1
|
Kim S, Jang S, Lee O. Muscle structure assessment using synchrotron radiation X-ray micro-computed tomography in murine with cerebral ischemia. Sci Rep 2024; 14:26825. [PMID: 39501018 PMCID: PMC11538359 DOI: 10.1038/s41598-024-78324-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024] Open
Abstract
Muscles are crucial for balance and walking, activities which depend specifically on the lower extremity muscles. Therefore, the evaluation of stroke induced atrophy and paralysis is essential; however, determining the extent of damage in the days after its occurrence remains challenging. In this study, we evaluated ischemic stroke-induced soleus muscle damage in gerbils using synchrotron radiation X-ray micro-computed tomography (SR-µCT), comparing a control group (n = 3), animals 7 days after stroke (7 d, n = 3), and animals 14 days after stroke (14 d, n = 3). The left muscle was paralyzed, whereas the right muscle was not. Subsequently, we quantified the assessment by segmenting the soleus muscle based on the extracellular space/matrix and fiber region to determine the degree of damage. The muscle fiber-to-extracellular space/matrix ratio were significantly damaged due to paralysis on the left side (control vs. 14 d, P = 0.040). Muscle area was significantly different at 14 d between the left and right sides (P = 0.010). Additionally, the left local fascicle surface area, thickness, global pennation angle, and local fascicle angle were significantly different between the control and 14 d groups (P = 0.002, P = 0.007, P = 0.005, and P = 0.014 respectively). These findings underscore the potential of post-stroke animal studies in improving rehabilitation treatment for the central nervous system by assessing the degree of muscle recovery.
Collapse
Affiliation(s)
- Subok Kim
- Department of Software Convergence, Graduate School, Soonchunhyang University, 22, Soonchunhyang-ro, 31538, Asan City, Chungnam, Republic of Korea
| | - Sanghun Jang
- Department of Physical Therapy, College of Health and Life Sciences, Korea National University of Transportation, 61, Daehak-ro, 27909, Jeungpyeong-eup, Chungbuk, Republic of Korea
| | - Onseok Lee
- Department of Software Convergence, Graduate School, Soonchunhyang University, 22, Soonchunhyang-ro, 31538, Asan City, Chungnam, Republic of Korea.
- Department of Medical IT Engineering, College of Medical Sciences, Soonchunhyang University, 22, Soonchunhyang-ro, 31538, Asan City, Chungnam, Republic of Korea.
| |
Collapse
|
2
|
Rust R, Nih LR, Liberale L, Yin H, El Amki M, Ong LK, Zlokovic BV. Brain repair mechanisms after cell therapy for stroke. Brain 2024; 147:3286-3305. [PMID: 38916992 PMCID: PMC11449145 DOI: 10.1093/brain/awae204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischaemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischaemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Lina R Nih
- Department of Brain Health, University of Nevada, Las Vegas, NV 89154, USA
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Lin Kooi Ong
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Zhang J, Price CJ, Zhao K, Tang Y, Zhong S, Lou J, Ye X, Liang F. Associations between amyloid-β load and cognition in cerebrovascular disease beyond cerebral amyloid angiopathy: a systematic review and meta-analysis of positron emission tomography studies. Age Ageing 2024; 53:afae240. [PMID: 39468728 DOI: 10.1093/ageing/afae240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND There is growing interest in the comorbidity of vascular and neurodegenerative pathologies in patients with cerebrovascular disease (CVD) beyond cerebral amyloid angiopathy (CAA). However, the relationship between amyloid-β and vascular cognitive impairment (VCI) remains debated. OBJECTIVE To investigate the association between VCI and amyloid-β deposition in non-CAA CVD patients. METHODS PubMed, Embase, Web of Science, PsycINFO and CENTRAL databases were systematically searched. Observational studies, including case-control and cohort studies, associating cognitive scores with amyloid load measured by positron emission tomography were selected. Meta-analyses were performed to assess the strength of amyloid-cognition associations across CVD subtypes and cognitive domains. A random-effects model using the inverse variance method was used, with heterogeneity evaluated by Q-statistics and I2 statistics. Meta-regression analyses were conducted to examine the influence of moderators, and publication bias was assessed using funnel plots and Egger's test. All statistical analyses were performed using StataMP 18. RESULTS Twenty-seven eligible studies encompassing 2894 participants were included. Among non-CAA CVD patients, global cognitive performance was significantly lower in those with higher amyloid-β deposition (standardized mean difference = -0.43, P < 0.001). The correlation strength varied across cognitive domains (executive function: r = -0.41; language: r = -0.36; memory: r = -0.29; all P < 0.001). The correlation was significant in patients with subcortical vascular disease (r = -0.43, P < 0.001) but not post-stroke patients (r = -0.19, P > 0.05). CONCLUSIONS Amyloid-β load is associated with cognitive decline in non-CAA CVD patients. This is more pronounced in patients with subcortical vascular disease than in post-stroke patients. Executive function is the most susceptible domain in VCI when the level of amyloid-β increases.
Collapse
Affiliation(s)
- Jie Zhang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, China
- Wellcome Center for Human Neuroimaging, Department of Imaging Neuroscience, Institute of Neurology, University College London, 12 Queen Square, London WC1N 3AR, United Kingdom
| | - Cathy J Price
- Wellcome Center for Human Neuroimaging, Department of Imaging Neuroscience, Institute of Neurology, University College London, 12 Queen Square, London WC1N 3AR, United Kingdom
| | - Ke Zhao
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Beijing 100101, China
- Institute of Cognitive Neuroscience, University College London, Alexandra House, 17 Queen Square, London WC1N 3AZ, United Kingdom
| | - Yuanyuan Tang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, China
| | - Shuchang Zhong
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, China
| | - Jingjing Lou
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, China
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, China
| | - Feng Liang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Road, Hangzhou 310014, China
| |
Collapse
|
4
|
Uppu RM, Chikkula K, Saneei S, Babu S, Fronczek FR. Structure of 2,3,5-tri-phenyl-tetra-zol-3-ium chloride hemi-penta-hydrate. Acta Crystallogr E Crystallogr Commun 2024; 80:1097-1100. [PMID: 39372184 PMCID: PMC11451482 DOI: 10.1107/s205698902400940x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
The title hydrated mol-ecular salt, C19H15N4 +·Cl-·2.5H2O, has two tri-phenyl-tetra-zolium cations, two chloride anions and five water mol-ecules in the asymmetric unit. The cations differ in the conformations of the phenyl rings with respect to the heterocyclic core, most notably for the C-bonded phenyl ring, for which the N-C-C-C torsion angles differ by 36.4 (3)°. This is likely a result of one cation accepting an O-H⋯N hydrogen bond from a water mol-ecule [O⋯N = 3.1605 (15) Å], while the other cation accepts no hydrogen bonds. In the extended structure, the water mol-ecules are involved in centrosymmetric (H2O)2Cl2 rings as well as (H2O)4 chains. An unusual O-H⋯π inter-action and weak C-H⋯O and C-H⋯Cl hydrogen bonds are also observed.
Collapse
Affiliation(s)
- Rao M Uppu
- Department of Environmental Toxicology Southern University and A&M College Baton Rouge LA 70813 USA
| | - Krishnaveni Chikkula
- Department of Environmental Toxicology Southern University and A&M College Baton Rouge LA 70813 USA
| | - Soheil Saneei
- Department of Environmental Toxicology Southern University and A&M College Baton Rouge LA 70813 USA
| | - Sainath Babu
- Department of Environmental Toxicology Southern University and A&M College Baton Rouge LA 70813 USA
| | - Frank R. Fronczek
- Department of Chemistry Louisiana State University,Baton Rouge LA 70803 USA
| |
Collapse
|
5
|
Chen R, Zhang MQ, Miao YL, Zhang SH, Cheng Y, Wang SS, Yin J, Yang CH, Zhang HF, Tang L, Li Y, Zhang Y, Fan YY. Targeting Neuronal GPR65 With Delayed BTB09089 Treatment Improves Neurorehabilitation Following Ischemic Stroke. Stroke 2024; 55:2151-2162. [PMID: 38946544 PMCID: PMC11259240 DOI: 10.1161/strokeaha.124.046954] [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: 01/26/2024] [Revised: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND GPR65 (G protein-coupled receptor 65) can sense extracellular acidic environment to regulate pathophysiological processes. Pretreatment with the GPR65 agonist BTB09089 has been proven to produce neuroprotection in acute ischemic stroke. However, whether delayed BTB09089 treatment and neuronal GPR65 activation promote neurorestoration remains unknown. METHODS Ischemic stroke was induced in wild-type (WT) or GPR65 knockout (GPR65-/-) mice by photothrombotic ischemia. Male mice were injected intraperitoneally with BTB09089 every other day at days 3, 7, or 14 poststroke. AAV-Syn-GPR65 (adenoassociated virus-synapsin-GPR65) was utilized to overexpress GPR65 in the peri-infarct cortical neurons of GPR65-/- and WT mice. Motor function was monitored by grid-walk and cylinder tests. The neurorestorative effects of BTB09089 were observed by immunohistochemistry, Golgi-Cox staining, and Western blotting. RESULTS BTB09089 significantly promoted motor outcomes in WT but not in GPR65-/- mice, even when BTB09089 was delayed for 3 to 7 days. BTB09089 inhibited the activation of microglia and glial scar progression in WT but not in GPR65-/- mice. Meanwhile, BTB09089 reduced the decrease in neuronal density in WT mice, but this benefit was abolished in GPR65-/- mice and reemerged by overexpressing GPR65 in peri-infarct cortical neurons. Furthermore, BTB09089 increased the GAP43 (growth-associated protein-43) and synaptophysin puncta density, dendritic spine density, dendritic branch length, and dendritic complexity by overexpressing GPR65 in the peri-infarct cortical neurons of GPR65-/- mice, which was accompanied by increased levels of p-CREB (phosphorylated cAMP-responsive element-binding protein). In addition, the therapeutic window of BTB09089 was extended to day 14 by overexpressing GPR65 in the peri-infarct cortical neurons of WT mice. CONCLUSIONS Our findings indicated that delayed BTB09089 treatment improved neurological functional recovery and brain tissue repair poststroke through activating neuronal GRP65. GPR65 overexpression may be a potential strategy to expand the therapeutic time window of GPR65 agonists for neurorehabilitation after ischemic stroke.
Collapse
Affiliation(s)
| | | | | | - Shu-Han Zhang
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Yao Cheng
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Shao-Shuai Wang
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Jing Yin
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Cai-Hong Yang
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Hui-Feng Zhang
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Li Tang
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Yan Li
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Yu Zhang
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| | - Yan-Ying Fan
- Department of Pharmacology, School of Basic Medical Science (R.C., M.-Q.Z., Y.-L.M., S.-H.Z., Y.C., J.Y., C.-H.Y., H.-H.Z., Y.L., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Medicinal Basic Research Innovation Centre of Chronic Kidney Disease, Ministry of Education (Y.C., L.T., Y.-Y.F.), Shanxi Medical University, Jinzhong, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China (Y.-L.M., C.-H.Y., H.-F.Z., Y.L., Y.Z.)
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China (S.-S.W.)
| |
Collapse
|
6
|
Hood RJ, Sanchez-Bezanilla S, Beard DJ, Rust R, Turner RJ, Stuckey SM, Collins-Praino LE, Walker FR, Nilsson M, Ong LK. Leakage beyond the primary lesion: A temporal analysis of cerebrovascular dysregulation at sites of hippocampal secondary neurodegeneration following cortical photothrombotic stroke. J Neurochem 2023; 167:733-752. [PMID: 38010732 DOI: 10.1111/jnc.16008] [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: 06/22/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023]
Abstract
We have previously demonstrated that a cortical stroke causes persistent impairment of hippocampal-dependent cognitive tasks concomitant with secondary neurodegenerative processes such as amyloid-β accumulation in the hippocampus, a region remote from the primary infarct. Interestingly, there is emerging evidence suggesting that deposition of amyloid-β around cerebral vessels may lead to cerebrovascular structural changes, neurovascular dysfunction, and disruption of blood-brain barrier integrity. However, there is limited knowledge about the temporal changes of hippocampal cerebrovasculature after cortical stroke. In the current study, we aimed to characterise the spatiotemporal cerebrovascular changes after cortical stroke. This was done using the photothrombotic stroke model targeting the motor and somatosensory cortices of mice. Cerebrovascular morphology as well as the co-localisation of amyloid-β with vasculature and blood-brain barrier integrity were assessed in the cortex and hippocampal regions at 7, 28 and 84 days post-stroke. Our findings showed transient cerebrovascular remodelling in the peri-infarct area up to 28 days post-stroke. Importantly, the cerebrovascular changes were extended beyond the peri-infarct region to the ipsilateral hippocampus and were sustained out to 84 days post-stroke. When investigating vessel diameter, we showed a decrease at 84 days in the peri-infarct and CA1 regions that were exacerbated in vessels with amyloid-β deposition. Lastly, we showed sustained vascular leakage in the peri-infarct and ipsilateral hippocampus, indicative of a compromised blood-brain-barrier. Our findings indicate that hippocampal vasculature may represent an important therapeutic target to mitigate the progression of post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Rebecca J Hood
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Sonia Sanchez-Bezanilla
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Daniel J Beard
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Ruslan Rust
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Switzerland
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Renée J Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Shannon M Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lyndsey E Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Centre for Rehab Innovations, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Michael Nilsson
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Centre for Rehab Innovations, The University of Newcastle, Callaghan, New South Wales, Australia
- School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, Queensland, Australia
| |
Collapse
|
7
|
Li Y, Fan Q, Li F, Pang R, Chen C, Li P, Wang X, Xuan W, Yu W. The multifaceted roles of activating transcription factor 3 (ATF3) in inflammatory responses - Potential target to regulate neuroinflammation in acute brain injury. J Cereb Blood Flow Metab 2023; 43:8-17. [PMID: 37165649 PMCID: PMC10638996 DOI: 10.1177/0271678x231171999] [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: 11/01/2022] [Revised: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 05/12/2023]
Abstract
Activating transcription factor 3 (ATF3) is one of the most important transcription factors that respond to and exert dual effects on inflammatory responses. Recently, the involvement of ATF3 in the neuroinflammatory response to acute brain injury (ABI) has been highlighted. It functions by regulating neuroimmune activation and the production of neuroinflammatory mediators. Notably, recent clinical evidence suggests that ATF3 may serve as a potential ideal biomarker of the long-term prognosis of ABI patients. This mini-review describes the essential inflammation modulatory roles of ATF3 in different disease contexts and summarizes the regulatory mechanisms of ATF3 in the ABI-induced neuroinflammation.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyue Fan
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengshi Li
- Department of Neurosurgery, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Pang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Wang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Xuan
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Collyer E, Blanco-Suarez E. Astrocytes in stroke-induced neurodegeneration: a timeline. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1240862. [PMID: 39086680 PMCID: PMC11285566 DOI: 10.3389/fmmed.2023.1240862] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/22/2023] [Indexed: 08/02/2024]
Abstract
Stroke is a condition characterized by sudden deprivation of blood flow to a brain region and defined by different post-injury phases, which involve various molecular and cellular cascades. At an early stage during the acute phase, fast initial cell death occurs, followed by inflammation and scarring. This is followed by a sub-acute or recovery phase when endogenous plasticity mechanisms may promote spontaneous recovery, depending on various factors that are yet to be completely understood. At later time points, stroke leads to greater neurodegeneration compared to healthy controls in both clinical and preclinical studies, this is evident during the chronic phase when recovery slows down and neurodegenerative signatures appear. Astrocytes have been studied in the context of ischemic stroke due to their role in glutamate re-uptake, as components of the neurovascular unit, as building blocks of the glial scar, and synaptic plasticity regulators. All these roles render astrocytes interesting, yet understudied players in the context of stroke-induced neurodegeneration. With this review, we provide a summary of previous research, highlight astrocytes as potential therapeutic targets, and formulate questions about the role of astrocytes in the mechanisms during the acute, sub-acute, and chronic post-stroke phases that may lead to neurorestoration or neurodegeneration.
Collapse
Affiliation(s)
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
9
|
Paul M, Paul JW, Hinwood M, Hood RJ, Martin K, Abdolhoseini M, Johnson SJ, Pollack M, Nilsson M, Walker FR. Clopidogrel Administration Impairs Post-Stroke Learning and Memory Recovery in Mice. Int J Mol Sci 2023; 24:11706. [PMID: 37511466 PMCID: PMC10380815 DOI: 10.3390/ijms241411706] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Clopidogrel, which is one of the most prescribed antiplatelet medications in the world, is given to stroke survivors for the prevention of secondary cardiovascular events. Clopidogrel exerts its antiplatelet activity via antagonism of the P2Y12 receptor (P2RY12). Although not widely known or considered during the initial clinical trials for clopidogrel, P2RY12 is also expressed on microglia, which are the brain's immune cells, where the receptor facilitates chemotactic migration toward sites of cellular damage. If microglial P2RY12 is blocked, microglia lose the ability to migrate to damaged sites and carry out essential repair processes. We aimed to investigate whether administering clopidogrel to mice post-stroke was associated with (i) impaired motor skills and cognitive recovery; (ii) physiological changes, such as survival rate and body weight; (iii) changes in the neurovascular unit, including blood vessels, microglia, and neurons; and (iv) changes in immune cells. Photothrombotic stroke (or sham surgery) was induced in adult male mice. From 24 h post-stroke, mice were treated daily for 14 days with either clopidogrel or a control. Cognitive performance (memory and learning) was assessed using a mouse touchscreen platform (paired associated learning task), while motor impairment was assessed using the cylinder task for paw asymmetry. On day 15, the mice were euthanized and their brains were collected for immunohistochemistry analysis. Clopidogrel administration significantly impaired learning and memory recovery, reduced mouse survival rates, and reduced body weight post-stroke. Furthermore, clopidogrel significantly increased vascular leakage, significantly increased the number and appearance of microglia, and significantly reduced the number of T cells within the peri-infarct region post-stroke. These data suggest that clopidogrel hampers cognitive performance post-stroke. This effect is potentially mediated by an increase in vascular permeability post-stroke, providing a pathway for clopidogrel to access the central nervous system, and thus, interfere in repair and recovery processes.
Collapse
Affiliation(s)
- Marina Paul
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Jonathan W Paul
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Madeleine Hinwood
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW 2308, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Rebecca J Hood
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Kristy Martin
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - Mahmoud Abdolhoseini
- School of Engineering, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Sarah J Johnson
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW 2308, Australia
- School of Engineering, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Michael Pollack
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW 2308, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Michael Nilsson
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW 2308, Australia
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
- LKC School of Medicine, Nanyang Technological University, Singapore 639798, Singapore
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
10
|
Zhuo Z, Wang H, Zhang S, Bartlett PF, Walker TL, Hou ST. Selenium supplementation provides potent neuroprotection following cerebral ischemia in mice. J Cereb Blood Flow Metab 2023; 43:1060-1076. [PMID: 36756891 PMCID: PMC10291447 DOI: 10.1177/0271678x231156981] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/30/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
Despite progress in reperfusion therapy, functional recovery remains suboptimal in many stroke patients, with oxidative stress, inflammation, dysbiosis, and secondary neurodegeneration constituting the major hurdles to recovery. The essential trace element selenium is emerging as a promising therapeutic agent for stroke. However, although several rodent studies have shown that selenium can protect against cell loss following cerebral ischemia, no study has yet examined whether selenium can enhance long-term functional recovery. Moreover, published studies have typically reported a single mechanism of action underlying selenium-mediated stroke recovery. However, we propose that selenium is more likely to have multifaceted actions. Here, we show that selenomethionine confers a potent neuroprotective effect in a canonical filament-induced transient middle cerebral artery occlusion (tMCAO) mouse model. Post-tMCAO selenium treatment significantly reduces the cerebral infarct volume, oxidative stress, and ferroptosis and enhances post-tMCAO motor performance in the acute phase after stroke. Moreover, analysis of the gut microbiota reveals that acute selenium treatment reverses stroke-induced gut dysbiosis. Longer-term selenium supplementation activates intrinsic neuroprotective mechanisms, prevents secondary neurodegeneration, alleviates systemic inflammation, and diminishes gut microbe-derived circulating trimethylamine N-oxide. These findings demonstrate that selenium treatment even after cerebral ischemia has long-term and multifaceted neuroprotective effects, highlighting its clinical potential.
Collapse
Affiliation(s)
- Zhan Zhuo
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, China
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Huimei Wang
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, China
- Present address: Hearing Research Group, Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, USA
| | - Shuai Zhang
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, China
| | - Perry F Bartlett
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Tara L Walker
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Sheng-Tao Hou
- Brain Research Centre, Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
11
|
Zbesko JC, Stokes J, Becktel DA, Doyle KP. Targeting foam cell formation to improve recovery from ischemic stroke. Neurobiol Dis 2023; 181:106130. [PMID: 37068641 PMCID: PMC10993857 DOI: 10.1016/j.nbd.2023.106130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/31/2023] [Accepted: 04/14/2023] [Indexed: 04/19/2023] Open
Abstract
Inflammation is a crucial part of the healing process after an ischemic stroke and is required to restore tissue homeostasis. However, the inflammatory response to stroke also worsens neurodegeneration and creates a tissue environment that is unfavorable to regeneration for several months, thereby postponing recovery. In animal models, inflammation can also contribute to the development of delayed cognitive deficits. Myeloid cells that take on a foamy appearance are one of the most prominent immune cell types within chronic stroke infarcts. Emerging evidence indicates that they form as a result of mechanisms of myelin lipid clearance becoming overwhelmed, and that they are a key driver of the chronic inflammatory response to stroke. Therefore, targeting lipid accumulation in foam cells may be a promising strategy for improving recovery. The aim of this review is to provide an overview of current knowledge regarding inflammation and foam cell formation in the brain in the weeks and months following ischemic stroke and identify targets that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Jacob C Zbesko
- Department of Immunobiology, University of Arizona, United States
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, United States
| | | | - Kristian P Doyle
- Department of Immunobiology, University of Arizona, United States; Departments of Neurology, Neurosurgery, Psychology, Arizona Center on Aging, and the BIO5 Institute, University of Arizona, United States.
| |
Collapse
|
12
|
Warren KE, Coupland KG, Hood RJ, Kang L, Walker FR, Spratt NJ. Movement of cerebrospinal fluid tracer into brain parenchyma and outflow to nasal mucosa is reduced at 24 h but not 2 weeks post-stroke in mice. Fluids Barriers CNS 2023; 20:27. [PMID: 37041551 PMCID: PMC10088200 DOI: 10.1186/s12987-023-00427-2] [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: 12/20/2022] [Accepted: 03/27/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Recent data indicates that cerebrospinal fluid (CSF) dynamics are disturbed after stroke. Our lab has previously shown that intracranial pressure rises dramatically 24 h after experimental stroke and that this reduces blood flow to ischaemic tissue. CSF outflow resistance is increased at this time point. We hypothesised that reduced transit of CSF through brain parenchyma and reduced outflow of CSF via the cribriform plate at 24 h after stroke may contribute to the previously identified post-stroke intracranial pressure elevation. METHODS Using a photothrombotic permanent occlusion model of stroke in C57BL/6 adult male mice, we examined the movement of an intracisternally infused 0.5% Texas Red dextran throughout the brain and measured tracer efflux into the nasal mucosa via the cribriform plate at 24 h or two weeks after stroke. Brain tissue and nasal mucosa were collected ex vivo and imaged using fluorescent microscopy to determine the change in CSF tracer intensity in these tissues. RESULTS At 24 h after stroke, we found that CSF tracer load was significantly reduced in brain tissue from stroke animals in both the ipsilateral and contralateral hemispheres when compared to sham. CSF tracer load was also reduced in the lateral region of the ipsilateral hemisphere when compared to the contralateral hemisphere in stroke brains. In addition, we identified an 81% reduction in CSF tracer load in the nasal mucosa in stroke animals compared to sham. These alterations to the movement of CSF-borne tracer were not present at two weeks after stroke. CONCLUSIONS Our data indicates that influx of CSF into the brain tissue and efflux via the cribriform plate are reduced 24 h after stroke. This may contribute to reported increases in intracranial pressure at 24 h after stroke and thus worsen stroke outcomes.
Collapse
Affiliation(s)
- K E Warren
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan and Hunter Medical Research Institute, University Drive, Callaghan, New Lambton Heights, NSW, 2308, Australia
- Hunter New England Health District, New Lambton Heights, NSW, Australia
| | - K G Coupland
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan and Hunter Medical Research Institute, University Drive, Callaghan, New Lambton Heights, NSW, 2308, Australia
- Hunter New England Health District, New Lambton Heights, NSW, Australia
| | - R J Hood
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan and Hunter Medical Research Institute, University Drive, Callaghan, New Lambton Heights, NSW, 2308, Australia
- Hunter New England Health District, New Lambton Heights, NSW, Australia
| | - L Kang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan and Hunter Medical Research Institute, University Drive, Callaghan, New Lambton Heights, NSW, 2308, Australia
- Hunter New England Health District, New Lambton Heights, NSW, Australia
| | - F R Walker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan and Hunter Medical Research Institute, University Drive, Callaghan, New Lambton Heights, NSW, 2308, Australia
- Hunter New England Health District, New Lambton Heights, NSW, Australia
| | - N J Spratt
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan and Hunter Medical Research Institute, University Drive, Callaghan, New Lambton Heights, NSW, 2308, Australia.
- Hunter New England Health District, New Lambton Heights, NSW, Australia.
| |
Collapse
|
13
|
Cotter KM, Bancroft GL, Haas HA, Shi R, Clarkson AN, Croxall ME, Stowe AM, Yun S, Eisch AJ. Use of an Automated Mouse Touchscreen Platform for Quantification of Cognitive Deficits After Central Nervous System Injury. Methods Mol Biol 2023; 2616:279-326. [PMID: 36715942 DOI: 10.1007/978-1-0716-2926-0_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Analyzing cognitive performance is an important aspect of assessing physiological deficits after stroke or other central nervous system (CNS) injuries in both humans and in basic science animal models. Cognitive testing on an automated touchscreen operant platform began in humans but is now increasingly popular in preclinical studies as it enables testing in many cognitive domains in a highly reproducible way while minimizing stress to the laboratory animal. Here, we describe the step-by-step setup and application of four operant touchscreen tests used on adult mice. In brief, mice are trained to touch a graphical image on a lit screen and initiate subsequent trials for a reward. Following initial training, mice can be tested on tasks that probe performance in many cognitive domains and thus infer the integrity of brain circuits and regions. There are already many outstanding published protocols on touchscreen cognitive testing. This chapter is designed to add to the literature in two specific ways. First, this chapter provides in a single location practical, behind-the-scenes tips for setup and testing of mice in four touchscreen tasks that are useful to assess in CNS injury models: Paired Associates Learning (PAL), a task of episodic, associative (object-location) memory; Location Discrimination Reversal (LDR), a test for mnemonic discrimination (also called behavioral pattern separation) and cognitive flexibility; Autoshaping (AUTO), a test of Pavlovian or classical conditioning; and Extinction (EXT), tasks of stimulus-response and response inhibition, respectively. Second, this chapter summarizes issues to consider when performing touchscreen tests in mouse models of CNS injury. Quantifying gross and fine aspects of cognitive function is essential to improved treatment for brain dysfunction after stroke or CNS injury as well as other brain diseases, and touchscreen testing provides a sensitive, reliable, and robust way to achieve this.
Collapse
Affiliation(s)
- Katherine M Cotter
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA
| | | | | | - Raymon Shi
- University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | | | - Ann M Stowe
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA. .,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Amelia J Eisch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA. .,Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Beyond the Primary Infarction: Focus on Mechanisms Related to Secondary Neurodegeneration after Stroke. Int J Mol Sci 2022; 23:ijms232416024. [PMID: 36555665 PMCID: PMC9782185 DOI: 10.3390/ijms232416024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Recently, a growing body of evidence has indicated that secondary neurodegeneration after stroke occurs at remote regions of the brain that are connected to the primary infarction site [...].
Collapse
|
15
|
Noll JM, Augello CJ, Kürüm E, Pan L, Pavenko A, Nam A, Ford BD. Spatial Analysis of Neural Cell Proteomic Profiles Following Ischemic Stroke in Mice Using High-Plex Digital Spatial Profiling. Mol Neurobiol 2022; 59:7236-7252. [PMID: 36151369 PMCID: PMC9616789 DOI: 10.1007/s12035-022-03031-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/09/2022] [Indexed: 10/14/2022]
Abstract
Stroke is ranked as the fifth leading cause of death and the leading cause of adult disability in the USA. The progression of neuronal damage after stroke is recognized to be a complex integration of glia, neurons, and the surrounding extracellular matrix, therefore potential treatments must target the detrimental effects created by these interactions. In this study, we examined the spatial cellular and neuroinflammatory mechanisms occurring early after ischemic stroke utilizing Nanostring Digital Spatial Profiling (DSP) technology. Male C57bl/6 mice were subjected to photothrombotic middle cerebral artery occlusion (MCAO) and sacrificed at 3 days post-ischemia. Spatial distinction of the ipsilateral hemisphere was studied according to the regions of interest: the ischemic core, peri-infarct tissues, and peri-infarct normal tissue (PiNT) in comparison to the contralateral hemisphere. We demonstrated that the ipsilateral hemisphere initiates distinct spatial regulatory proteomic profiles with DSP technology that can be identified consistently with the immunohistochemical markers, FJB, GFAP, and Iba-1. The core border profile demonstrated an induction of neuronal death, apoptosis, autophagy, immunoreactivity, and early degenerative proteins. Most notably, the core border resulted in a decrease of the neuronal proteins Map2 and NeuN; an increase in the autophagy proteins BAG3 and CTSD; an increase in the microglial and peripheral immune invasion proteins Iba1, CD45, CD11b, and CD39; and an increase in the neurodegenerative proteins BACE1, APP, amyloid β 1-42, ApoE, and hyperphosphorylated tau protein S-199. The peri-infarct region demonstrated increased astrocytic, immunoreactivity, apoptotic, and neurodegenerative proteomic profiles, with an increase in BAG3, GFAP, and hyperphosphorylated tau protein S-199. The PiNT region displayed minimal changes compared to the contralateral cortex with only an increase in GFAP. In this study, we showed that mechanisms known to be associated with stroke, such as apoptosis and inflammation, occur in distinct spatial domains of the injured brain following ischemia. We also demonstrated the dysregulation of specific autophagic pathways that may lead to neurodegeneration in peri-infarct brain tissues. Taken together, these data suggest that identifying post-ischemic mechanisms occurring in a spatiotemporal manner may lead to more precise targets for successful therapeutic interventions to treat stroke.
Collapse
Affiliation(s)
- Jessica M Noll
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, 900 University Ave, Riverside, CA, 92521, USA
| | - Catherine J Augello
- Division of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Esra Kürüm
- Department of Statistics, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Liuliu Pan
- Nanostring Technologies, Seattle, WA, 98109, USA
| | - Anna Pavenko
- Nanostring Technologies, Seattle, WA, 98109, USA
| | - Andy Nam
- Nanostring Technologies, Seattle, WA, 98109, USA
| | - Byron D Ford
- Division of Biomedical Sciences, University of California-Riverside School of Medicine, 900 University Ave, Riverside, CA, 92521, USA.
| |
Collapse
|
16
|
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
|
17
|
Wang H, Zhang M, Li J, Liang J, Yang M, Xia G, Ren Y, Zhou H, Wu Q, He Y, Yin J. Gut microbiota is causally associated with poststroke cognitive impairment through lipopolysaccharide and butyrate. J Neuroinflammation 2022; 19:76. [PMID: 35379265 PMCID: PMC8981610 DOI: 10.1186/s12974-022-02435-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022] Open
Abstract
Background Poststroke cognitive impairment (PSCI) is prevalent in stroke patients. The etiology of PSCI remains largely unknown. We previously found that stroke induces gut microbiota dysbiosis which affects brain injury. Hereby, we aimed to investigate whether the gut microbiota contributes to the pathogenesis of PSCI. Methods 83 stroke patients were recruited and their cognitive function were measured by Montreal Cognitive Assessment (MoCA) scores 3 months after stroke onset. The peripheral inflammatory factor levels and gut microbiota compositions of the patients were analyzed. Fecal microbiota transplantation from patients to stroke mice was performed to examine the causal relationship between the gut microbiota and PSCI. The cognitive function of mice was evaluated by Morris water maze test. Results 34 and 49 stroke patients were classified as PSCI and non-PSCI, respectively. Compared with non-PSCI patients, PSCI patients showed significantly higher levels of gut Enterobacteriaceae, lipopolysaccharide (LPS) and peripheral inflammation markers. Consistently, stroke mice that received microbiota from PSCI patients (PSCI mice) presented a higher level of Enterobacteriaceae, intestinal Toll-like receptor-4 (TLR4) expression, circulating LPS, LPS-binding protein (LBP) and inflammatory cytokines, and a lower level of fecal butyrate, severer intestine destruction and cognitive impairment than mice that received microbiota from nPSCI patients (nPSCI mice). In addition, we observed exacerbations in blood–brain barrier (BBB) integrity, microglial activation, neuronal apoptosis in the CA1 region of the hippocampus, and Aβ deposition in the thalamus of PSCI mice in comparison with nPSCI mice. Intraperitoneal injection of LPS after stroke caused similar pathology to those seen in PSCI mice. Supplementation with sodium butyrate (NaB) via drinking water rescued these detrimental changes in PSCI mice. Conclusions Our data indicate a cause–effect relationship between gut microbiota and PSCI for the first time, which is likely mediated by inflammation-regulating metabolites including LPS and butyrate. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02435-9.
Collapse
Affiliation(s)
- Huidi Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingsi Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhai Liang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mengjia Yang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Genghong Xia
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yueran Ren
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiheng Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jia Yin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Kiselev A, Kotov A, Mikhaleva M, Stovbun S, Kotov S. Ampakines — a promising approach to neuroprotection. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:54-62. [DOI: 10.17116/jnevro202212209154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
19
|
Brown GC. Neuronal Loss after Stroke Due to Microglial Phagocytosis of Stressed Neurons. Int J Mol Sci 2021; 22:13442. [PMID: 34948237 PMCID: PMC8707068 DOI: 10.3390/ijms222413442] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/17/2022] Open
Abstract
After stroke, there is a rapid necrosis of all cells in the infarct, followed by a delayed loss of neurons both in brain areas surrounding the infarct, known as 'selective neuronal loss', and in brain areas remote from, but connected to, the infarct, known as 'secondary neurodegeneration'. Here we review evidence indicating that this delayed loss of neurons after stroke is mediated by the microglial phagocytosis of stressed neurons. After a stroke, neurons are stressed by ongoing ischemia, excitotoxicity and/or inflammation and are known to: (i) release "find-me" signals such as ATP, (ii) expose "eat-me" signals such as phosphatidylserine, and (iii) bind to opsonins, such as complement components C1q and C3b, inducing microglia to phagocytose such neurons. Blocking these factors on neurons, or their phagocytic receptors on microglia, can prevent delayed neuronal loss and behavioral deficits in rodent models of ischemic stroke. Phagocytic receptors on microglia may be attractive treatment targets to prevent delayed neuronal loss after stroke due to the microglial phagocytosis of stressed neurons.
Collapse
Affiliation(s)
- Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
20
|
Stuckey SM, Ong LK, Collins-Praino LE, Turner RJ. Neuroinflammation as a Key Driver of Secondary Neurodegeneration Following Stroke? Int J Mol Sci 2021; 22:ijms222313101. [PMID: 34884906 PMCID: PMC8658328 DOI: 10.3390/ijms222313101] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023] Open
Abstract
Ischaemic stroke involves the rapid onset of focal neurological dysfunction, most commonly due to an arterial blockage in a specific region of the brain. Stroke is a leading cause of death and common cause of disability, with over 17 million people worldwide suffering from a stroke each year. It is now well-documented that neuroinflammation and immune mediators play a key role in acute and long-term neuronal tissue damage and healing, not only in the infarct core but also in distal regions. Importantly, in these distal regions, termed sites of secondary neurodegeneration (SND), spikes in neuroinflammation may be seen sometime after the initial stroke onset, but prior to the presence of the neuronal tissue damage within these regions. However, it is key to acknowledge that, despite the mounting information describing neuroinflammation following ischaemic stroke, the exact mechanisms whereby inflammatory cells and their mediators drive stroke-induced neuroinflammation are still not fully understood. As a result, current anti-inflammatory treatments have failed to show efficacy in clinical trials. In this review we discuss the complexities of post-stroke neuroinflammation, specifically how it affects neuronal tissue and post-stroke outcome acutely, chronically, and in sites of SND. We then discuss current and previously assessed anti-inflammatory therapies, with a particular focus on how failed anti-inflammatories may be repurposed to target SND-associated neuroinflammation.
Collapse
Affiliation(s)
- Shannon M. Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan 2308, Australia
| | - Lyndsey E. Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Renée J. Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
- Correspondence: ; Tel.: +61-8-8313-3114
| |
Collapse
|
21
|
Is Cerebral Amyloid-β Deposition Related to Post-stroke Cognitive Impairment? Transl Stroke Res 2021; 12:946-957. [PMID: 34195928 DOI: 10.1007/s12975-021-00921-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 01/20/2023]
Abstract
Approximately two-thirds of ischemic stroke patients suffer from different levels of post-stroke cognitive impairment (PSCI), but the underlying mechanisms of PSCI remain unclear. Cerebral amyloid-β (Aβ) deposition, a pathological hallmark of Alzheimer's disease, has been discovered in the brains of stroke patients in some autopsy studies. However, less is known about the role of Aβ pathology in the development of PSCI. It is hypothesized that cerebral ischemic injury may lead to neurotoxic Aβ accumulation in the brain, which further induces secondary neurodegeneration and progressive cognitive decline after stroke onset. In this review, we summarized available evidence from pre-clinical and clinical studies relevant to the aforementioned hypothesis. We found inconsistency in the results obtained from studies in rodents, nonhuman primates, and stroke patients. Moreover, the causal relationship between post-stroke cerebral Aβ deposition and PSCI has been uncertain and controversial. Taken together, evidence supporting the hypothesis that brain ischemia induces cerebral Aβ deposition has been insufficient so far. And, there is still no consensus regarding the contribution of cerebral amyloid pathology to PSCI. Other non-amyloid neurodegenerative mechanisms might be involved and remain to be fully elucidated.
Collapse
|
22
|
Corticosterone Administration Alters White Matter Tract Structure and Reduces Gliosis in the Sub-Acute Phase of Experimental Stroke. Int J Mol Sci 2021; 22:ijms22136693. [PMID: 34206635 PMCID: PMC8269094 DOI: 10.3390/ijms22136693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 11/17/2022] Open
Abstract
White matter tract (WMT) degeneration has been reported to occur following a stroke, and it is associated with post-stroke functional disturbances. White matter pathology has been suggested to be an independent predictor of post-stroke recovery. However, the factors that influence WMT remodeling are poorly understood. Cortisol is a steroid hormone released in response to prolonged stress, and elevated levels of cortisol have been reported to interfere with brain recovery. The objective of this study was to investigate the influence of corticosterone (CORT; the rodent equivalent of cortisol) on WMT structure post-stroke. Photothrombotic stroke (or sham surgery) was induced in 8-week-old male C57BL/6 mice. At 72 h, mice were exposed to standard drinking water ± CORT (100 µg/mL). After two weeks of CORT administration, mice were euthanised and brain tissue collected for histological and biochemical analysis of WMT (particularly the corpus callosum and corticospinal tract). CORT administration was associated with increased tissue loss within the ipsilateral hemisphere, and modest and inconsistent WMT reorganization. Further, a structural and molecular analysis of the WMT components suggested that CORT exerted effects over axons and glial cells. Our findings highlight that CORT at stress-like levels can moderately influence the reorganization and microstructure of WMT post-stroke.
Collapse
|