1
|
Wang S, Li C, Kang X, Su X, Liu Y, Wang Y, Liu S, Deng X, Huang H, Li T, Lu D, Cai W, Lu Z, Wei L, Lu T. Agomelatine promotes differentiation of oligodendrocyte precursor cells and preserves white matter integrity after cerebral ischemic stroke. J Cereb Blood Flow Metab 2024; 44:1487-1500. [PMID: 38853430 PMCID: PMC11574932 DOI: 10.1177/0271678x241260100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/11/2024]
Abstract
White matter injury contributes to neurological disorders after acute ischemic stroke (AIS). The repair of white matter injury is dependent on the re-myelination by oligodendrocytes. Both melatonin and serotonin antagonist have been proved to protect against post-stroke white matter injury. Agomelatine (AGM) is a multi-functional treatment which is both a melatonin receptor agonist and selective serotonin receptor antagonist. Whether AGM protects against white matter injury after stroke and the underlying mechanisms remain elusive. Here, using the transient middle cerebral artery occlusion (tMCAO) model, we evaluated the therapeutic effects of AGM in stroke mice. Sensorimotor and cognitive functions, white matter integrity, oligodendroglial regeneration and re-myelination in stroke hemisphere after AGM treatment were analyzed. We found that AGM efficiently preserved white matter integrity, reduced brain tissue loss, attenuated long-term sensorimotor and cognitive deficits in tMCAO models. AGM treatment promoted OPC differentiation and enhanced re-myelination both in vitro, ex vivo and in vivo, although OPC proliferation was unaffected. Mechanistically, AGM activated low density lipoprotein receptor related protein 1 (LRP1), peroxisome proliferator-activated receptor γ (PPARγ) signaling thus promoted OPC differentiation and re-myelination after stroke. Inhibition of PPARγ or knock-down of LRP1 in OPCs reversed the beneficial effects of AGM. Altogether, our data indicate that AGM represents a novel therapy against white matter injury after cerebral ischemia.
Collapse
Affiliation(s)
- Shisi Wang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunyi Li
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinmei Kang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaotao Su
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuxin Liu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuge Wang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sanxin Liu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaohui Deng
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huipeng Huang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tiemei Li
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Danli Lu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Cai
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China
| | - Zhengqi Lu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei Wei
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tingting Lu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Sümer Coşkun A, Bülbül M, Çeker T, Özak A, Tanrıöver G, Elif Gürer İ, Tuzcu Balaban H, Göksu E, Aslan M. Protective Effects of Adropin in Experimental Subarachnoid Hemorrhage. Neuroscience 2024; 551:307-315. [PMID: 38851381 DOI: 10.1016/j.neuroscience.2024.05.037] [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: 03/21/2024] [Revised: 05/01/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE We aimed to investigate early effects of exogenously administered adropin (AD) on neurological function, endothelial nitric oxide synthase (eNOS) expression, nitrite/nitrate levels, oxidative stress, and apoptosis in subarachnoid hemorrhage (SAH). METHODS Following intracerebroventricular AD administration (10 µg/5 µl at a rate of 1 µl/min) SAH model was carried out in Sprague-Dawley rats by injection of autologous blood into the prechiasmatic cistern. The effects of AD were assessed 24 h following SAH. The modified Garcia score was employed to evaluate functional insufficiencies. Adropin and caspase-3 proteins were measured by ELISA, while nitrite/nitrate levels, total antioxidant capacity (TAC) and reactive oxygen/nitrogen species (ROS/RNS) were assayed by standard kits. eNOS expression and apoptotic neurons were detected by immunohistochemical analysis. RESULTS The SAH group performed notably lower on the modified Garcia score compared to sham and SAH + AD groups. Adropin administration increased brain eNOS expression, nitrite/nitrate and AD levels compared to SHAM and SAH groups. SAH produced enhanced ROS/RNS generation and reduced antioxidant capacity in the brain. Adropin boosted brain TAC and diminished ROS/RNS production in SAH rats and no considerable change amongst SHAM and SAH + AD groups were detected. Apoptotic cells were notably increased in intensity and number after SAH and were reduced by AD administration. CONCLUSIONS Adropin increases eNOS expression and reduces neurobehavioral deficits, oxidative stress, and apoptotic cell death in SAH model. Presented results indicate that AD provides protection in early brain injury associated with SAH.
Collapse
Affiliation(s)
- Ayşenur Sümer Coşkun
- Division of Anesthesia and Reanimation, Kepez State Hospital, 07320 Antalya, Turkey.
| | - Mehmet Bülbül
- Department of Physiology, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| | - Tuğçe Çeker
- Department of Medical Biochemistry, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| | - Ahmet Özak
- Department of Neurosurgery, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| | - Gamze Tanrıöver
- Department of Histology, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| | - İnanç Elif Gürer
- Department Pathology, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| | - Hazal Tuzcu Balaban
- Department Pathology, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| | - Ethem Göksu
- Department of Neurosurgery, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| | - Mutay Aslan
- Department of Medical Biochemistry, Akdeniz University Faculty of Medicine, Antalya 07070, Turkey.
| |
Collapse
|
3
|
Geraghty JR, Butler M, Maharathi B, Tate AJ, Lung TJ, Balasubramanian G, Testai FD, Loeb JA. Diffuse microglial responses and persistent EEG changes correlate with poor neurological outcome in a model of subarachnoid hemorrhage. Sci Rep 2024; 14:13618. [PMID: 38871799 PMCID: PMC11176397 DOI: 10.1038/s41598-024-64631-2] [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: 02/14/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024] Open
Abstract
The mechanism by which subarachnoid hemorrhage (SAH) leads to chronic neurologic deficits is unclear. One possibility is that blood activates microglia to drive inflammation that leads to synaptic loss and impaired brain function. Using the endovascular perforation model of SAH in rats, we investigated short-term effects on microglia together with long-term effects on EEG and neurologic function for up to 3 months. Within the first week, microglia were increased both at the site of injury and diffusely across the cortex (2.5-fold increase in SAH compared to controls, p = 0.012). Concomitantly, EEGs from SAH animals showed focal increases in slow wave activity and diffuse reduction in fast activity. When expressed as a fast-slow spectral ratio, there were significant interactions between group and time (p < 0.001) with less ipsilateral recovery over time. EEG changes were most pronounced during the first week and correlated with neurobehavioral impairment. In vitro, the blood product hemin was sufficient to increase microglia phagocytosis nearly six-fold (p = 0.032). Immunomodulatory treatment with fingolimod after SAH reduced microglia, improved neurological function, and increased survival. These findings, which parallel many of the EEG changes seen in patients, suggest that targeting neuroinflammation could reduce long-term neurologic dysfunction following SAH.
Collapse
Affiliation(s)
- Joseph R Geraghty
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, 3400 Spruce St, Philadelphia, PA, 19104, USA
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA
| | - Mitchell Butler
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 851 S Morgan St, Chicago, IL, 60607, USA
| | - Biswajit Maharathi
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 851 S Morgan St, Chicago, IL, 60607, USA
| | - Alexander J Tate
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA
- Neuroscience Doctoral Program, Medical College of Wisconsin, Suite H2200, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Tyler J Lung
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA
- The Ohio State University School of Medicine, 1645 Neil Ave, Columbus, OH, 43210, USA
| | - Giri Balasubramanian
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, 851 S Morgan St, Chicago, IL, 60607, USA
| | - Fernando D Testai
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA
| | - Jeffrey A Loeb
- Department of Neurology & Rehabilitation, University of Illinois College of Medicine, 912 S Wood St, NPI Suite 174N, Chicago, IL, 60612, USA.
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, NPI North Bldg., Room 657, M/C 796, 912 S. Wood Street, Chicago, IL, 60612, USA.
| |
Collapse
|
4
|
Hao J, Wang T, Cao C, Li X, Li H, Gao H, Li J, Shen H, Chen G. LPCAT3 exacerbates early brain injury and ferroptosis after subarachnoid hemorrhage in rats. Brain Res 2024; 1832:148864. [PMID: 38484924 DOI: 10.1016/j.brainres.2024.148864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
AIMS Lysophosphatidylcholine acyltransferase 3 (LPCAT3) is known to play a pivotal role in lipid metabolism, but its role in the early brain injury (EBI) following subarachnoid hemorrhage (SAH) remains unclear. This study provides insights into LPCAT3 expression alterations and functional implications in EBI following SAH. METHODS SAH models of adult male Sprague-Dawley (SD) rats were established by intravascular perforation. Lentivirus vectors were administered by intracerebroventricular injection (i.c.v.) to either induce LPCAT3 overexpression or knockdown 14 days before SAH induction. Western blot, immunofluorescence, Nissl staining, MDA detection, ROS detection, iron content detection, and short-term and long-term neurobehavioral tests were performed to investigate the effects of regulated-LPCAT3 after SAH. RESULTS LPCAT3 levels were found to be significantly elevated in SAH. Suppression of LPCAT3 expression via shRNA improved oxidative stress, reduced brain edema, alleviated behavioral and cognitive deficits following SAH and decreased neuronal death, while upregulating LPCAT3 expression showed opposing effects. CONCLUSION LPCAT3 is involved in SAH-induced EBI and associated with ferroptosis. Our findings provide a referential basis for potential therapeutic interventions aimed at alleviating EBI following SAH.
Collapse
Affiliation(s)
- Jiahui Hao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Tong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Intensive Care Unit, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China; Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Heng Gao
- Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China
| | - Jinquan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
5
|
Li C, Ajmal E, Alok K, Powell K, Wadolowski S, Tambo W, Turpin J, Barthélemy E, Al-Abed Y, LeDoux D. CGRP as a potential mediator for the sexually dimorphic responses to traumatic brain injury. Biol Sex Differ 2024; 15:44. [PMID: 38816868 PMCID: PMC11138127 DOI: 10.1186/s13293-024-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND The outcomes of traumatic brain injury (TBI) exhibit variance contingent upon biological sex. Although female sex hormones exert neuroprotective effects, the administration of estrogen and progesterone has not yielded conclusive results. Hence, it is conceivable that additional mediators, distinct from female sex hormones, merit consideration due to their potential differential impact on TBI outcomes. Calcitonin gene-related peptide (CGRP) exhibits sexually dimorphic expression and demonstrates neuroprotective effects in acute brain injuries. In this study, we aimed to examine sex-based variations in TBI structural and functional outcomes with respect to CGRP expression. METHODS Male and female Sprague Dawley rats were exposed to controlled cortical impact to induce severe TBI, followed by interventions with and without CGRP inhibition. In the acute phase of TBI, the study centered on elucidating the influence of CGRP on oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) and endothelial nitric oxide synthase (eNOS) signaling in the peri-impact tissue. Subsequently, during the chronic phase of TBI, the investigation expanded to evaluate CGRP expression in relation to lesion volume, microvascular dysfunction, and white matter injury, as well as working and spatial memory, anxiety-like, and depression-like behaviors in subjects of both sexes. RESULTS Female rats exhibited elevated levels of CGRP in the peri-impact brain tissue during both baseline conditions and in the acute and chronic phases of TBI, in comparison to age-matched male counterparts. Enhanced CGRP levels in specific brain sub-regions among female rats correlated with superior structural and functional outcomes following TBI compared to their male counterparts. CGRP inhibition induced heightened oxidative stress and a reduction in the expression of Nrf2 and eNOS in both male and female rats, with the observed alteration being more pronounced in females than in males. CONCLUSIONS This study marks the inaugural identification of CGRP as a downstream mediator contributing to the sexually dimorphic response observed in TBI outcomes.
Collapse
Affiliation(s)
- Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA.
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
| | - Erum Ajmal
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Khaled Alok
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Keren Powell
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Steven Wadolowski
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Willians Tambo
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Justin Turpin
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Ernest Barthélemy
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Yousef Al-Abed
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - David LeDoux
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| |
Collapse
|
6
|
Kanamaru H, Zhu S, Dong S, Takemoto Y, Huang L, Sherchan P, Suzuki H, Tang J, Zhang JH. UDP-Glucose/P2Y14 Receptor Signaling Exacerbates Neuronal Apoptosis After Subarachnoid Hemorrhage in Rats. Stroke 2024; 55:1381-1392. [PMID: 38525592 PMCID: PMC11039370 DOI: 10.1161/strokeaha.123.044422] [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: 07/05/2023] [Accepted: 02/13/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a severe subtype of stroke with poor outcomes. Abnormal glucose metabolism often occurs after SAH, but the strict control of blood glucose levels is not always beneficial. This study aimed to investigate the contribution of uridine diphosphate glucose (UDP-G), an intermediate of glucose/glycogen metabolism, and its receptor P2Y14 (P2Y purinoceptor 14) to SAH pathology and explored the potential targeted treatments in rats. METHODS A total of 218 Sprague-Dawley male rats were used. SAH was induced by endovascular perforation. Brain expressions of P2Y14, uridine diphosphate glucose (UDP-G), and its converting enzyme UGP2 (UDP-G pyrophosphorylase-2) were evaluated. Exogenous UDP-G or selective P2Y14 inhibitor was administered intranasally at 1 hour after SAH to explore their potential effects. Intranasal Ugp2 or P2ry14 siRNA was delivered 24 hours before SAH for mechanistic evaluation. Primary neuron culture and hemoglobin stimulation were used as in vitro model of SAH. Post-SAH evaluation included liquid chromatography-mass spectrometry measurement of brain endogenous UDP-G level, neurobehavioral assessments, Western blotting, immunohistochemistry, TUNEL staining, and Nissl staining. RESULTS There was an acute elevation of endogenous brain UDP-G and UGP2 after SAH, and P2Y14 was expressed in neurons. Although P2Y14 inhibitor decreased neurological dysfunction, neuronal apoptosis, and proapoptotic molecules, exogenous UDP-G exacerbated these outcomes at 24 hours after SAH. Early inhibition of P2Y14 preserved long-term neuronal survival in the hippocampus, amygdala, and cortex with improved neurocognition and depressive-like behavior. In addition, in vivo knockdown of Ugp2- and P2ry14-reduced neurological deficits and proapoptotic molecules at 24 hours after SAH, and furthermore in vitro knockdown of P2ry14-reduced apoptosis in hemoglobin stimulated primary neuron. CONCLUSIONS These findings suggest a detrimental role of brain UDP-G/P2Y14 signaling in SAH, as a part of glucose metabolic pathology at the tissue level. P2Y14 inhibitor 4-[4-(4-piperidinyl)phenyl]-7-[4-(trifluoromethyl)phenyl]-2-naphthalenecarboxylic acid hydrochloride may serve as a potential therapeutic target in treating patients with SAH.
Collapse
Affiliation(s)
- Hideki Kanamaru
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan (H.K., H.S.)
| | - Shiyi Zhu
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
| | - Siyuan Dong
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
| | - Yushin Takemoto
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
- Department of Neurosurgery, Kumamoto University School of Medicine, Japan (Y.T.)
| | - Lei Huang
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
- Neurosurgery, (L.H., J.H.Z.), Loma Linda University, CA
| | - Prativa Sherchan
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan (H.K., H.S.)
| | - Jiping Tang
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
| | - John H Zhang
- Departments of Physiology and Pharmacology (H.K., S.Z., S.D., Y.T., L.H., P.S., J.T., J.H.Z.,), Loma Linda University, CA
- Neurosurgery, (L.H., J.H.Z.), Loma Linda University, CA
- Anesthesiology (J.H.Z.), Loma Linda University, CA
| |
Collapse
|
7
|
Morelli M, Adcock J, Yim TW, Rook J, Mocco J, Brophy C, Cheung-Flynn J. The Cell Permeant Phosphopetpide mimetic of VASP Alleviates Motor Function Deficits After Experimental Subarachnoid Hemorrhage. J Mol Neurosci 2024; 74:9. [PMID: 38214771 DOI: 10.1007/s12031-023-02180-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 01/13/2024]
Abstract
Subarachnoid hemorrhage (SAH) due to the rupture of an intracranial aneurysm leads to delayed vasospasm and neuroischemia, which can result in profound neurologic deficit and death. Therapeutic options after SAH are currently limited to hemodynamic optimization and nimodipine, which have limited clinical efficacy. Experimental SAH results in cerebral vasospasm have demonstrated the downregulation of nitric oxide (NO)-protein kinase G (PKG) signaling elements. VP3 is a novel cell permeant phosphopeptide mimetic of VASP, a substrate of PKG and an actin-associated protein that modulates vasorelaxation in vascular smooth muscle cells. In this study, we determined that intravenous administration of high doses of VP3 did not induce systemic hypotension in rats except at the maximal soluble dose, implying that VP3 is well-tolerated and has a wide therapeutic window. Using a single cisterna magna injection rat model of SAH, we demonstrated that intravenous administration of low-dose VP3 after SAH improved neurologic deficits for up to 14 days as determined by the rotarod test. These findings suggest that strategies aimed at targeting the cerebral vasculature with VP3 may improve neurologic deficits associated with SAH.
Collapse
Affiliation(s)
- Madeleine Morelli
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jamie Adcock
- Division of Surgical Research, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tsz Wing Yim
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jerri Rook
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - J Mocco
- Cerebrovascular Center, Department of Neurosurgery, Mount Sinai Health System, New York, NY, USA
| | - Colleen Brophy
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joyce Cheung-Flynn
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
8
|
Gu J, Lu J, Yang J, Liu Y, Zhu X, Zhang J, Shen H, Li X, Yu Z, Li H. Norad Competently Binds with Pum2 to Regulate Neuronal Apoptosis and Play a Neuroprotective Role After SAH in Mice. Neuroscience 2023; 535:108-123. [PMID: 37913857 DOI: 10.1016/j.neuroscience.2023.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 11/03/2023]
Abstract
Subarachnoid Hemorrhage (SAH) is a cerebrovascular disorder that has been found to have severe consequences, including a high mortality and disability rate. Research has indicated that neuronal death, particularly apoptosis, plays a major role in the neurological impairment that follows SAH. RNA-binding protein Pum2 can interfere with translation or other biological functions by connecting to the UGUAHAUA sequence on RNA. Noncoding RNA activated by DNA damage (Norad) contains some Pum2 recognition sequences, which may bind to Pum2 protein and affect its capacity to attach to target mRNA. The time course expression of Norad and Pum2 after SAH is analyzed by establishing a mouse SAH model. Subsequently, the purpose of this study is to investigate the potential role and mechanism of the Norad-Pum2 axis after SAH using lentivirus overexpression of Pum2 and knockdown of Norad. Analysis of Pum2 and Norad levels reveal that the former is significantly reduce and the latter is significantly increased in the SAH group compared to the sham group. Subsequent overexpression of Pum2 and Norad knockdown is found to reduce SAH-induced oxidative stress, neuronal apoptosis, and ultimately improve behavioral and cognitive changes in SAH mice. Our study indicates that Norad-Pum2 acts as a neuromodulator in SAH, and that by increasing Pum2 and decreasing Norad levels, SAH-induced neuronal apoptosis can be reduced and neurological deficits alleviated. Consequently, Norad-Pum2 may be a promising therapeutic target for SAH.
Collapse
Affiliation(s)
- Junyi Gu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Jian Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Yangyang Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xunan Zhu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
9
|
Cao C, Lu T, Cheng Q, Cui G, Wang Z, Li X, Li H, Gao H, Shen H, Sun Q. Restoring System xc- activity by xCT overexpression inhibited neuronal ferroptosis and improved neurological deficits after experimental subarachnoid hemorrhage. Brain Res 2023; 1820:148556. [PMID: 37648093 DOI: 10.1016/j.brainres.2023.148556] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Ferroptosis is an important therapeutic target to alleviate early brain injury (EBI) after subarachnoid hemorrhage (SAH), yet the mechanism of neuronal ferroptosis after SAH remains unclear. System xc- dysfunction is one of the key pathways to induce ferroptosis. System xc- activity is mainly regulated by the expression of xCT. This study was designed to investigate the effect of xCT expression and System xc- activity on ferroptosis and EBI in an experimental SAH model both in vitro and in vivo. METHODS SAH was induced in adult male Sprague-Dawley rats by injecting autologous blood into the prechiasmatic cistern. Primary neurons treated with oxyhemoglobin (10 µM) were used to mimic SAH in vitro. Plasmid transfection was used to induce xCT overexpression. Western blotting, immunofluorescence staining, measurement of cystine uptake, enzyme-linked immunosorbent assay, transmission electron microscopy, Nissl staining, and a series of neurobehavioral tests were conducted to explore the role of xCT and System xc- activity in ferroptosis and EBI after SAH. RESULTS We found that System xc- dysfunction induced ferroptosis and exacerbated EBI after SAH in rats. xCT deficiency after SAH resulted in System xc- dysfunction, weakened neuronal antioxidant capacity and activated neuronal ferroptosis. xCT overexpression improved neuronal antioxidant capacity and inhibited neuronal ferroptosis by restoring System xc- activity. Rats with xCT overexpression after SAH presented with attenuated brain edema and inflammation, increased neuronal survival, and ameliorated neurological deficits. CONCLUSIONS Our study revealed that restoring System xc- activity by xCT overexpression inhibited neuronal ferroptosis and EBI and improved neurological deficits after SAH.
Collapse
Affiliation(s)
- Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Intensive Care Unit, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China; Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China.
| | - Ting Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Qian Cheng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Heng Gao
- Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China.
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| |
Collapse
|
10
|
Szczygielski J, Hubertus V, Kruchten E, Müller A, Albrecht LF, Schwerdtfeger K, Oertel J. Prolonged course of brain edema and neurological recovery in a translational model of decompressive craniectomy after closed head injury in mice. Front Neurol 2023; 14:1308683. [PMID: 38053795 PMCID: PMC10694459 DOI: 10.3389/fneur.2023.1308683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Background The use of decompressive craniectomy in traumatic brain injury (TBI) remains a matter of debate. According to the DECRA trial, craniectomy may have a negative impact on functional outcome, while the RescueICP trial revealed a positive effect of surgical decompression, which is evolving over time. This ambivalence of craniectomy has not been studied extensively in controlled laboratory experiments. Objective The goal of the current study was to investigate the prolonged effects of decompressive craniectomy (both positive and negative) in an animal model. Methods Male mice were assigned to the following groups: sham, decompressive craniectomy, TBI and TBI followed by craniectomy. The analysis of functional outcome was performed at time points 3d, 7d, 14d and 28d post trauma according to the Neurological Severity Score and Beam Balance Score. At the same time points, magnetic resonance imaging was performed, and brain edema was analyzed. Results Animals subjected to both trauma and craniectomy presented the exacerbation of the neurological impairment that was apparent mostly in the early course (up to 7d) after injury. Decompressive craniectomy also caused a significant increase in brain edema volume (initially cytotoxic with a secondary shift to vasogenic edema and gliosis). Notably, delayed edema plus gliosis appeared also after decompression even without preceding trauma. Conclusion In prolonged outcomes, craniectomy applied after closed head injury in mice aggravates posttraumatic brain edema, leading to additional functional impairment. This effect is, however, transient. Treatment options that reduce brain swelling after decompression may accelerate neurological recovery and should be explored in future experiments.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Instutute of Neuropathology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Medical Sciences, University of Rzeszów, Rzeszow, Poland
| | - Vanessa Hubertus
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Department of Neurosurgery, Charité University Medicine, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| | - Eduard Kruchten
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Interventional and Diagnostic Radiology, Karlsruhe, Germany
| | - Andreas Müller
- Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Lisa Franziska Albrecht
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| |
Collapse
|
11
|
Peng J, He Y, He J, Zhang J, Yu Z, Xia Y. GPR30 agonist G1 combined with hypothermia alleviates cognitive impairment and anxiety-like behavior after subarachnoid hemorrhage in rats. Brain Behav 2023; 13:e3204. [PMID: 37548479 PMCID: PMC10570468 DOI: 10.1002/brb3.3204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/08/2023] [Accepted: 07/08/2023] [Indexed: 08/08/2023] Open
Abstract
INTRODUCTION This study aimed to investigate the treatment effect of G protein-coupled receptor 30 (GPR30) agonist G1 combined with hypothermia (HT) on cognitive impairment and anxiety-like behavior after subarachnoid hemorrhage (SAH) in rats. METHODS Fifty male rats were randomly assigned to one of five groups: Sham group, SAH group, SAH + G1 group, SAH + HT group, and SAH + G1 + HT group. The SAH rat model was established by modified endovascular puncture in all groups except the Sham group. Neurological function after the operation was assessed by Garcia scoring. The degree of rat cerebral edema was determined using dry-wet weighing method on the 28th day after operation. Moreover, the behavioral test was performed on rats on the 4th and 28th days after operation. RESULTS Compared with Sham group, the Garcia score of each SAH rat model group decreased significantly on the first day and thereafter increased gradually. However, the recovery rate of each treatment group was higher than the SAH group (no treatment), and the Garcia score of SAH + G1 + HT group was much higher than the SAH group on the seventh day after operation. In addition, each treatment group could obviously reduce the cerebral edema degree of SAH rats, among which rats in SAH + G1 + HT group had lower cerebral edema degree than SAH + G1 group and SAH + HT group. Behavioral test results showed that the combination of GPR30 agonist G1 and HT markedly improved the learning and memory ability of SAH rats, alleviated their anxiety- and emotion-related behavior, and enhanced their social interaction. CONCLUSION GPR30 agonist G1 combined with HT reduces cognitive impairment and anxiety-like behavior in rats with SAH.
Collapse
Affiliation(s)
- Jun Peng
- Department of neurosurgeryHaikou Affiliated Hospital of Central South University Xiangya School of MedicineHaikouHainanChina
| | - Yang He
- Department of NeurologyHainan Medical College First Affiliated HospitalHaikouHainanChina
| | - Jun He
- Department of neurosurgeryHaikou Affiliated Hospital of Central South University Xiangya School of MedicineHaikouHainanChina
| | - Ji‐kun Zhang
- Department of neurosurgeryHaikou Affiliated Hospital of Central South University Xiangya School of MedicineHaikouHainanChina
| | - Zheng‐tao Yu
- Department of neurosurgeryHaikou Affiliated Hospital of Central South University Xiangya School of MedicineHaikouHainanChina
| | - Ying Xia
- Department of neurosurgeryHaikou Affiliated Hospital of Central South University Xiangya School of MedicineHaikouHainanChina
| |
Collapse
|
12
|
Patsouris V, Blecharz-Lang KG, Nieminen-Kelhä M, Schneider UC, Vajkoczy P. Resolution of Cerebral Inflammation Following Subarachnoid Hemorrhage. Neurocrit Care 2023; 39:218-228. [PMID: 37349601 PMCID: PMC10499726 DOI: 10.1007/s12028-023-01770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/31/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Aneurismal subarachnoid hemorrhage (SAH) is a type of hemorrhagic stroke that, despite improvement through therapeutic interventions, remains a devastating cerebrovascular disorder that has a high mortality rate and causes long-term disability. Cerebral inflammation after SAH is promoted through microglial accumulation and phagocytosis. Furthermore, proinflammatory cytokine release and neuronal cell death play key roles in the development of brain injury. The termination of these inflammation processes and restoration of tissue homeostasis are of utmost importance regarding the possible chronicity of cerebral inflammation and the improvement of the clinical outcome for affected patients post SAH. Thus, we evaluated the inflammatory resolution phase post SAH and considered indications for potential tertiary brain damage in cases of incomplete resolution. METHODS Subarachnoid hemorrhage was induced through endovascular filament perforation in mice. Animals were killed 1, 7 and 14 days and 1, 2 and 3 months after SAH. Brain cryosections were immunolabeled for ionized calcium-binding adaptor molecule-1 to detect microglia/macrophages. Neuronal nuclei and terminal deoxyuridine triphosphate-nick end labeling staining was used to visualize secondary cell death of neurons. The gene expression of various proinflammatory mediators in brain samples was analyzed by quantitative polymerase chain reaction. RESULTS We observed restored tissue homeostasis due to decreased microglial/macrophage accumulation and neuronal cell death 1 month after insult. However, the messenger RNA expression levels of interleukin 6 and tumor necrosis factor α were still elevated at 1 and 2 months post SAH, respectively. The gene expression of interleukin 1β reached its maximum on day 1, whereas at later time points, no significant differences between the groups were detected. CONCLUSIONS By the herein presented molecular and histological data we provide an important indication for an incomplete resolution of inflammation within the brain parenchyma after SAH. Inflammatory resolution and the return to tissue homeostasis represent an important contribution to the disease's pathology influencing the impact on brain damage and outcome after SAH. Therefore, we consider a novel complementary or even superior therapeutic approach that should be carefully rethought in the management of cerebral inflammation after SAH. An acceleration of the resolution phase at the cellular and molecular levels could be a potential aim in this context.
Collapse
Affiliation(s)
- Victor Patsouris
- Institute of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Kinga G Blecharz-Lang
- Institute of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Institute of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Ulf C Schneider
- Department of Neurosurgery, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Peter Vajkoczy
- Institute of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
13
|
The protective effect of low-dose minocycline on brain microvascular ultrastructure in a rodent model of subarachnoid hemorrhage. Histochem Cell Biol 2023; 159:91-114. [PMID: 36153470 PMCID: PMC9899762 DOI: 10.1007/s00418-022-02150-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 02/07/2023]
Abstract
The multifaceted nature of subarachnoid hemorrhage (SAH) pathogenesis is poorly understood. To date, no pharmacological agent has been found to be efficacious for the prevention of brain injury when used for acute SAH intervention. This study was undertaken to evaluate the beneficial effects of low-dose neuroprotective agent minocycline on brain microvascular ultrastructures that have not been studied in detail. We studied SAH brain injury using an in vivo prechiasmatic subarachnoid hemorrhage rodent model. We analyzed the qualitative and quantitative ultrastructural morphology of capillaries and surrounding neuropil in the rodent brains with SAH and/or minocycline administration. Here, we report that low-dose minocycline (1 mg/kg) displayed protective effects on capillaries and surrounding cells from significant SAH-induced changes. Ultrastructural morphology analysis revealed also that minocycline stopped endothelial cells from abnormal production of vacuoles and vesicles that compromise blood-brain barrier (BBB) transcellular transport. The reported ultrastructural abnormalities as well as neuroprotective effects of minocycline during SAH were not directly mediated by inhibition of MMP-2, MMP-9, or EMMPRIN. However, SAH brain tissue treated with minocycline was protected from development of other morphological features associated with oxidative stress and the presence of immune cells in the perivascular space. These data advance the knowledge on the effect of SAH on brain tissue ultrastructure in an SAH rodent model and the neuroprotective effect of minocycline when administered in low doses.
Collapse
|
14
|
Kim HY, Back DB, Choi BR, Choi DH, Kwon KJ. Rodent Models of Post-Stroke Dementia. Int J Mol Sci 2022; 23:ijms231810750. [PMID: 36142661 PMCID: PMC9501431 DOI: 10.3390/ijms231810750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Post-stroke cognitive impairment is one of the most common complications in stroke survivors. Concomitant vascular risk factors, including aging, diabetes mellitus, hypertension, dyslipidemia, or underlying pathologic conditions, such as chronic cerebral hypoperfusion, white matter hyperintensities, or Alzheimer’s disease pathology, can predispose patients to develop post-stroke dementia (PSD). Given the various clinical conditions associated with PSD, a single animal model for PSD is not possible. Animal models of PSD that consider these diverse clinical situations have not been well-studied. In this literature review, diverse rodent models that simulate the various clinical conditions of PSD have been evaluated. Heterogeneous rodent models of PSD are classified into the following categories: surgical technique, special structure, and comorbid condition. The characteristics of individual models and their clinical significance are discussed in detail. Diverse rodent models mimicking the specific pathomechanisms of PSD could provide effective animal platforms for future studies investigating the characteristics and pathophysiology of PSD.
Collapse
Affiliation(s)
- Hahn Young Kim
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea
- Correspondence: ; Tel.: +82-2-2030-7563; Fax: +82-2-2030-5169
| | - Dong Bin Back
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea
| | - Bo-Ryoung Choi
- Department of Neurology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea
| | - Dong-Hee Choi
- Department of Medicine, Konkuk University School of Medicine, Seoul 05030, Korea
| | - Kyoung Ja Kwon
- Department of Medicine, Konkuk University School of Medicine, Seoul 05030, Korea
| |
Collapse
|
15
|
Cao C, Ding J, Cao D, Li B, Wu J, Li X, Li H, Cui G, Shen H, Chen G. TREM2 modulates neuroinflammation with elevated IRAK3 expression and plays a neuroprotective role after experimental SAH in rats. Neurobiol Dis 2022; 171:105809. [PMID: 35781003 DOI: 10.1016/j.nbd.2022.105809] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 06/20/2022] [Accepted: 06/26/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The modulation of neuroinflammation is a new direction that may alleviate the early brain injury after subarachnoid hemorrhage (SAH). Brain resident microglia/macrophages (Mi/MΦ) are the key drivers of neuroinflammation. Triggering receptor expressed on myeloid cells 2 (TREM2) has been reported to play a neuroprotective role by activating phagocytosis and suspending inflammatory response in experimental ischemic stroke and intracerebral hemorrhage. This study was designed to investigate the role of TREM2 on neuroinflammation and neuroprotective effects in a rat SAH model. METHODS Adult male Sprague-Dawley rats were induced SAH through endovascular perforation. Lentivirus vectors were administered by i.c.v. to induce TREM2 overexpression or knockdown 7 days before SAH induction. Short- and long-term neurobehavioral tests, western blotting, immunofluorescence, enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick end labeling and Nissl staining were performed to explore the neuroprotective role of TREM2 after SAH. RESULTS The expression of TREM2 elevated in a rat SAH model with a peak at 48 h after SAH and mainly expressed in Mi/MΦ in brain. TREM2 overexpression improved short- and long-term neurological deficits induced by SAH in rats, while TREM2 knockdown worsened neurological dysfunction. The rats with TREM2 overexpressed presented less neuronal apoptosis and more neuronal survival at 48 h after SAH, while the rats with TREM2 knockdown presented on the contrary. TREM2 overexpression manifested activated phagocytosis and suppressed inflammatory response, with the increase of CD206+/CD11b+ cells and IL-10 expression as well as the decrease of the infiltration of MPO+ cells and the expression of TNF-α, IL-1β. While TREM2 knockdown abolished these effects. The protein level of IRAK3, a negative regulatory factor of inflammation, was significantly elevated after TREM2 overexpression and declined after TREM2 knockdown. CONCLUSIONS Our research suggested TREM2 played a neuroprotective role and improved the short- and long-term neurological deficits by modulating neuroinflammation after SAH. The modulation on neuroinflammation of TREM2 after SAH was related with the elevated protein level of IRAK3.
Collapse
Affiliation(s)
- Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Neurocritical Intensive Care Unit, The Affiliated Jiangyin Hospital, School of Medicine, Southeast University, Jiangyin City 214400, Jiangsu Province, China
| | - Jiasheng Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Demao Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Bing Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
16
|
Regnier-Golanov AS, Gulinello M, Hernandez MS, Golanov EV, Britz GW. Subarachnoid Hemorrhage Induces Sub-acute and Early Chronic Impairment in Learning and Memory in Mice. Transl Stroke Res 2022; 13:625-640. [PMID: 35260988 DOI: 10.1007/s12975-022-00987-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/20/2022]
Abstract
Subarachnoid hemorrhage (SAH) leads to significant long-term cognitive deficits, so-called the post-SAH syndrome. Existing neurological scales used to assess outcomes of SAH are focused on sensory-motor functions. To better evaluate short-term and chronic consequences of SAH, we explored and validated a battery of neurobehavioral tests to gauge the functional outcomes in mice after the circle of Willis perforation-induced SAH. The 18-point Garcia scale, applied up to 4 days, detected impairment only at 24-h time point and showed no significant difference between the Sham and SAH group. A decrease in locomotion was detected at 4-days post-surgery in the open field test but recovered at 30 days in Sham and SAH groups. However, an anxiety-like behavior undetected at 4 days developed at 30 days in SAH mice. At 4-days post-surgery, Y-maze revealed an impairment in working spatial memory in SAH mice, and dyadic social interactions showed a decrease in the sociability in SAH mice, which spent less time interacting with the stimulus mouse. At 30 days after ictus, SAH mice displayed mild spatial learning and memory deficits in the Barnes maze as they committed significantly more errors and used more time to find the escape box but still were able to learn the task. We also observed cognitive dysfunction in the SAH mice in the novel object recognition test. Taken together, these data suggest dysfunction of the limbic system and hippocampus in particular. We suggest a battery of 5 basic behavioral tests allowing to detect neurocognitive deficits in a sub-acute and chronic phase following the SAH.
Collapse
Affiliation(s)
| | - M Gulinello
- Rodent Behavior Core, Department of Neuroscience, Albert Einstein University, Bronx, NY, 10461, USA
| | - M S Hernandez
- Department of Neurosurgery, Houston Methodist Hospital, Houston, USA
| | - E V Golanov
- Department of Neurosurgery, Houston Methodist Hospital, Houston, USA
| | - G W Britz
- Department of Neurosurgery, Houston Methodist Hospital, Houston, USA.
| |
Collapse
|
17
|
Liu Q, Hou C, Zhang H, Fu C, Wang W, Wang B, Li J, Zhao Y, Yang X. Impaired meningeal lymphatic vessels exacerbate early brain injury after experimental subarachnoid hemorrhage. Brain Res 2021; 1769:147584. [PMID: 34303696 DOI: 10.1016/j.brainres.2021.147584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/26/2021] [Accepted: 07/14/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND PURPOSE Blood that enters the subarachnoid space (SAS) and its breakdown products are neurotoxic and are the principal inducers of brain injury after subarachnoid hemorrhage (SAH). Recently, meningeal lymphatic vessels (MLVs) have been proven to play an important role in clearing erythrocytes that arise from SAH, as well as other macromolecular solutes. However, evidence demonstrating the relationship between MLVs and brain injury after SAH is still limited. Therefore, we performed this study to observe the effects of meningeal lymphatic impairment on early brain injury (EBI) after experimental SAH. METHODS The MLVs of C57BL/6 male adult mice were ablated by injecting Visudyne into the cisterna magna and transcranially photoconverting it with laser light. The MLVs were then examined by immunofluorescence staining for lyve-1. Next, both the MLV-ablated group and the control group (normal mice) underwent filament perforation to model SAH or sham operation. We assessed the cortical perfusion of all the mice before SAH induction, 5 min after SAH and 24 h after SAH. In addition, we evaluated neurological function deficits by Garcia scores and measured brain water content at 24 h post SAH. Then, neuroinflammation and neural apoptosis in the mouse brain were also examined. RESULTS Visudyne and transcranial photoconversion treatment notably ablated mouse MLVs. Five minutes after SAH induction, cortical perfusion was significantly impaired, and after 24 h, this impairment was ameliorated considerably in the control group but ameliorated only slightly or worsened in the MLV-ablated group. Additionally, the MLVablated group presented worse neurological function deficits and more severe brain edema than the control group. More notably, neuroinflammation and neural apoptosis were also observed. CONCLUSION Ablation of MLVs by Visudyne treatment exacerbated EBI after experimental SAH in mice. The worsening of EBI may have arisen from limited drainage of blood and other breakdown products, which are thought to cause brain edema, neuroinflammation, neuronal apoptosis and other pathological processes.
Collapse
Affiliation(s)
- Quanlei Liu
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Changkai Hou
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Hao Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Cong Fu
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Weihan Wang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China
| | - Bangyue Wang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Jian Li
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Yan Zhao
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China
| | - Xinyu Yang
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, China; Department of Neurosurgery, Tianjin Medical University General Hospital, China.
| |
Collapse
|
18
|
Zheng ZY, Lu G, Xiong ZQ, Leung CK, Su XW, Li T, Poon WS, Chan WY, Wong GKC. Integrated analysis of gait parameters and gene expression profiles in a murine model of subarachnoid hemorrhage. GENES BRAIN AND BEHAVIOR 2021; 20:e12728. [PMID: 33641236 DOI: 10.1111/gbb.12728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/02/2021] [Accepted: 02/26/2021] [Indexed: 11/28/2022]
Abstract
Gait analysis has been widely used to examine the behavioral presentation of numerous neurological disorders. Thorough murine model evaluation of the subarachnoid hemorrhage (SAH)-associated gait deficits is missing. This study measures gait deficits using a clinically relevant murine model of SAH to examine associations between gait variability and SAH-associated gene expressions. A total of 159 dynamic and static gait parameters from the endovascular perforation murine model for simulating clinical human SAH were determined using the CatWalk system. Eighty gait parameters and the mRNA expression levels of 35 of the 88 SAH-associated genes were differentially regulated in the diseased models. Totals of 42 and 38 gait parameters correlated with the 35 SAH-associated genes positively and negatively with Pearson's correlation coefficients of >0.7 and <-0.7, respectively. p-SP1453 expression in the motor cortex in SAH animal models displays a significant correlation with a subset of gait parameters associated with muscular strength and coordination of limb movements. Our data highlights a strong correlation between gait variability and SAH-associated gene expression. p-SP1453 expression could act as a biomarker to monitor SAH pathological development and a therapeutic target for SAH.
Collapse
Affiliation(s)
- Zhi Yuan Zheng
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhi Qiang Xiong
- Bioinformatics Unit, SDIVF R&D Centre, Hong Kong Science and Technology Parks, Hong Kong, China
| | - Chi Kwan Leung
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xian Wei Su
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Tu Li
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - George Kwok Chu Wong
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
19
|
Turan N, Heider RA, Nadeem M, Miller BA, Wali B, Yousuf S, Sayeed I, Stein DG, Pradilla G. Neurocognitive Outcomes in a Cisternal Blood Injection Murine Model of Subarachnoid Hemorrhage. J Stroke Cerebrovasc Dis 2020; 29:105249. [PMID: 33066928 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/13/2020] [Accepted: 08/10/2020] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) results in neurocognitive dysfunction and anxiety in humans and in animal models. Neurobehavioral tests such as the Morris Water Maze (MWM) and Elevated Plus Maze (EPM) tests are validated in several models of SAH but have not been tested in the murine cisternal blood injection SAH model. METHODS Adult C57BL/6 mice (n=16) were randomized into two groups. Group 1 (n=8) received sham surgery. Group 2 (n=8) underwent SAH with 60 µL of autologous blood injected into the cisterna magna. Mice were then tested using the Modified Garcia Score on post-operative day 2 (POD2), EPM on POD5 & POD16, and MWM on POD6-16.Brain tissues harvested on POD16 were stained with Fluoro-Jade C to identify neurodegeneration in the hippocampus and cortex and Iba-1 immunofluorescence staining for microglial activation in the dentate gyrus and CA1 region of the hippocampus. RESULTS SAH mice showed increased escape latency on POD10. Swim distance was significantly increased on POD9-10 and swim speed was significantly decreased on POD6&POD10 in SAH mice. SAH mice exhibited a trend for lowered proportion of covered arm entries in EPM on POD16. Modified Garcia Score was not significantly different between the groups on POD2. The area of microglial activation in the dentate gyrus and CA1 region of the hippocampus was mildly increased but not significantly different at day 16 after SAH. Similarly, no significant differences were noted in the number of Fluoro-Jade C (+) cells in cortex or hippocampus. CONCLUSIONS Cisternal single blood injection in mice produces mild neurocognitive deficits most pronounced in spatial learning and most evident 10 days after SAH.
Collapse
Affiliation(s)
- Nefize Turan
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA; Cerebrovascular Research Laboratory, Department of Neurosurgery, Atlanta, GA, USA
| | - Robert A Heider
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA
| | - Maheen Nadeem
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA
| | - Brandon A Miller
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA; Cerebrovascular Research Laboratory, Department of Neurosurgery, Atlanta, GA, USA
| | - Bushra Wali
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Seema Yousuf
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Iqbal Sayeed
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Donald G Stein
- Emory University School of Medicine, Department of Emergency Medicine, Atlanta, GA, USA
| | - Gustavo Pradilla
- Emory University School of Medicine, Department of Neurosurgery, Atlanta, GA, USA; Cerebrovascular Research Laboratory, Department of Neurosurgery, Atlanta, GA, USA.
| |
Collapse
|
20
|
Matsumura K, Kumar TP, Guddanti T, Yan Y, Blackburn SL, McBride DW. Neurobehavioral Deficits After Subarachnoid Hemorrhage in Mice: Sensitivity Analysis and Development of a New Composite Score. J Am Heart Assoc 2020; 8:e011699. [PMID: 30971151 PMCID: PMC6507191 DOI: 10.1161/jaha.118.011699] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Because of the failure of numerous clinical trials, various recommendations have been made to improve the usefulness of preclinical studies. Specifically, the STAIR (Stroke Therapy Academic Industry Roundtable) recommendations highlighted functional outcome as a critical measure. Recent reviews of experimental subarachnoid hemorrhage (SAH) studies have brought to light the numerous neurobehavioral scoring systems that are used in preclinical SAH studies. To gain insight into the utility of these scoring systems, as well as to identify a scoring system that best captures the deficits caused by SAH in mice, we designed the current study. Methods and Results Adult male C57BL/6J mice were used. One cohort of mice was randomly allocated to either sham or SAH and had functional testing performed on days 1 to 3 post‐SAH using the modified Bederson Score, Katz Score, Garcia Neuroscore, and Parra Neuroscore, as well as 21 individual subtests. A new composite neuroscore was developed using the 8 most diagnostically accurate subtests. To validate the use of the developed composite neuroscore, another cohort of mice was randomly assigned to either the sham or SAH group and neurobehavior was evaluated on days 1 to 3, 5, and 7 after injury. Receiver operating characteristic curves were used to analyze the diagnostic accuracy of each scoring system, as well as the subtests. Of the 4 published scoring systems, the Parra Neuroscore was diagnostically accurate for SAH injury in mice versus the modified Bederson and Katz Scores, but not the Garcia Neuroscore. However, the newly developed composite neuroscore was found to be statistically more diagnostically accurate than even the Parra Neuroscore. Conclusions The findings of this study promote use of the newly developed composite neuroscore for experimental SAH studies in mice.
Collapse
Affiliation(s)
- Kanako Matsumura
- 1 The Vivian L. Smith Department of Neurosurgery McGovern Medical School The University of Texas Health Science Center at Houston Houston TX
| | - T Peeyush Kumar
- 1 The Vivian L. Smith Department of Neurosurgery McGovern Medical School The University of Texas Health Science Center at Houston Houston TX
| | - Tejesh Guddanti
- 1 The Vivian L. Smith Department of Neurosurgery McGovern Medical School The University of Texas Health Science Center at Houston Houston TX
| | - Yuanqing Yan
- 1 The Vivian L. Smith Department of Neurosurgery McGovern Medical School The University of Texas Health Science Center at Houston Houston TX
| | - Spiros L Blackburn
- 1 The Vivian L. Smith Department of Neurosurgery McGovern Medical School The University of Texas Health Science Center at Houston Houston TX
| | - Devin W McBride
- 1 The Vivian L. Smith Department of Neurosurgery McGovern Medical School The University of Texas Health Science Center at Houston Houston TX
| |
Collapse
|
21
|
van Lieshout JH, Marbacher S, Muhammad S, Boogaarts HD, Bartels RHMA, Dibué M, Steiger HJ, Hänggi D, Kamp MA. Proposed Definition of Experimental Secondary Ischemia for Mouse Subarachnoid Hemorrhage. Transl Stroke Res 2020; 11:1165-1170. [PMID: 32152960 PMCID: PMC7496000 DOI: 10.1007/s12975-020-00796-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/17/2020] [Accepted: 02/21/2020] [Indexed: 01/01/2023]
Abstract
Inconsistency in outcome parameters for delayed cerebral ischemia (DCI) makes it difficult to compare results between mouse studies, in the same way inconsistency in outcome parameters in human studies has for long obstructed adequate comparison. The absence of an established definition may in part be responsible for the failed translational results. The present article proposes a standardized definition for DCI in experimental mouse models, which can be used as outcome measure in future animal studies. We used a consensus-building approach to propose a definition for "experimental secondary ischemia" (ESI) in experimental mouse subarachnoid hemorrhage that can be used as an outcome measure in preclinical studies. We propose that the outcome measure should be as follows: occurrence of focal neurological impairment or a general neurological impairment compared with a control group and that neurological impairment should occur secondarily following subarachnoid hemorrhage (SAH) induction compared with an initial assessment following SAH induction. ESI should not be used if the condition can be explained by general anesthesia or if other means of assessments sufficiently explain function impairment. If neurological impairment cannot reliably be evaluated, due to scientific setup. Verification of a significant secondary impairment of the cerebral perfusion compared with a control group is mandatory. This requires longitudinal examination in the same animal. The primary aim is that ESI should be distinguished from intervention-related ischemia or neurological deficits, in order establish a uniform definition for experimental SAH in mice that is in alignment with outcome measures in human studies.
Collapse
Affiliation(s)
- Jasper Hans van Lieshout
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, D-40225, Düsseldorf, Germany. .,Department of Neurosurgery, Radboudumc Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands.
| | - Serge Marbacher
- Department of Neurosurgery c/o Neuro Research Office, Kantonsspital Aarau, Tellstrasse 1, 5001, Aarau, Switzerland
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, D-40225, Düsseldorf, Germany
| | - Hieronymus D Boogaarts
- Department of Neurosurgery, Radboudumc Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands
| | - Ronald H M A Bartels
- Department of Neurosurgery, Radboudumc Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands
| | - Maxine Dibué
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, D-40225, Düsseldorf, Germany
| | - Hans-Jakob Steiger
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, D-40225, Düsseldorf, Germany
| | - Daniel Hänggi
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, D-40225, Düsseldorf, Germany
| | - Marcel A Kamp
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, D-40225, Düsseldorf, Germany
| |
Collapse
|
22
|
Garland P, Morton MJ, Haskins W, Zolnourian A, Durnford A, Gaastra B, Toombs J, Heslegrave AJ, More J, Okemefuna AI, Teeling JL, Graversen JH, Zetterberg H, Moestrup SK, Bulters DO, Galea I. Haemoglobin causes neuronal damage in vivo which is preventable by haptoglobin. Brain Commun 2020; 2:fcz053. [PMID: 32346673 PMCID: PMC7188517 DOI: 10.1093/braincomms/fcz053] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After subarachnoid haemorrhage, prolonged exposure to toxic extracellular haemoglobin occurs in the brain. Here, we investigate the role of haemoglobin neurotoxicity in vivo and its prevention. In humans after subarachnoid haemorrhage, haemoglobin in cerebrospinal fluid was associated with neurofilament light chain, a marker of neuronal damage. Most haemoglobin was not complexed with haptoglobin, an endogenous haemoglobin scavenger present at very low concentration in the brain. Exogenously added haptoglobin bound most uncomplexed haemoglobin, in the first 2 weeks after human subarachnoid haemorrhage, indicating a wide therapeutic window. In mice, the behavioural, vascular, cellular and molecular changes seen after human subarachnoid haemorrhage were recapitulated by modelling a single aspect of subarachnoid haemorrhage: prolonged intrathecal exposure to haemoglobin. Haemoglobin-induced behavioural deficits and astrocytic, microglial and synaptic changes were attenuated by haptoglobin. Haptoglobin treatment did not attenuate large-vessel vasospasm, yet improved clinical outcome by restricting diffusion of haemoglobin into the parenchyma and reducing small-vessel vasospasm. In summary, haemoglobin toxicity is of clinical importance and preventable by haptoglobin, independent of large-vessel vasospasm.
Collapse
Affiliation(s)
- Patrick Garland
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Matthew J Morton
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - William Haskins
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Ardalan Zolnourian
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Andrew Durnford
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Ben Gaastra
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Jamie Toombs
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.,Department of Neurodegenerative Disease, Institute of Neurology, London, WC1N 3BG, UK
| | - Amanda J Heslegrave
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.,Department of Neurodegenerative Disease, Institute of Neurology, London, WC1N 3BG, UK
| | - John More
- Research & Development Department, Bio Products Laboratory Limited, Elstree, Hertfordshire, WD6 3BX, UK
| | - Azubuike I Okemefuna
- Research & Development Department, Bio Products Laboratory Limited, Elstree, Hertfordshire, WD6 3BX, UK
| | - Jessica L Teeling
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, SO16 6YD, UK
| | - Jonas H Graversen
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.,Department of Neurodegenerative Disease, Institute of Neurology, London, WC1N 3BG, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mo¨ lndal, S-431 80, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mo¨ lndal, S-431 80, Sweden
| | - Soren K Moestrup
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark.,Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus N, Denmark.,Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Diederik O Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK.,Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
| |
Collapse
|
23
|
Immune response mediates the cardiac damage after subarachnoid hemorrhage. Exp Neurol 2019; 323:113093. [PMID: 31676318 DOI: 10.1016/j.expneurol.2019.113093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/09/2019] [Accepted: 10/25/2019] [Indexed: 11/23/2022]
Abstract
Cardiac dysfunction is a common adverse effect of subarachnoid hemorrhage (SAH). Autopsy of SAH patients shows immunocyte infiltration into the heart. In this study, a SAH model of endovascular perforation was performed in adult male mice in order to test whether SAH causes cardiac dysfunction in non-primary cardiac disease young adult male mice and whether immune response mediates SAH induced cardiac and neurological deficit. Splenectomy was performed on a subpopulation of mice one week prior to induction of the SAH. Neurological functional tests, transthoracic Doppler echocardiography, immunofluorescent staining, and flow cytometry were performed to investigate neurological and cardiac function and immune/inflammatory effects of SAH in mice with or without splenectomy. We found that SAH significantly induces ventricular fibrillation and cardiac dysfunction identified by significantly reduced left ventricular ejection fraction, left ventricular fractional shortening, decreased heart rate, as well as increased macrophage and neutrophil infiltration into heart and inflammatory factor expression in the heart compared to sham control mice. SAH also induces neurological deficit, increases astrocyte and microglial activity, and inflammatory cell infiltration into brain as well as up-regulates inflammatory factor expression in the brain tissue. Splenectomy not only significantly improves neurological function, but also reduces cardiac dysfunction compared to SAH alone mice. Splenectomy in SAH mice significantly reduces inflammatory cell infiltration, and decreases NADPH oxidase-2 and macrophage chemokine protein-1 expression in heart and brain when compared to non-splenectomy SAH mice. Our data suggest that, SAH induces acute cardiac dysfunction in non-primary cardiac disease mice. Secondary immune response may play an important role in mediating brain-heart damage after SAH.
Collapse
|
24
|
Pu T, Zou W, Feng W, Zhang Y, Wang L, Wang H, Xiao M. Persistent Malfunction of Glymphatic and Meningeal Lymphatic Drainage in a Mouse Model of Subarachnoid Hemorrhage. Exp Neurobiol 2019; 28:104-118. [PMID: 30853828 PMCID: PMC6401547 DOI: 10.5607/en.2019.28.1.104] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/19/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular event that often is followed by permanent brain impairments. It is necessary to explore the pathogenesis of secondary pathological damages in order to find effective interventions for improving the prognosis of SAH. Blockage of brain lymphatic drainage has been shown to worsen cerebral ischemia and edema after acute SAH. However, whether or not there is persistent dysfunction of cerebral lymphatic drainage following SAH remains unclear. In this study, autologous blood was injected into the cisterna magna of mice to establish SAH model. One week after surgery, SAH mice showed decreases in fluorescent tracer drainage to the deep cervical lymph nodes (dcLNs) and influx into the brain parenchyma after injection into the cisterna magna. Moreover, SAH impaired polarization of astrocyte aquaporin-4 (AQP4) that is a functional marker of glymphatic clearance and resulted in accumulations of Tau proteins as well as CD3+, CD4+, and CD8+ cells in the brain. In addition, pathological changes, including microvascular spasm, activation of glial cells, neuroinflammation, and neuronal apoptosis were observed in the hippocampus of SAH mice. Present results demonstrate persistent malfunction of glymphatic and meningeal lymphatic drainage and related neuropathological damages after SAH. Targeting improvement of brain lymphatic clearance potentially serves as a new strategy for the treatment of SAH.
Collapse
Affiliation(s)
- Tinglin Pu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Wenyan Zou
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Yanli Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Linmei Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Hongxing Wang
- Deptment of Rehabilitation Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
25
|
RP001 hydrochloride improves neurological outcome after subarachnoid hemorrhage. J Neurol Sci 2019; 399:6-14. [PMID: 30738334 DOI: 10.1016/j.jns.2019.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 02/08/2023]
Abstract
Subarachnoid hemorrhage (SAH) results in neurological damage, acute cardiac damage and has a high mortality rate. Immunoresponse in the acute phase after SAH plays a key role in mediating vasospasm, edema, inflammation and neuronal damage. The S1P/S1PR pathway impacts multiple cellular functions, exerts anti-inflammatory and anti-apoptotic effects, promotes remyelination, and improves outcome in several central nervous system (CNS) diseases. RP001 hydrochloride is a novel S1PR agonist, which sequesters lymphocytes within their secondary tissues and prevents infiltration of immune cells into the CNS thereby reducing immune response. In this study, we investigated whether RP001 attenuates neuronal injury after SAH by reducing inflammation. S1PRs, specifically S1PR1, 3 not only exerts anti-inflammatory effects, but also decreases heart rate and induces atrioventricular conduction abnormalities. Therefore, we also tested whether RP001 treatment of SAH regulates cardiac functional outcome. Male adult C57BL/6 mice were subjected to SAH, and neurological function tests, echocardiography, and immunohistochemical analysis were performed. SAH induces neurological deficits and acute cardiac dysfunction compared to sham control mice. Treatment of SAH with a low-dose of RP001 induces better neurological outcome and cardiac function compared to a high-dose of RP001. Low-dose-RP001 treatment significantly decreases apoptosis, white matter damage, blood brain barrier permeability, microglial/astrocyte activation, macrophage chemokine protein-1, matrix metalloproteinase-9 and NADPH oxidase-2 expression in the brain compared to SAH control mice. Our findings indicate that low-dose of RP001 alleviates neurological damage after SAH, in part by decreasing neuroinflammation.
Collapse
|
26
|
Pu T, Zou W, Feng W, Zhang Y, Wang L, Wang H, Xiao M. Persistent Malfunction of Glymphatic and Meningeal Lymphatic Drainage in a Mouse Model of Subarachnoid Hemorrhage. Exp Neurobiol 2019; 28:104-118. [PMID: 30853828 PMCID: PMC6401547 DOI: 10.5607/en.2019.28.1.104;17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 06/09/2024] Open
Abstract
Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular event that often is followed by permanent brain impairments. It is necessary to explore the pathogenesis of secondary pathological damages in order to find effective interventions for improving the prognosis of SAH. Blockage of brain lymphatic drainage has been shown to worsen cerebral ischemia and edema after acute SAH. However, whether or not there is persistent dysfunction of cerebral lymphatic drainage following SAH remains unclear. In this study, autologous blood was injected into the cisterna magna of mice to establish SAH model. One week after surgery, SAH mice showed decreases in fluorescent tracer drainage to the deep cervical lymph nodes (dcLNs) and influx into the brain parenchyma after injection into the cisterna magna. Moreover, SAH impaired polarization of astrocyte aquaporin-4 (AQP4) that is a functional marker of glymphatic clearance and resulted in accumulations of Tau proteins as well as CD3+, CD4+, and CD8+ cells in the brain. In addition, pathological changes, including microvascular spasm, activation of glial cells, neuroinflammation, and neuronal apoptosis were observed in the hippocampus of SAH mice. Present results demonstrate persistent malfunction of glymphatic and meningeal lymphatic drainage and related neuropathological damages after SAH. Targeting improvement of brain lymphatic clearance potentially serves as a new strategy for the treatment of SAH.
Collapse
Affiliation(s)
- Tinglin Pu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Wenyan Zou
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Yanli Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Linmei Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| | - Hongxing Wang
- Deptment of Rehabilitation Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
27
|
Barrow JW, Turan N, Wangmo P, Roy AK, Pradilla G. The role of inflammation and potential use of sex steroids in intracranial aneurysms and subarachnoid hemorrhage. Surg Neurol Int 2018; 9:150. [PMID: 30105144 PMCID: PMC6080146 DOI: 10.4103/sni.sni_88_18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
Background Aneurysmal subarachnoid hemorrhage (aSAH) continues to be a devastating neurological condition with a high risk of associated morbidity and mortality. Inflammation has been shown to increase the risk of complications associated with aSAH such as vasospasm and brain injury in animal models and humans. The goal of this review is to discuss the inflammatory mechanisms of aneurysm formation, rupture and vasospasm and explore the role of sex hormones in the inflammatory response to aSAH. Methods A literature review was performed using PubMed using the following search terms: "intracranial aneurysm," "cerebral aneurysm," "dihydroepiandrosterone sulfate" "estrogen," "hormone replacement therapy," "inflammation," "oral contraceptive," "progesterone," "sex steroids," "sex hormones" "subarachnoid hemorrhage," "testosterone." Only studies published in English language were included in the review. Results Studies have shown that administration of sex hormones such as progesterone and estrogen at early stages in the inflammatory cascade can lower the risk and magnitude of subsequent complications. The exact mechanism by which these hormones act on the brain, as well as their role in the inflammatory cascade is not fully understood. Moreover, conflicting results have been published on the effect of hormone replacement therapy in humans. This review will scrutinize the variations in these studies to provide a more detailed understanding of sex hormones as potential therapeutic agents for intracranial aneurysms and aSAH. Conclusion Inflammation may play a role in the pathogenesis of intracranial aneurysm formation and subarachnoid hemorrhage, and administration of sex hormones as anti-inflammatory agents has been associated with improved functional outcome in experimental models. Further studies are needed to determine the therapeutic role of these hormones in the intracranial aneurysms and aSAH.
Collapse
Affiliation(s)
- Jack W Barrow
- Cerebrovascular Research Laboratory, Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Mercer University School of Medicine, Savannah, Georgia, USA
| | - Nefize Turan
- Cerebrovascular Research Laboratory, Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Pasang Wangmo
- Cerebrovascular Research Laboratory, Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anil K Roy
- Cerebrovascular Research Laboratory, Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Gustavo Pradilla
- Cerebrovascular Research Laboratory, Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Duris K, Lipkova J, Splichal Z, Madaraszova T, Jurajda M. Early Inflammatory Response in the Brain and Anesthesia Recovery Time Evaluation After Experimental Subarachnoid Hemorrhage. Transl Stroke Res 2018; 10:10.1007/s12975-018-0641-z. [PMID: 29926382 DOI: 10.1007/s12975-018-0641-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/09/2018] [Accepted: 06/12/2018] [Indexed: 01/05/2023]
Abstract
The main objective was to evaluate, whether the subarachnoid hemorrhage (SAH)-associated early inflammatory response has focal or global character, i.e., whether areas distant to hematoma may be affected by an early inflammatory response. The second objective was to evaluate the association of anesthesia recovery time for basic reflexes/neurological functions with severity of SAH. SAH was induced in rats using an endovascular perforation model. Anesthesia recovery time was evaluated for pain reaction recovery time (spinal level), spontaneous ventilation recovery time (brain stem level), and consciousness recovery time (neocortical level). mRNA expressions of TNFα, IL-1β, IL-6, ICAM-1, and VCAM-1 in areas adjacent and distant to hematoma were evaluated between 2 and 8 h after SAH. Serum levels of TNFα, IL-1β, and IL-6 were assessed at 4 and 8 h after SAH. Anesthesia recovery time of all selected parameters was associated with severity of SAH. The consciousness recovery time test had the best predictive value, while the spontaneous ventilation recovery time test was able to bring information in the shortest time. The mRNA expressions of pro-inflammatory cytokines were significantly increased in severe SAH groups in both adjacent and distant areas. The inflammatory response in mild/moderate SAH groups was less strong, peaking at 4 h after SAH. Serum levels of pro-inflammatory cytokines were ambiguous. Anesthesia recovery time may be useful for bleeding severity prediction in the SAH model; however, further validation is needed. Severe subarachnoid hemorrhage is associated with the strong early inflammatory response, which has a global character, while mild subarachnoid hemorrhage is accompanied by a weaker inflammation.
Collapse
Affiliation(s)
- K Duris
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Neurosurgery, The University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - J Lipkova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Z Splichal
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - T Madaraszova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Neurosurgery, The University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michal Jurajda
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| |
Collapse
|
29
|
Effect of Progesterone on Cerebral Vasospasm and Neurobehavioral Outcomes in a Rodent Model of Subarachnoid Hemorrhage. World Neurosurg 2017; 110:e150-e159. [PMID: 29097330 DOI: 10.1016/j.wneu.2017.10.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/19/2017] [Accepted: 10/21/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) induces widespread inflammation leading to cellular injury, vasospasm, and ischemia. Evidence suggests that progesterone (PROG) can improve functional recovery in acute brain injury owing to its anti-inflammatory and neuroprotective properties, which could also be beneficial in SAH. We hypothesized that PROG treatment attenuates inflammation-mediated cerebral vasospasm and microglial activation, improves synaptic connectivity, and ameliorates functional recovery after SAH. METHODS We investigated the effect of PROG in a cisternal SAH model in adult male C57BL/6 mice. Neurobehavioral outcomes were evaluated using rotarod latency and grip strength tests. Basilar artery perimeter, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptor 1 (GluR1)/synaptophysin colocalization, and Iba-1 immunoreactivity were quantified histologically. RESULTS PROG (8 mg/kg) significantly improved rotarod latency at day 6 and grip strength at day 9. PROG-treated mice had significantly reduced basilar artery vasospasm at 24 hours. GluR1/synaptophysin colocalization, indicative of synaptic GluR1, was significantly reduced in the SAH+Vehicle group at 24 hours, and PROG treatment significantly attenuated this reduction. PROG treatment significantly reduced microglial cell activation and proliferation in cerebellum and cortex but not in the brainstem at 10 days. CONCLUSIONS PROG treatment ameliorated cerebral vasospasm, reduced microglial activation, restored synaptic GluR1 localization, and improved neurobehavioral performance in a murine model of SAH. These results provide a rationale for further translational testing of PROG therapy in SAH.
Collapse
|
30
|
Vellimana AK, Zhou ML, Singh I, Aum DJ, Nelson JW, Harris GR, Athiraman U, Han BH, Zipfel GJ. Minocycline protects against delayed cerebral ischemia after subarachnoid hemorrhage via matrix metalloproteinase-9 inhibition. Ann Clin Transl Neurol 2017; 4:865-876. [PMID: 29296615 PMCID: PMC5740245 DOI: 10.1002/acn3.492] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/06/2017] [Accepted: 09/25/2017] [Indexed: 12/11/2022] Open
Abstract
Objective Delayed cerebral ischemia (DCI) is an independent risk factor for poor outcome after aneurysmal subarachnoid hemorrhage (SAH) and is multifactorial in etiology. While prior studies have suggested a role for matrix metalloproteinase-9 (MMP-9) in early brain injury after SAH, its contribution to the pathophysiology of DCI is unclear. Methods In the first experiment, wild-type (WT) and MMP-9-/- mice were subjected to sham or endovascular perforation SAH surgery. In separate experiments, WT and MMP-9-/-mice were administered vehicle or minocycline either pre- or post-SAH. All mice underwent assessment of multiple components of DCI including vasospasm, neurobehavioral function, and microvessel thrombosis. In another experiment, rabbits were subjected to sham or cisterna magna injection SAH surgery, and administered vehicle or minocycline followed by vasospasm assessment. Results MMP-9 expression and activity was increased after SAH. Genetic (MMP-9-/- mice) and pharmacological (pre-SAH minocycline administration) inhibition of MMP-9 resulted in decreased vasospasm and neurobehavioral deficits. A therapeutically feasible strategy of post-SAH administration of minocycline resulted in attenuation of multiple components of DCI. Minocycline administration to MMP-9-/- mice did not yield additional protection. Consistent with experiments in mice, both pre- and post-SAH administration of minocycline attenuated SAH-induced vasospasm in rabbits. Interpretation MMP-9 is a key player in the pathogenesis of DCI. The consistent attenuation of multiple components of DCI with both pre- and post-SAH administration of minocycline across different species and experimental models of SAH, combined with the excellent safety profile of minocycline in humans suggest that a clinical trial in SAH patients is warranted.
Collapse
Affiliation(s)
- Ananth K Vellimana
- Department of Neurological Surgery Washington University School of Medicine St. Louis Missouri
| | - Meng-Liang Zhou
- Department of Neurosurgery Jinling Hospital School of Medicine Nanjing University Nanjing Jiangsu Province China
| | - Itender Singh
- Department of Neurological Surgery Washington University School of Medicine St. Louis Missouri
| | - Diane J Aum
- Department of Neurological Surgery Washington University School of Medicine St. Louis Missouri
| | - James W Nelson
- Department of Neurological Surgery Washington University School of Medicine St. Louis Missouri
| | - Glenn R Harris
- Department of Neurological Surgery Washington University School of Medicine St. Louis Missouri
| | - Umeshkumar Athiraman
- Department of Anesthesiology Washington University School of Medicine St. Louis Missouri
| | - Byung H Han
- Department of Pharmacology A.T. Still University of Health Sciences Kirksville College of Osteopathic Medicine Kirksville Missouri
| | - Gregory J Zipfel
- Department of Neurological Surgery Washington University School of Medicine St. Louis Missouri
| |
Collapse
|
31
|
Fanizzi C, Sauerbeck AD, Gangolli M, Zipfel GJ, Brody DL, Kummer TT. Minimal Long-Term Neurobehavioral Impairments after Endovascular Perforation Subarachnoid Hemorrhage in Mice. Sci Rep 2017; 7:7569. [PMID: 28790425 PMCID: PMC5548778 DOI: 10.1038/s41598-017-07701-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
Cognitive deficits are among the most severe and pervasive consequences of aneurysmal subarachnoid hemorrhage (SAH). A critical step in developing therapies targeting such outcomes is the characterization of experimentally-tractable pre-clinical models that exhibit multi-domain neurobehavioral deficits similar to those afflicting humans. We therefore searched for neurobehavioral abnormalities following endovascular perforation induction of SAH in mice, a heavily-utilized model. We instituted a functional screen to manage variability in injury severity, then assessed acute functional deficits, as well as activity, anxiety-related behavior, learning and memory, socialization, and depressive-like behavior at sub-acute and chronic time points (up to 1 month post-injury). Animals in which SAH was induced exhibited reduced acute functional capacity and reduced general activity to 1 month post-injury. Tests of anxiety-related behavior including central area time in the elevated plus maze and thigmotaxis in the open field test revealed increased anxiety-like behavior at subacute and chronic time-points, respectively. Effect sizes for subacute and chronic neurobehavioral endpoints in other domains, however, were small. In combination with persistent variability, this led to non-significant effects of injury on all remaining neurobehavioral outcomes. These results suggest that, with the exception of anxiety-related behavior, alternate mouse models are required to effectively analyze cognitive outcomes after SAH.
Collapse
Affiliation(s)
- Claudia Fanizzi
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
- Department of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrew D Sauerbeck
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
| | - Mihika Gangolli
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
| | - Gregory J Zipfel
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, Missouri, USA
| | - David L Brody
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA
| | - Terrance T Kummer
- Department of Neurology, Washington University School of Medicine in St. Louis, Missouri, USA.
| |
Collapse
|
32
|
Chou SHY, Lan J, Esposito E, Ning M, Balaj L, Ji X, Lo EH, Hayakawa K. Extracellular Mitochondria in Cerebrospinal Fluid and Neurological Recovery After Subarachnoid Hemorrhage. Stroke 2017; 48:2231-2237. [PMID: 28663512 DOI: 10.1161/strokeaha.117.017758] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE Recent studies suggest that extracellular mitochondria may be involved in the pathophysiology of stroke. In this study, we assessed the functional relevance of endogenous extracellular mitochondria in cerebrospinal fluid (CSF) in rats and humans after subarachnoid hemorrhage (SAH). METHODS A standard rat model of SAH was used, where an intraluminal suture was used to perforate a cerebral artery, thus leading to blood extravasation into subarachnoid space. At 24 and 72 hours after SAH, neurological outcomes were measured, and the standard JC1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzimidazolylcarbocyanineiodide) assay was used to quantify mitochondrial membrane potentials in the CSF. To further support the rat model experiments, CSF samples were obtained from 41 patients with SAH and 27 control subjects. Mitochondrial membrane potentials were measured with the JC1 assay, and correlations with clinical outcomes were assessed at 3 months. RESULTS In the standard rat model of SAH, extracellular mitochondria was detected in CSF at 24 and 72 hours after injury. JC1 assays demonstrated that mitochondrial membrane potentials in CSF were decreased after SAH compared with sham-operated controls. In human CSF samples, extracellular mitochondria were also detected, and JC1 levels were also reduced after SAH. Furthermore, higher mitochondrial membrane potentials in the CSF were correlated with good clinical recovery at 3 months after SAH onset. CONCLUSIONS This proof-of-concept study suggests that extracellular mitochondria may provide a biomarker-like glimpse into brain integrity and recovery after injury.
Collapse
Affiliation(s)
- Sherry H-Y Chou
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Jing Lan
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Elga Esposito
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - MingMing Ning
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Leonora Balaj
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Xunming Ji
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Eng H Lo
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Kazuhide Hayakawa
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.).
| |
Collapse
|
33
|
Long-Lasting Cerebral Vasospasm, Microthrombosis, Apoptosis and Paravascular Alterations Associated with Neurological Deficits in a Mouse Model of Subarachnoid Hemorrhage. Mol Neurobiol 2017; 55:2763-2779. [PMID: 28455691 DOI: 10.1007/s12035-017-0514-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/04/2017] [Indexed: 12/21/2022]
Abstract
Subarachnoid hemorrhage (SAH) is a devastating disease with high mortality and morbidity. Long-term cognitive and sensorimotor deficits are serious complications following SAH but still not well explained and described in mouse preclinical models. The aim of our study is to characterize a well-mastered SAH murine model and to establish developing pathological mechanisms leading to cognitive and motor deficits, allowing identification of specific targets involved in these long-term troubles. We hereby demonstrate that the double blood injection model of SAH induced long-lasting large cerebral artery vasospasm (CVS), microthrombosis formation and cerebral brain damage including defect in potential paravascular diffusion. These neurobiological alterations appear to be associated with sensorimotor and cognitive dysfunctions mainly detected 10 days after the bleeding episode. In conclusion, this characterized model of SAH in mice, stressing prolonged neurobiological pathological mechanisms and associated sensitivomotor deficits, will constitute a validated preclinical model to better decipher the link between CVS, long-term cerebral apoptosis and cognitive disorders occurring during SAH and to allow investigating novel therapeutic approaches in transgenic mice.
Collapse
|
34
|
Overview of Traumatic Brain Injury: An Immunological Context. Brain Sci 2017; 7:brainsci7010011. [PMID: 28124982 PMCID: PMC5297300 DOI: 10.3390/brainsci7010011] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) afflicts people of all ages and genders, and the severity of injury ranges from concussion/mild TBI to severe TBI. Across all spectrums, TBI has wide-ranging, and variable symptomology and outcomes. Treatment options are lacking for the early neuropathology associated with TBIs and for the chronic neuropathological and neurobehavioral deficits. Inflammation and neuroinflammation appear to be major mediators of TBI outcomes. These systems are being intensively studies using animal models and human translational studies, in the hopes of understanding the mechanisms of TBI, and developing therapeutic strategies to improve the outcomes of the millions of people impacted by TBIs each year. This manuscript provides an overview of the epidemiology and outcomes of TBI, and presents data obtained from animal and human studies focusing on an inflammatory and immunological context. Such a context is timely, as recent studies blur the traditional understanding of an “immune-privileged” central nervous system. In presenting the evidence for specific, adaptive immune response after TBI, it is hoped that future studies will be interpreted using a broader perspective that includes the contributions of the peripheral immune system, to central nervous system disorders, notably TBI and post-traumatic syndromes.
Collapse
|