation, neuronal apoptosis, and autophagy in the striatum and contribute to motor or behavioral deficits. Pomalidomide (Pom) is a Food and Drug Administration (FDA)-approved immunomodulatory drug clinically used in treating multiple myeloma. We previously showed that Pom reduces neuroinflammation and neuronal death induced by TBI in rat cerebral cortex. Here, we further compared the effects of Pom in cortex and striatum focusing on neurodegeneration, oxidative and nitrosative damages, as well as neuroinflammation following TBI. Sprague-Dawley rats subjected to a controlled cortical impact were used as the animal model of TBI. Systemic administration of Pom (0.5 mg/kg, intravenous [i.v.]) at 5 h post-injury alleviated motor behavioral deficits, contusion volume at 24 h after TBI. Pom alleviated TBI-induced neurodegeneration stained by Fluoro-Jade C in both cortex and striatum. Notably, Pom treatment reduces oxidative and nitrosative damages in cortex and striatum and is more efficacious in striatum (93% reduction in 4-HNE-positive and 84% reduction in 3-NT-positive neurons) than in cerebral cortex (42% reduction in 4-HNE-positive and 55% reduction in 3-NT-positive neurons). In addition, Pom attenuated microgliosis, astrogliosis, and elevations of proinflammatory cytokines in cortical and striatal tissue. We conclude that Pom may contribute to improved motor behavioral outcomes after TBI through targeting oxidative/nitrosative damages and neuroinflammation.
Collapse
Affiliation(s)
- Ya-Ni Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan City
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pen-Sen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei
- Neuroscience Research Center, Taipei Medical University, Taipei
| | - Alan Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Chih-Hao Yang
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ying Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei
| | - Ju-Chi Ou
- Neuroscience Research Center, Taipei Medical University, Taipei
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei
- Neuroscience Research Center, Taipei Medical University, Taipei
| |
Collapse
|
47
|
Kapate N, Liao R, Sodemann RL, Stinson T, Prakash S, Kumbhojkar N, Suja VC, Wang LLW, Flanz M, Rajeev R, Villafuerte D, Shaha S, Janes M, Park KS, Dunne M, Golemb B, Hone A, Adebowale K, Clegg J, Slate A, McGuone D, Costine-Bartell B, Mitragotri S. Backpack-mediated anti-inflammatory macrophage cell therapy for the treatment of traumatic brain injury. PNAS NEXUS 2024; 3:pgad434. [PMID: 38187808 PMCID: PMC10768983 DOI: 10.1093/pnasnexus/pgad434] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024]
Abstract
Traumatic brain injury (TBI) is a debilitating disease with no current therapies outside of acute clinical management. While acute, controlled inflammation is important for debris clearance and regeneration after injury, chronic, rampant inflammation plays a significant adverse role in the pathophysiology of secondary brain injury. Immune cell therapies hold unique therapeutic potential for inflammation modulation, due to their active sensing and migration abilities. Macrophages are particularly suited for this task, given the role of macrophages and microglia in the dysregulated inflammatory response after TBI. However, maintaining adoptively transferred macrophages in an anti-inflammatory, wound-healing phenotype against the proinflammatory TBI milieu is essential. To achieve this, we developed discoidal microparticles, termed backpacks, encapsulating anti-inflammatory interleukin-4, and dexamethasone for ex vivo macrophage attachment. Backpacks durably adhered to the surface of macrophages without internalization and maintained an anti-inflammatory phenotype of the carrier macrophage through 7 days in vitro. Backpack-macrophage therapy was scaled up and safely infused into piglets in a cortical impact TBI model. Backpack-macrophages migrated to the brain lesion site and reduced proinflammatory activation of microglia in the lesion penumbra of the rostral gyrus of the cortex and decreased serum concentrations of proinflammatory biomarkers. These immunomodulatory effects elicited a 56% decrease in lesion volume. The results reported here demonstrate, to the best of our knowledge, a potential use of a cell therapy intervention for a large animal model of TBI and highlight the potential of macrophage-based therapy. Further investigation is required to elucidate the neuroprotection mechanisms associated with anti-inflammatory macrophage therapy.
Collapse
Affiliation(s)
- Neha Kapate
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rick Liao
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Ryan Luke Sodemann
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tawny Stinson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Supriya Prakash
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Ninad Kumbhojkar
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Vineeth Chandran Suja
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Lily Li-Wen Wang
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mikayla Flanz
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Rohan Rajeev
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Dania Villafuerte
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Suyog Shaha
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Morgan Janes
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kyung Soo Park
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Michael Dunne
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Bryan Golemb
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alexander Hone
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kolade Adebowale
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - John Clegg
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Andrea Slate
- Center of Comparative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Declan McGuone
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Beth Costine-Bartell
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| |
Collapse
|
48
|
Sakul AA, Balcikanli Z, Ozsoy NA, Orhan C, Sahin N, Tuzcu M, Juturu V, Kilic E, Sahin K. A highly bioavailable curcumin formulation ameliorates inflammation cytokines and neurotrophic factors in mice with traumatic brain injury. Chem Biol Drug Des 2024; 103:e14439. [PMID: 38230778 DOI: 10.1111/cbdd.14439] [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: 04/27/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
A novel curcumin formulation increases relative absorption by 46 times (CurcuWIN®) of the total curcuminoids over the unformulated standard curcumin form. However, the exact mechanisms by which curcumin demonstrates its neuroprotective effects are not fully understood. This study aimed to investigate the impact of a novel formulation of curcumin on the expression of brain-derived neurotrophic factor (BDNF), glial fibrillary acidic protein (GFAP), a main component of the glial scar and growth-associated protein-43 (GAP-43), a signaling molecule in traumatic brain injury (TBI). Mice (adult, male, C57BL/6j) were randomly divided into three groups as follows: TBI group (TBI-induced mice); TBI + CUR group (TBI mice were injected i.p. curcumin just after TBI); TBI+ CurcuWIN® group (TBI mice were injected i.p. CurcuWIN® just after TBI). Brain injury was induced using a cold injury model. Injured brain tissue was stained with Cresyl violet to evaluate infarct volume and brain swelling, analyzed, and measured using ImageJ by Bethesda (MD, USA). Western blot analysis was performed to determine the protein levels related to injury. While standard curcumin significantly reduced brain injury, CurcuWIN® showed an even greater reduction associated with reductions in glial activation, NF-κB, and the inflammatory cytokines IL-1β and IL-6. Additionally, both standard curcumin and CurcuWIN® led to increased BDNF, GAP-43, ICAM-1, and Nrf2 expression. Notably, CurcuWIN® enhanced their expression more than standard curcumin. This data suggests that highly bioavailable curcumin formulation has a beneficial effect on the traumatic brain in mice.
Collapse
Affiliation(s)
- Ayse Arzu Sakul
- Department of Pharmacology, School of Medicine, University of Istanbul Medipol, Istanbul, Turkey
| | - Zeynep Balcikanli
- Department of Physiology, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Nilay Ates Ozsoy
- Department of Pharmacology, School of Medicine, University of Istanbul Medipol, Istanbul, Turkey
- Regenerative and Restorative Medical Research Center, Experimental Neurology Laboratory, Istanbul Medipol University, Istanbul, Turkey
| | - Cemal Orhan
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - Nurhan Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - Mehmet Tuzcu
- Department of Biology, Faculty of Science, Firat University Elazig, Elazig, Turkey
| | - Vijaya Juturu
- Scientific and Clinical Affairs, Research, and Development, OmniActives Health Technologies Inc., Morristown, New Jersey, USA
| | - Ertugrul Kilic
- Department of Physiology, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Kazim Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
49
|
Keezer MR, Bauer PR, Sander JW. The Future: Moving from Phenotypically Defined Diseases Toward Pathophysiological Systems. Can J Neurol Sci 2024; 51:110-112. [PMID: 36855937 DOI: 10.1017/cjn.2023.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Affiliation(s)
- Mark R Keezer
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, H2X 0A9, Canada
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede2103SW, The Netherlands
| | - Prisca R Bauer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Freiburg, University of Freiburg, 79104, Germany
| | - Josemir W Sander
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede2103SW, The Netherlands
- UCL Queen Square Institute of Neurology, London WC1N 3BG, & Chalfont Centre for Epilepsy, Chalfont St Peter, SL9 0RJ, UK
- Neurology Department, West of China Hospital, Sichuan University, Chengdu 61004, China
| |
Collapse
|
50
|
Ichwan K, Gazali S, Suherman S, Desiana D, Nurjannah N. Plasma interleukin 6 as an outcome predictor of traumatic brain injury patients. NARRA J 2023; 3:e234. [PMID: 38455629 PMCID: PMC10919736 DOI: 10.52225/narra.v3i3.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/26/2023] [Indexed: 03/09/2024]
Abstract
Traumatic brain injury is one of the leading causes of death and disability in young adults. Previous studies have suggested that neuroinflammatory process involves the overexpression of interleukin 6 (IL-6); however, data on the predictive ability of IL-6 is limited and conflicting in traumatic head injury patients. The aim of this study was to assess the ability of plasma IL-6 as a predictor of outcome in head injury patients. A cross-sectional study was conducted between June and December 2020 among traumatic head injury patients admitted to Dr. Zainoel Abidin Hospital, Banda Aceh, Indonesia. Demographic, clinical data, and IL-6 level were collected and measured on admission. The outcome was assessed by the Glasgow outcome scale extended (GOSE) in the first- and third-month of post-injury. A total of 50 traumatic brain injury patients were recruited of which 54% were male, 64% had mild head injury, 82% had leukocytosis, and 60% had non-bleeding head CT scan. The mean of IL-6 level was 79.32 pg/mL while the GOSE scores ranged from 1 (death) to 8 (upper good recovery). Early IL-6 level (<24 hours post-injury) was significantly correlated with worse outcome in traumatic head injury, though the correlation strength was moderate (p<0.001; r=-0.42). As a predictor, IL-6 yielded the area under curve (AUC) value of 93.5% (p<0.001) and a cut-off point of 46.33 pg/mL. The sensitivity and specificity of this predictor were 87.5% and 95.24%, respectively. In conclusion, early IL-6 level can be used as a predictor for traumatic head injury. Nevertheless, further multi-center study with a bigger sample size is needed to confirm this finding.
Collapse
Affiliation(s)
- Khairunnisa Ichwan
- Department of Neurology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Indonesia
| | - Syahrul Gazali
- Department of Neurology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Indonesia
| | - Suherman Suherman
- Department of Neurology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Indonesia
| | - Desiana Desiana
- Department of Clinical Pathology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Clinical Pathology, Dr. Zainoel Abidin Hospital, Banda Aceh, Indonesia
| | - Nurjannah Nurjannah
- Department of Public Health, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| |
Collapse
|