1
|
Zhang J, Hong J, Chen J, Zhao F, Ye Q, Shan Y, Li C, Wen H. ScRNAs reveals high-frequency rTMS-induced pericyte differentiation: Potential implications for vascular regeneration and blood-brain barrier stability in stroke. Heliyon 2024; 10:e35339. [PMID: 39229501 PMCID: PMC11369456 DOI: 10.1016/j.heliyon.2024.e35339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/14/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Stroke is a major cause of adult disability worldwide, often involving disruption of the blood-brain barrier (BBB). Repairing the BBB is crucial for stroke recovery, and pericytes, essential components of the BBB, are potential intervention targets. Repetitive transcranial magnetic stimulation (rTMS) has been proposed as a treatment for functional impairments after stroke, with potential effects on BBB integrity. However, the underlying mechanisms remain unclear. In this study using a transient middle cerebral artery occlusion (tMCAO) rat model, we investigated the impact of rTMS on post-stroke BBB. Through single-cell sequencing (ScRNAs), we observed developmental relationships among pericytes, endothelial cells, and vascular smooth muscle cells, suggesting the differentiation potential of pericytes. A distinct subcluster of pericytes emerged as a potential therapeutic target for stroke. Additionally, our results revealed enhanced cellular communication among these cell types, enriching signaling pathways such as IGF, TNF, NOTCH, and ICAM. Analysis of differentially expressed genes highlighted processes related to stress, differentiation, and development. Notably, rTMS intervention upregulated Reck in vascular smooth muscle cells, implicating its role in the classical Wnt signaling pathway. Overall, our bioinformatics findings suggest that rTMS may modulate BBB permeability and promote vascular regeneration following stroke. This might happen through 20 Hz rTMS promoting pericyte differentiation into vascular smooth muscle cells, upregulating Reck, then activating the classical Wnt signaling pathway, and facilitating vascular regeneration and BBB stability.
Collapse
Affiliation(s)
| | | | - Jiemei Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Fei Zhao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Qiuping Ye
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yilong Shan
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Chao Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Hongmei Wen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| |
Collapse
|
2
|
Yu F, Wang F, Li K, Du G, Deng B, Xie H, Yang G, Xiao T. Real-time X-ray imaging of mouse cerebral microvessels in vivo using a pixel temporal averaging method. JOURNAL OF SYNCHROTRON RADIATION 2022; 29:239-246. [PMID: 34985441 PMCID: PMC8733992 DOI: 10.1107/s1600577521012522] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/25/2021] [Indexed: 06/14/2023]
Abstract
Rodents are used extensively as animal models for the preclinical investigation of microvascular-related diseases. However, motion artifacts in currently available imaging methods preclude real-time observation of microvessels in vivo. In this paper, a pixel temporal averaging (PTA) method that enables real-time imaging of microvessels in the mouse brain in vivo is described. Experiments using live mice demonstrated that PTA efficiently eliminated motion artifacts and random noise, resulting in significant improvements in contrast-to-noise ratio. The time needed for image reconstruction using PTA with a normal computer was 250 ms, highlighting the capability of the PTA method for real-time angiography. In addition, experiments with less than one-quarter of photon flux in conventional angiography verified that motion artifacts and random noise were suppressed and microvessels were successfully identified using PTA, whereas conventional temporal subtraction and averaging methods were ineffective. Experiments performed with an X-ray tube verified that the PTA method could also be successfully applied to microvessel imaging of the mouse brain using a laboratory X-ray source. In conclusion, the proposed PTA method may facilitate the real-time investigation of cerebral microvascular-related diseases using small animal models.
Collapse
Affiliation(s)
- Fucheng Yu
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201204, People’s Republic of China
- Shanghai Synchrotron Radiation Facility/Zhang Jiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201800, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Feixiang Wang
- Shanghai Synchrotron Radiation Facility/Zhang Jiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201800, People’s Republic of China
| | - Ke Li
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201204, People’s Republic of China
- Shanghai Synchrotron Radiation Facility/Zhang Jiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201800, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Guohao Du
- Shanghai Synchrotron Radiation Facility/Zhang Jiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201800, People’s Republic of China
| | - Biao Deng
- Shanghai Synchrotron Radiation Facility/Zhang Jiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201800, People’s Republic of China
| | - Honglan Xie
- Shanghai Synchrotron Radiation Facility/Zhang Jiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201800, People’s Republic of China
| | - Guoyuan Yang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Tiqiao Xiao
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201204, People’s Republic of China
- Shanghai Synchrotron Radiation Facility/Zhang Jiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201800, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
3
|
Mamtilahun M, Jiang L, Song Y, Shi X, Liu C, Jiang Y, Deng L, Zheng H, Shen H, Li Y, Zhang Z, Wang Y, Tang Y, Yang GY. Plasma from healthy donors protects blood-brain barrier integrity via FGF21 and improves the recovery in a mouse model of cerebral ischaemia. Stroke Vasc Neurol 2021; 6:561-571. [PMID: 33785536 PMCID: PMC8717795 DOI: 10.1136/svn-2020-000774] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 01/14/2023] Open
Abstract
Background Healthy plasma therapy reverses cognitive deficits and promotes neuroplasticity in ageing brain disease. However, whether healthy plasma therapy improve blood–brain barrier integrity after stroke remains unknown. Methods Here, we intravenously injected healthy female mouse plasma into adult female ischaemic stroke C57BL/6 mouse induced by 90 min transient middle cerebral artery occlusion for eight consecutive days. Infarct volume, brain atrophy and neurobehavioural tests were examined to assess the outcomes of plasma treatment. Cell apoptosis, blood–brain barrier integrity and fibroblast growth factor 21 knockout mice were used to explore the underlying mechanism. Results Plasma injection improved neurobehavioural recovery and decreased infarct volume, brain oedema and atrophy after stroke. Immunostaining showed that the number of transferase dUTP nick end labelling+/NeuN+ cells decreased in the plasma-injected group. Meanwhile, plasma injection reduced ZO-1, occluding and claudin-5 tight junction gap formation and IgG extravasation at 3 days after ischaemic stroke. Western blot results showed that the FGF21 expression increased in the plasma-injected mice. However, using FGF21 knockout mouse plasma injecting to the ischaemic wild-type mice diminished the neuroprotective effects. Conclusions Our study demonstrated that healthy adult plasma treatment protected the structural and functional integrity of blood–brain barrier, reduced neuronal apoptosis and improved functional recovery via FGF21, opening a new avenue for ischaemic stroke therapy.
Collapse
Affiliation(s)
- Muyassar Mamtilahun
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Lu Jiang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Yaying Song
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Xiaojing Shi
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Chang Liu
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Yixu Jiang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Lidong Deng
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Haoran Zheng
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Hui Shen
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Yongfang Li
- Department of Neurology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Yaohui Tang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China .,Department of Neurology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| |
Collapse
|
4
|
Platelet endothelial cell adhesion molecule-1 is a gatekeeper of neutrophil transendothelial migration in ischemic stroke. Brain Behav Immun 2021; 93:277-287. [PMID: 33388423 DOI: 10.1016/j.bbi.2020.12.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/24/2020] [Accepted: 12/22/2020] [Indexed: 11/21/2022] Open
Abstract
RATIONALE Adhesion molecules are key elements in stroke-induced brain injury by regulating the migration of effector immune cells from the circulation to the lesion site. Platelet endothelial cell adhesion molecule-1 (PECAM-1) is an adhesion molecule highly expressed on endothelial cells and leukocytes, which controls the final steps of trans-endothelial migration. A functional role for PECAM-1 in post-ischemic brain injury has not yet been demonstrated. OBJECTIVE Using genetic Pecam-1 depletion and PECAM-1 blockade using a neutralizing anti-PECAM-1 antibody, we evaluated the role of PECAM-1 mediated trans-endothelial immune cell migration for ischemic injury, delayed brain atrophy, and brain immune cell infiltrates. Trans-endothelial immune cell migration was furthermore evaluated in cultured human cerebral microvascular endothelial cells. METHODS AND RESULTS Transient middle cerebral artery occlusion (tMCAO) was induced in 10-12-week-old male Pecam-1-/- and Pecam-1+/+ wildtype mice. PECAM-1 levels increased in the ischemic brain tissue due to the infiltration of PECAM-1+ leukocytes. Using magnetic resonance imaging, we observed smaller infarct volume, less edema formation, and less brain atrophy in Pecam-1-/- compared with Pecam-1+/+ wildtype mice. The transmigration of leukocytes, specifical neutrophils, was selectively reduced by Pecam-1-/-, as shown by immune fluorescence and flow cytometry in vivo and transmigration assays in vitro. Importantly, inhibition with an anti-PECAM-1 antibody in wildtype mice decreased neutrophil brain influx and infarct. CONCLUSION PECAM-1 controls the trans-endothelial migration of neutrophils in a mouse model of ischemic stroke. Antibody blockade of PECAM-1 after stroke onset ameliorates stroke severity in mice, making PECAM-1 an interesting target to dampen post-stroke neuroinflammation, reduce ischemic brain injury, and enhance post-ischemic brain remodeling.
Collapse
|
5
|
Lian MJ, Huang CL, Lee TM. Novel system in vitro of classifying oral carcinogenesis based on feature extraction for gray-level co-occurrence matrix using scanned laser pico projector. Lasers Med Sci 2021; 37:215-224. [PMID: 33528670 DOI: 10.1007/s10103-020-03215-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/26/2020] [Indexed: 11/29/2022]
Abstract
Oral cancer is among the top 10 causes of death due to cancer worldwide. The prognosis for oral cancer patients is not good, with a 5-year survival rate of only 50%. Earlier and more precise classification will help clinicians make a diagnosis and patients survive. With the advancement of technology, computer-aided detection methods are used to help clinicians form therapy strategies. Gray-level co-occurrence matrix (GLCM) feature extraction of images describing the spatial distribution of gray levels is widely used in medical imaging analysis. Scanned laser pico projector (SLPP) has advantages such as high intensity, directivity, coherence, and mono-color with low bandwidth. In this study, GLCM feature extraction and SLPP reflex images were combined to make a small, non-staining, noninvasive classification system. According to the various image characteristics in oral carcinogenesis, SLPP reflex images better define the borders and three-dimensional structures and provide effective GLCM features such as contrast, energy, and homogeneity to classify carcinogenesis in dysplastic oral keratinocyte (DOK) and normal oral keratinocyte (NOK) cells. Moreover, it also reliably classifies highly metastatic (HSC-3) and tongue cancer (CAL-27) cells. A promising computer-aided classification system for oral cancer was developed to build a reliable intraoral examination system for in situ computer-aided diagnosis in normal clinics.
Collapse
Affiliation(s)
- Meng-Jia Lian
- School of Dentistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Chih-Ling Huang
- Center for Fundamental Science, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Tzer-Min Lee
- School of Dentistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
6
|
Hong J, Chen J, Li C, An D, Tang Z, Wen H. High-Frequency rTMS Improves Cognitive Function by Regulating Synaptic Plasticity in Cerebral Ischemic Rats. Neurochem Res 2021; 46:276-286. [PMID: 33136229 DOI: 10.1007/s11064-020-03161-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 01/07/2023]
Abstract
Poststroke cognitive impairment (PSCI) is one of the most severe sequelae of stroke and lacks effective treatment. Previous studies have shown that high-frequency repetitive transcranial magnetic stimulation (rTMS) may be a promising PSCI therapeutic approach, but the underlying mechanism is unclear. To uncover the effect of rTMS on PSCI, a transient middle cerebral artery occlusion (tMCAO) model was established. Modified Neurological Severity Score (mNSS) test and Morris Water Maze (MWM) test were performed to assess the neurological and cognitive function of rats. Furthermore, to explore the underlying mechanism, differentially expressed genes (DEGs) in the hippocampus of rats in the rTMS group and tMCAO group were compared using RNA sequencing. Then, bioinformatics analysis, including gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and protein-protein interaction (PPI) network analysis, was conducted to elaborate these DEGs. Our results indicated that high-frequency rTMS could significantly improve neurological and cognitive function, according to mNSS and MWM tests. We found 85 DEGs, including 71 upregulated genes and 14 downregulated genes, between the rTMS group and tMCAO group. The major functional category was related to chemical synaptic transmission modulation and several DEGs were significantly upregulated in processes related to synaptic plasticity, such as glutamatergic synapses. Calb2, Zic1, Kcnk9, and Grin3a were notable in PPI analysis. These results demonstrate that rTMS has a beneficial effect on PSCI, and its mechanism may be related to the regulation of synaptic plasticity and functional genes such as Calb2, Zic1, Kcnk9, and Grin3a in the hippocampus.
Collapse
Affiliation(s)
- Jiena Hong
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jiemei Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Chao Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Delian An
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Zhiming Tang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Hongmei Wen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
7
|
Mamtilahun M, Wei Z, Qin C, Wang Y, Tang Y, Shen FX, Tian HL, Zhang Z, Yang GY. DL-3n-Butylphthalide Improves Blood-Brain Barrier Integrity in Rat After Middle Cerebral Artery Occlusion. Front Cell Neurosci 2021; 14:610714. [PMID: 33510620 PMCID: PMC7835508 DOI: 10.3389/fncel.2020.610714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/23/2020] [Indexed: 11/13/2022] Open
Abstract
Objective: DL-3n-butylphthalide (NBP) has beneficial effects in different stages of ischemic stroke. Our previous studies have demonstrated that NBP promoted angiogenesis in the perifocal region of the ischemic brain. However, the molecular mechanism of NBP for blood–brain barrier protection in acute ischemic stroke was unclear. Here, we explored the neuroprotective effects of NBP on blood–brain barrier integrity in the acute phase of ischemic stroke in a rat model. Methods: Adult male Sprague–Dawley rats (n = 82) underwent 2 h of transient middle cerebral artery occlusion and received 90 mg/kg of NBP for 3 days. Brain edema, infarct volume, surface blood flow, and neurological severity score were evaluated. Blood–brain barrier integrity was evaluated by Evans blue leakage and changes in tight junction proteins. We further examined AQP4 and eNOS expression, MMP-9 enzyme activity, and possible signaling pathways for the role of NBP after ischemic stroke. Results: NBP treatment significantly increased eNOS expression and surface blood flow in the brain, reduced brain edema and infarct volume, and improved neurological severity score compared to the control group (p < 0.05). Furthermore, NBP attenuated Evans blue and IgG leakage and increased tight junction protein expression compared to the control after 1 and 3 days of ischemic stroke (p < 0.05). Finally, NBP decreased AQP4 expression, MMP-9 enzyme activity, and increased MAPK expression during acute ischemic stroke. Conclusion: NBP protected blood–brain barrier integrity and attenuated brain injury in the acute phase of ischemic stroke by decreasing AQP4 expression and MMP-9 enzyme activity. The MAPK signaling pathway may be associated in this process.
Collapse
Affiliation(s)
- Muyassar Mamtilahun
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenyu Wei
- University of Shanghai for Science and Technology Affiliated Shidong Hospital, Shanghai, China
| | - Chuan Qin
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongting Wang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yaohui Tang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Fan-Xia Shen
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Heng-Li Tian
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Ma Y, Jiang L, Wang L, Li Y, Liu Y, Lu W, Shi R, Zhang L, Fu Z, Qu M, Liu Y, Wang Y, Zhang Z, Yang GY. Endothelial progenitor cell transplantation alleviated ischemic brain injury via inhibiting C3/C3aR pathway in mice. J Cereb Blood Flow Metab 2020; 40:2374-2386. [PMID: 31865842 PMCID: PMC7820683 DOI: 10.1177/0271678x19892777] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endothelial progenitor cell transplantation is a potential therapeutic approach in brain ischemia. However, whether the therapeutic effect of endothelial progenitor cells is via affecting complement activation is unknown. We established a mouse focal ischemia model (n = 111) and transplanted endothelial progenitor cells into the peri-infarct region immediately after brain ischemia. Neurological outcomes and brain infarct/atrophy volume were examined after ischemia. Expression of C3, C3aR and pro-inflammatory factors were further examined to explore the role of endothelial progenitor cells in ischemic brain. We found that endothelial progenitor cells improved neurological outcomes and reduced brain infarct/atrophy volume after 1 to 14 days of ischemia compared to the control (p < 0.05). C3 and C3aR expression in the brain was up-regulated at 1 day up to 14 days (p < 0.05). Endothelial progenitor cells reduced astrocyte-derived C3 (p < 0.05) and C3aR expression (p < 0.05) after ischemia. Endothelial progenitor cells also reduced inflammatory response after ischemia (p < 0.05). Endothelial progenitor cell transplantation reduced astrocyte-derived C3 expression in the brain after ischemic stroke, together with decreased C3aR and inflammatory response contributing to neurological function recovery. Our results indicate that modulating complement C3/C3aR pathway is a novel therapeutic target for the ischemic stroke.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Jiang
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Liping Wang
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongfang Li
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqun Liu
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wenjing Lu
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Rubing Shi
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Linyuan Zhang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zongjie Fu
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Meijie Qu
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yingling Liu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yongting Wang
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Yamato K, Nakajo Y, Yamamoto-Imoto H, Kokame K, Miyata T, Takahashi JC, Kataoka H, Yanamoto H. Low-Dose Activated Protein C Suppresses the Development of Cerebral Infarction and Neurological Deficits in Mice. NEUROSURGERY OPEN 2020. [DOI: 10.1093/neuopn/okaa014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
BACKGROUND
A large prospective study previously reported that a higher plasma level of protein C (PC) was associated with a lower incidence of ischemic stroke.
OBJECTIVE
To investigate the neuroprotective properties of activated PC (APC) against acute ischemic stroke using the 3-vessel occlusion model.
METHODS
Male C57BL/6J mice received APC (human APC) at 0.25, 0.5, or 1.0 (low dose) or 2.0, 4.0, or 8.0 mg/kg (high dose). Edaravone (Eda) (1.0, 3.0, or 10 mg/kg, a neuroprotectant approved for use in Japan), albumin (2.0 mg/kg), heparin (100 or 600 U/kg), or saline was used as the control. The drug or control was administered intravenously twice in the initial 24 h or 5 times in 3 d, starting 5 min after the induction of ischemia.
RESULTS
Low-dose APC significantly reduced lesion volumes, not affecting the depth of ischemia. High-dose APC did not significantly reduce lesion volumes, causing hemorrhagic transformation in some cases. In the chronic phase, lesion volumes were significantly suppressed in the APC or Eda group, and only the APC group showed a significant attenuation of neurological deficits. The protease-activated receptor (PAR)-1 antagonist SCH79797, administered during preischemia, completely abolished APC-induced neuroprotection. The overshoot-like abrupt recovery in regional cerebral blood flow observed in the control in the initial reperfusion phase was significantly suppressed by the APC treatment, indicating that the cerebral autoregulation system, consisting of endothelial cells and blood-brain barrier functions, was preserved.
CONCLUSION
Low-dose APC, potentially via the PAR-1-dependent anti-inflammatory cascade, protects the brain against ischemic stroke without increasing the risk of hemorrhagic transformation or death.
Collapse
Affiliation(s)
- Keiko Yamato
- Laboratory of Neurology and Neurosurgery, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yukako Nakajo
- Laboratory of Neurology and Neurosurgery, National Cerebral and Cardiovascular Center, Suita, Japan
- Research Laboratories, Rakuwa-kai Otowa Hospital, Kyoto, Japan
| | - Hitomi Yamamoto-Imoto
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Koichi Kokame
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Toshiyuki Miyata
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Jun C Takahashi
- Department of Neurosurgery, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hiroharu Kataoka
- Department of Neurosurgery, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hiroji Yanamoto
- Laboratory of Neurology and Neurosurgery, National Cerebral and Cardiovascular Center, Suita, Japan
- Department of Cardiovascular Science, Division of Surgical Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
10
|
Li Y, Yao N, Zhang T, Guo F, Niu X, Wu Z, Hou S. Ability of Post-treatment Glycyrrhizic Acid to Mitigate Cerebral Ischemia/Reperfusion Injury in Diabetic Mice. Med Sci Monit 2020; 26:e926551. [PMID: 32981927 PMCID: PMC7531204 DOI: 10.12659/msm.926551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background Diabetes aggravates cerebral ischemia/reperfusion (I/R) injury by increasing inflammatory reactions, but its specific mechanism is currently unclear. Material/Methods Diabetes was induced in mice with a high-fat diet combined with streptozotocin. These mice were subjected to transient middle cerebral artery occlusion (tMCAO) for 60 min, followed by reperfusion for 24–72 h and post-treatment glycyrrhizic acid (GA). Control and diabetic mice were randomly allocated to 8 groups of 18 mice each. Blood glucose, brain infarction, brain edema, and neurological function were monitored. Necrosis was determined by Nissl staining, loss of neurons by immunofluorescent (IF) staining for NeuN, and activation of inflammatory microglia by IF staining for Iba-1. Levels of HMGB1, TLR4, Myd88, and NF-κB mRNA and protein in ischemic brain were determined by qRT-PCR and western blotting, respectively, and serum concentrations of IL-1β, IL-6, and TNF-α by ELISA. Results Infarction volume, brain edema, and neurological function after tMCAO were significantly aggravated in diabetes, but ameliorated by post-treatment GA. GA also reduced neuronal loss and microglial activation. Cerebral Myd88 level showed a positive correlation with neurological scores. GA suppressed the expression of Myd88 and a proinflammatory pathway that included Myd88, HMGB1, TLR4, and NF-κB, as well as reducing serum concentrations of IL-1β, IL-6, and TNF-α. Conclusions Post-treatment inhibited inflammatory responses and provided therapeutic benefits in diabetic mice with cerebral I/R injury, suggesting that GA may be a candidate drug to suppress cerebral I/R in diabetic patients.
Collapse
Affiliation(s)
- Yuan Li
- School of Nursing, Ningxia Medical University, Yinchuan, Ningxia, China (mainland).,School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China (mainland)
| | - Na Yao
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China (mainland)
| | - Ting Zhang
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China (mainland)
| | - Fengying Guo
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia, China (mainland)
| | - Xiangying Niu
- Department of Clinical Pathology, Ningxia Medical University, Yinchuan, Ningxia, China (mainland)
| | - Zhihui Wu
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China (mainland)
| | - Shaozhang Hou
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China (mainland)
| |
Collapse
|
11
|
Qin C, Zhou P, Wang L, Mamtilahun M, Li W, Zhang Z, Yang GY, Wang Y. Dl-3-N-butylphthalide attenuates ischemic reperfusion injury by improving the function of cerebral artery and circulation. J Cereb Blood Flow Metab 2019; 39:2011-2021. [PMID: 29762050 PMCID: PMC6775578 DOI: 10.1177/0271678x18776833] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dl-3-N-butylphthalide (NBP) is approved in China for the treatment of ischemic stroke. Previous studies have shown that NBP promotes recovery after stroke via multiple mechanisms. However, the effect of NBP on vascular function and thrombosis remains unclear. Here, we aim to study the effect of NBP on vascular function using a rat model of transient middle cerebral artery occlusion (MCAO) and a state-of-the-art high-resolution synchrotron radiation angiography. Eighty SD rats underwent MCAO surgery. NBP (90 mg/kg) was administrated daily by gavage. Synchrotron radiation angiography was used to evaluate the cerebral vascular perfusion, vasoconstriction, and vasodilation in real-time. Neurological scores, brain infarction and atrophy were evaluated. Real-time PCR was used to assess the expression levels of thrombosis and vasoconstriction-related genes. Results revealed that NBP attenuated thrombosis after MCAO and reduced brain infarct and atrophy volume. NBP administrated at 1 and 4 h after MCAO prevented the vasoconstriction of the artery and maintained its diameter at normal level. Administrated at one week after surgery, NBP functioned as a vasodilator in rats after MCAO while displayed no vasodilating effect in sham group. Our results suggested that NBP attenuates brain injury via increasing the regional blood flow by reducing thrombosis and vasoconstriction.
Collapse
Affiliation(s)
- Chuan Qin
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Panting Zhou
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Liping Wang
- Department of Neurology, School of Medicine, Shanghai Jiao Tong University, Ruijin Hospital, Shanghai, China
| | - Muyassar Mamtilahun
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wanlu Li
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Neurology, School of Medicine, Shanghai Jiao Tong University, Ruijin Hospital, Shanghai, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
12
|
Wang J, Lin X, Mu Z, Shen F, Zhang L, Xie Q, Tang Y, Wang Y, Zhang Z, Yang GY. Rapamycin Increases Collateral Circulation in Rodent Brain after Focal Ischemia as detected by Multiple Modality Dynamic Imaging. Am J Cancer Res 2019; 9:4923-4934. [PMID: 31410191 PMCID: PMC6691378 DOI: 10.7150/thno.32676] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/17/2019] [Indexed: 01/09/2023] Open
Abstract
Rationale: Brain collaterals contribute to improving ischemic stroke outcomes. However, dynamic and timely investigations of collateral blood flow and collateral restoration in whole brains of living animals have rarely been reported. Methods: Using multiple modalities of imaging, including synchrotron radiation angiography, laser speckle imaging, and micro-CT imaging, we dynamically explored collateral circulation throughout the whole brain in the rodent middle cerebral artery occlusion model. Results: We demonstrated that compared to control animals, 4 neocollaterals gradually formed between the intra- and extra-arteries in the skull base of model animals after occlusion (p<0.05). Two main collaterals were critical to the supply of blood from the posterior to the middle cerebral artery territory in the deep brain (p<0.05). Abundant small vessel and capillary anastomoses were detected on the surface of the cortex between the posterior and middle cerebral artery and between the anterior and middle cerebral artery (p<0.05). Collateral perfusion occurred immediately (≈15 min) and was maintained for up to 14 days after occlusion. Further study revealed that administration of rapamycin at 15 min after MCAO dilated the existing collateral vessels and promoted collateral perfusion. Principal conclusions: Our results provide evidence of collateral functional perfusion in the skull base, deep brain, and surface of the cortex. Rapamycin was capable of enlarging the diameter of collaterals, potentially extending the time window for ischemic stroke therapy.
Collapse
|
13
|
Dynamic Detection of Thrombolysis in Embolic Stroke Rats by Synchrotron Radiation Angiography. Transl Stroke Res 2019; 10:695-704. [PMID: 30680639 DOI: 10.1007/s12975-019-0687-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/30/2018] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
Abstract
A rodent model of embolic middle cerebral artery occlusion is used to mimic cerebral embolism in clinical patients. Thrombolytic therapy is the effective treatment for this ischemic injury. However, it is difficult to detect thrombolysis dynamically in living animals. Synchrotron radiation angiography may provide a novel approach to directly monitor the thrombolytic process and assess collateral circulation after embolic stroke. Thirty-six adult Sprague-Dawley rats underwent the embolic stroke model procedure and were then treated with tissue plasminogen activator. The angiographic images were obtained in vivo by synchrotron radiation angiography. Synchrotron radiation angiography confirmed the successful establishment of occlusion and detected the thrombolysis process after the thrombolytic treatment. The time of thrombolytic recanalization was unstable during embolic stroke. The infarct volume increased as the recanalization time was delayed from 2 to 6 h (p < 0.05). The collateral circulation of the internal carotid artery to the ophthalmic artery, the olfactory artery to the ophthalmic artery, and the posterior cerebral artery to the middle cerebral artery opened after embolic stroke and manifested different opening rates (59%, 24%, and 75%, respectively) in the rats. The opening of the collateral circulation from the posterior cerebral artery to the middle cerebral artery alleviated infarction in rats with successful thrombolysis (p < 0.05). The cerebral vessels of the circle of Willis narrowed after thrombolysis (p < 0.05). Synchrotron radiation angiography provided a unique tool to dynamically detect and assess the thrombolysis process and the collateral circulation during thrombolytic therapy.
Collapse
|
14
|
Mangas A, Heredia M, Riolobos A, De la Fuente A, Criado JM, Yajeya J, Geffard M, Coveñas R. Overexpression of kynurenic acid and 3-hydroxyanthranilic acid after rat traumatic brain injury. Eur J Histochem 2018; 62:2985. [PMID: 30426733 PMCID: PMC6275464 DOI: 10.4081/ejh.2018.2985] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/02/2018] [Indexed: 12/14/2022] Open
Abstract
Using an immunohistochemical technique, we have studied the distribution of kynuneric acid (KYNA) and 3-hydroxyanthranilic acid (3-HAA) in a rat brain injury model (trauma). The study was carried out inducing a cerebral ablation of the frontal motor cortex. Two mouse monoclonal specific antibodies previously developed by our group directed against KYNA and 3-HAA were used. In control animals (sham-operated), the expression of both KYNA and 3-HAA was not observed. In animals in which the ablation was performed, the highest number of immunoreactive cells containing KYNA or 3-HAA was observed in the region surrounding the lesion and the number of these cells decreased moving away from the lesion. KYNA and 3-HAA were also observed in the white matter (ipsilateral side) located close to the injured region and in some cells placed in the white matter of the contralateral side. The distribution of KYNA and 3-HAA perfectly matched with the peripheral injured regions. The results found were identical independently of the perfusion date of animals (17, 30 or 54 days after brain injury). For the first time, the presence of KYNA and 3-HAA has been described in a rat trauma model. Moreover, by using a double immunocytochemistry protocol, it has been demonstrated that both metabolites were located in astrocytes. The findings observed suggest that, in cerebral trauma, KYNA and 3-HAA are involved in tissue damage and that these compounds could act, respectively, as a neuroprotector and a neurotoxic. This means that, in trauma, a counterbalance occurs and that a regulation of the indoleamine 2,3 dioxygenase (IDO) pathway could be required after a brain injury in order to decrease the deleterious effects of ending metabolites (the neurotoxic picolinic acid). Moreover, the localization of KYNA and 3-HAA in the contralateral side of the lesion suggests that the IDO pathway is also involved in the sprouting and pathfinding that follows a traumatic brain injury.
Collapse
Affiliation(s)
- Arturo Mangas
- Gemacbio, France; University of Salamanca, Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla y León.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Song H, Mylvaganam SM, Wang J, Mylvaganam SMK, Wu C, Carlen PL, Eubanks JH, Feng J, Zhang L. Contributions of the Hippocampal CA3 Circuitry to Acute Seizures and Hyperexcitability Responses in Mouse Models of Brain Ischemia. Front Cell Neurosci 2018; 12:278. [PMID: 30210302 PMCID: PMC6123792 DOI: 10.3389/fncel.2018.00278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/08/2018] [Indexed: 12/29/2022] Open
Abstract
The hippocampal circuitry is widely recognized as susceptible to ischemic injury and seizure generation. However, hippocampal contribution to acute non-convulsive seizures (NCS) in models involving middle cerebral artery occlusion (MCAO) remains to be determined. To address this, we occluded the middle cerebral artery in adult C57 black mice and monitored electroencephalographic (EEG) discharges from hippocampal and neocortical areas. Electrographic discharges in the absence of convulsive motor behaviors were observed within 90 min following occlusion of the middle cerebral artery. Hippocampal discharges were more robust than corresponding cortical discharges in all seizure events examined, and hippocampal discharges alone or with minimal cortical involvement were also observed in some seizure events. Seizure development was associated with ipsilateral hippocampal injuries as determined by subsequent histological examinations. We also introduced hypoxia-hypoglycemia episodes in mouse brain slices and examined regional hyperexcitable responses ex vivo. Extracellular recordings showed that the hippocampal CA3 region had a greater propensity for exhibiting single/multiunit activities or epileptiform field potentials following hypoxic-hypoglycemic (HH) episodes compared to the CA1, dentate gyrus, entorhinal cortical (EC) or neocortical regions. Whole-cell recordings revealed that CA3 pyramidal neurons exhibited excessive excitatory postsynaptic currents, attenuated inhibitory postsynaptic currents and intermittent or repetitive spikes in response to HH challenge. Together, these observations suggest that hippocampal discharges, possibly as a result of CA3 circuitry hyperexcitability, are a major component of acute NCS in a mouse model of MCAO.
Collapse
Affiliation(s)
- Hongmei Song
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | | | - Justin Wang
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Chiping Wu
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Peter L. Carlen
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine (Neurology), University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James H. Eubanks
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Surgery (Neurosurgery), University of Toronto, Toronto, ON, Canada
| | - Jiachun Feng
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Liang Zhang
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine (Neurology), University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Early Detection of Cerebral Infarction After Focal Ischemia Using a New MRI Indicator. Mol Neurobiol 2018; 56:658-670. [DOI: 10.1007/s12035-018-1073-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 04/10/2018] [Indexed: 10/16/2022]
|
17
|
Wang L, Mu Z, Lin X, Geng J, Xiao TQ, Zhang Z, Wang Y, Guan Y, Yang GY. Simultaneous Imaging of Cerebrovascular Structure and Function in Hypertensive Rats Using Synchrotron Radiation Angiography. Front Aging Neurosci 2017; 9:359. [PMID: 29163140 PMCID: PMC5673661 DOI: 10.3389/fnagi.2017.00359] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 10/19/2017] [Indexed: 12/12/2022] Open
Abstract
Hypertension has a profound influence on the structure and function of blood vessels. Cerebral vessels undergo both structural and functional changes in hypertensive animals. However, dynamic changes of cerebrovasculature and the factors involved in this process are largely unknown. In this study, we explored the dynamic changes of vascular structure in hypertensive rats using novel synchrotron radiation angiography. Twenty-four spontaneously hypertensive rats (SHR) and 24 Sprague–Dawley (SD) rats underwent synchrotron radiation (SR) angiography. Each group had 8 animals. We studied the cerebral vascular changes in SHR over a time period of 3–12-month and performed quantitative analysis. No vascular morphology differences between SHR and SD rats were observed in the early stage of hypertension. The number of twisted blood vessels in the front brain significantly increased at the 9- and 12-month observation time-points in the SHR compared to the SD rats (p < 0.01). The vessel density of the cortex and the striatum in SHR was consistently higher than that in SD rats at time points of 3-, 9-, and 12-month (p < 0.001). Vascular elasticity decreased both in SHR and SD rats with aging. There were statistically significant differences in the relative vascular elasticity of extracranial/intracranial internal carotid artery, middle cerebral artery, posterior cerebral artery and anterior cerebral artery between SHR and SD rats at 12-month (p < 0.01). We concluded that the dynamic vascular alterations detected by SR angiography provided novel imaging data for the study of hypertension in vivo. The longer the course of hypertension was, the more obvious the vascular differences between the SHR and the SD rats became.
Collapse
Affiliation(s)
- Liping Wang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhihao Mu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Xiaojie Lin
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jieli Geng
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ti Qiao Xiao
- Shanghai Synchrotron Radiation Facility, Shanghai Institute of Applied Physics, CAS, Shanghai, China
| | - Zhijun Zhang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongting Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongjing Guan
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Tang Y, Yuan F, Cai B, Xia W, Wang Y, Yang GY. Effect of ischaemic brain injury on sexual function in adult mice. Stroke Vasc Neurol 2017; 1:127-132. [PMID: 28959474 PMCID: PMC5435199 DOI: 10.1136/svn-2016-000013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 01/09/2023] Open
Abstract
Objective Priapism refers to a condition with persistent abnormal erection of the penis, which is usually caused by disease or injury in the brain or spinal cord, or obstruction to the outflow of blood through the dorsal vein at the root of the penis, without sexual desires. The effect of cerebral ischaemia on sexual function is unknown. The aim of this study is to explore whether priapism occurs in adult mice. Furthermore, we examined the relationship between priapism and the region of infarct in the brain. Design Adult male CD-1 mice who underwent permanent middle cerebral artery occlusion (pMCAO) were closely examined from 2 hours to 14 days postoperation. Results We found that priapism occurs in ∼80% of the mice with pMCAO, which could persist up to 14 days. Further study has demonstrated that the occurrence of priapism is related to the infarct region: priapism is found only in mice with ischaemic injury extending to the hypothalamus and the hippocampus regions. Conclusion Our result suggested priapism may be used as a deep brain injury marker for evaluating brain injury in mice after pMCAO.
Collapse
Affiliation(s)
- Yaohui Tang
- Neuroscience and Neuroengineering Center, Med-X Research Institute Shanghai Jiao Tong University, Shanghai, China
| | - Falei Yuan
- Neuroscience and Neuroengineering Center, Med-X Research Institute Shanghai Jiao Tong University, Shanghai, China
| | - Beibei Cai
- Neuroscience and Neuroengineering Center, Med-X Research Institute Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- Neuroscience and Neuroengineering Center, Med-X Research Institute Shanghai Jiao Tong University, Shanghai, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Center, Med-X Research Institute Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute Shanghai Jiao Tong University, Shanghai, China.,Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Guo F, Lou J, Han X, Deng Y, Huang X. Repetitive Transcranial Magnetic Stimulation Ameliorates Cognitive Impairment by Enhancing Neurogenesis and Suppressing Apoptosis in the Hippocampus in Rats with Ischemic Stroke. Front Physiol 2017; 8:559. [PMID: 28824455 PMCID: PMC5539749 DOI: 10.3389/fphys.2017.00559] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/17/2017] [Indexed: 01/03/2023] Open
Abstract
Cognitive impairment is a serious mental deficit caused by stroke that can severely affect the quality of a survivor's life. Repetitive transcranial magnetic stimulation (rTMS) is a well-known rehabilitation modality that has been reported to exert neuroprotective effects after cerebral ischemic injury. In the present study, we evaluated the therapeutic efficacy of rTMS against post-stroke cognitive impairment (PSCI) and investigated the mechanisms underlying its effects in a middle cerebral artery occlusion (MCAO) rat model. The results showed that rTMS ameliorated cognitive deficits and tended to reduce the sizes of cerebral lesions. In addition, rTMS significantly improved cognitive function via a mechanism involving increased neurogenesis and decreased apoptosis in the ipsilateral hippocampus. Moreover, brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), were clearly upregulated in ischemic hippocampi after treatment with rTMS. Additionally, further studies demonstrated that rTMS markedly enhanced the expression of the apoptosis-related B cell lymphoma/leukemia gene 2 (Bcl-2) and decreased the expression of the Bcl-2-associated protein X (Bax) and the number of TUNEL-positive cells in the ischemic hippocampus. Both protein levels and mRNA levels were investigated. Our findings suggest that after ischemic stroke, treatment with rTMS promoted the functional recovery of cognitive impairments by inhibiting apoptosis and enhancing neurogenesis in the hippocampus and that this mechanism might be mediated by the BDNF signaling pathway.
Collapse
Affiliation(s)
- Feng Guo
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Jicheng Lou
- Department of Obstetrics and Gynecology, The Central Hospital of WuhanWuhan, China
| | - Xiaohua Han
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Yuguo Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Xiaolin Huang
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| |
Collapse
|
20
|
Mangas A, Yajeya J, González N, Ruiz I, Pernìa M, Geffard M, Coveñas R. Gemst: a taylor-made combination that reverts neuroanatomical changes in stroke. Eur J Histochem 2017; 61:2790. [PMID: 28735520 PMCID: PMC5452634 DOI: 10.4081/ejh.2017.2790] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 01/25/2023] Open
Abstract
In a single transient middle cerebral artery occlusion model of stroke and using immunohistochemical techniques, the effects of a new therapeutic approach named Gemst (a member of the Poly-L-Lysine innovative therapies) have been studied in the rat brain. The expression of inflammatory (CD45, CD11b), oxidative (NO-tryptophan, NO2-tyrosine) and indoleamine 2, 3-dioxygenase pathway (kynurenic acid, 3-hydroxy anthranilic acid) markers has been evaluated in early and late phases of stroke. For this purpose, we have developed eight highly specific monoclonal antibodies directed against some of these markers. In the early phase (3 and 5 days of the stroke, we observed no effect of Gemst treatment (7.5 mg/day, subcutaneously for 3, 5 days). In the late phase (21 days) of stroke and exclusively in the ipsilateral side of non-treated animals an overexpression of kynurenic acid, 3-hydroxy anthranilic acid, CD45, CD11b, GFAP and ionized calcium-binding adapter molecule 1 (IBA-1) was found. In treated animals, the overexpression of the four former markers was completely abolished whereas the overexpression of the two latter ones was decreased down to normal levels. Gemst reversed the pathological conditions of stroke to normal situations. Gemst exerts a multifunctional action: down-regulates the indoleamine 2, 3-dioxygenase pathway and abolishes brain infiltration, microglial activation and gliosis. Moreover, Gemst has no effect on the expression of doublecortin, a protein involved in neuronal migration. Gemst could be a new drug for the treatment of stroke since it reverses the pathological findings of stroke and normalizes brain tissue conditions following the ischemic insult.
Collapse
|
21
|
Mangas A, Yajeya J, González N, Ruiz I, Duleu S, Geffard M, Coveñas R. Overexpression of kynurenic acid in stroke: An endogenous neuroprotector? Ann Anat 2017; 211:33-38. [PMID: 28163204 DOI: 10.1016/j.aanat.2017.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/08/2016] [Accepted: 01/04/2017] [Indexed: 10/20/2022]
Abstract
It is known that kynurenic acid (KYNA) exerts a neuroprotective effect against the neuronal loss induced by ischemia; acting as a scavenger, and exerting antioxidant action. In order to study the distribution of KYNA, a highly specific monoclonal antibody directed against KYNA was developed. This distribution was studied in control rats and in animals in which a middle cerebral artery occlusion (stroke model) was induced. By double immunohistochemistry, astrocytes containing KYNA and GFAP were exclusively found in the ipsilateral cerebral cortex and/or striatum, at 2, 5 and 21days after the induction of stroke. In control animals and in the contralateral side of the stroke animals, no immunoreactivity for KYNA was found. Under pathological conditions, the presence of KYNA is reported for the first time in the mammalian brain from early phases of stroke. The distribution of KYNA matches perfectly with the infarcted regions suggesting that, in stroke, this overexpressed molecule could be involved in neuroprotective/scavenger/antioxidant mechanisms.
Collapse
Affiliation(s)
- A Mangas
- Gemacbio, Saint Jean d'Illac, France; Institut pour le Développement de la Recherche en Pathologie Humaine et Thérapeutique (IDRPHT), Talence, France; Institute of Neurosciences of Castilla y León (INCYL), Laboratory of Neuroanatomy of the Peptidergic Systems, University of Salamanca, Salamanca, Spain.
| | - J Yajeya
- School of Medicine, Department of Physiology, University of Salamanca, Salamanca, Spain
| | - N González
- Gemacbio, Saint Jean d'Illac, France; Institut pour le Développement de la Recherche en Pathologie Humaine et Thérapeutique (IDRPHT), Talence, France
| | - I Ruiz
- Gemacbio, Saint Jean d'Illac, France
| | - S Duleu
- Institut pour le Développement de la Recherche en Pathologie Humaine et Thérapeutique (IDRPHT), Talence, France
| | - M Geffard
- Gemacbio, Saint Jean d'Illac, France; Institut pour le Développement de la Recherche en Pathologie Humaine et Thérapeutique (IDRPHT), Talence, France
| | - R Coveñas
- Institute of Neurosciences of Castilla y León (INCYL), Laboratory of Neuroanatomy of the Peptidergic Systems, University of Salamanca, Salamanca, Spain
| |
Collapse
|
22
|
Mangas A, Yajeya J, González N, Ruiz I, Geffard M, Coveñas R. 3-hydroxi-anthranilic acid is early expressed in stroke. Eur J Histochem 2016; 60:2709. [PMID: 28076933 PMCID: PMC5159783 DOI: 10.4081/ejh.2016.2709] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/22/2016] [Accepted: 11/27/2016] [Indexed: 12/14/2022] Open
Abstract
Using an immunohistochemical technique, we have studied the distribution of 3-OH-anthranilic acid (3-HAA) in the rat brain. Our study was carried out in control animals and in rats in which a stroke model (single transient middle cerebral artery occlusion) was performed. A monoclonal antibody directed against 3-HAA was also developed. 3-HAA was exclusively observed in the infarcted regions (ipsilateral striatum/cerebral cortex), 2, 5 and 21 days after the induction of stroke. In control rats and in the contralateral side of the stroke animals, no immunoreactivity for 3-HAA was visualized. Under pathological conditions (from early phases of stroke), we reported for the first time the presence of 3-HAA in the mammalian brain. By double immunohistochemistry, the coexistence of 3-HAA and GFAP was observed in astrocytes. The distribution of 3-HAA matched perfectly with the infarcted regions. Our findings suggest that, in stroke, 3-HAA could be involved in the tissue damage observed in the infarcted regions, since it is well known that 3-HAA exerts cytotoxic effects.
Collapse
Affiliation(s)
- A Mangas
- Gemacbio - Institute for the Development of Research in Human Pathology and Therapeutic (IDRPHT) - University of Salamanca.
| | | | | | | | | | | |
Collapse
|
23
|
Systematic review of survival time in experimental mouse stroke with impact on reliability of infarct estimation. J Neurosci Methods 2016; 261:10-8. [PMID: 26620203 DOI: 10.1016/j.jneumeth.2015.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/24/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Stroke is the second most common cause of death worldwide. Only one treatment for acute ischemic stroke is currently available, thrombolysis with rt-PA, but it is limited in its use. Many efforts have been invested in order to find additive treatments, without success. A multitude of reasons for the translational problems from mouse experimental stroke to clinical trials probably exists, including infarct size estimations around the peak time of edema formation. Furthermore, edema is a more prominent feature of stroke in mice than in humans, because of the tendency to produce larger infarcts with more substantial edema. PURPOSE This paper will give an overview of previous studies of experimental mouse stroke, and correlate survival time to peak time of edema formation. Furthermore, investigations of whether the included studies corrected the infarct measurements for edema and a comparison of correction methods will be discussed. METHOD Relevant terms were searched in the National Library of Medicine PubMed database. A method for classification of infarct measurement methods was made using a naming convention. CONCLUSION Our study shows that infarct size estimations are often performed around the peak time of edema, with a median of 24h. Most studies do consider edema formation, however, there is no consensus on what method to use to correct for edema. Furthermore, investigations into neuroprotective drugs should use longer survival times to ensure completion of the investigated process. Our findings indicate a need for more research in this area, and establishment of common correction methodology.
Collapse
|
24
|
Mu ZH, Jiang Z, Lin XJ, Wang LP, Xi Y, Zhang ZJ, Wang YT, Yang GY. Vessel Dilation Attenuates Endothelial Dysfunction Following Middle Cerebral Artery Occlusion in Hyperglycemic Rats. CNS Neurosci Ther 2016; 22:316-24. [PMID: 26842484 DOI: 10.1111/cns.12500] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES Dynamically observe cerebral vascular changes in hyperglycemic rats in vivo and explore the effect of diabetes on endothelial function after ischemic stroke. BACKGROUND Diabetes affects both large and small vessels in the brain, but the dynamic process and mechanism are unclear. METHODS We investigated the structural and functional changes of brain vasculature in living hyperglycemic rats and their impact on stroke outcomes via a novel technique: synchrotron radiation angiography. We also examined the effect of prolonged fasudil treatment on arterial reactivity and hemorrhagic transformation. Adult Sprague Dawley rats were treated by streptozotocin to induce type 1 diabetes. These hyperglycemic rats received fasudil pretreatment and then underwent transient middle cerebral artery occlusion. RESULTS We found that diabetes caused arteries narrowing in the circus Willis as early as 2 weeks after streptozotocin injection (P < 0.05). These vessels were further constricted after middle cerebral artery occlusion. L-NAME could induce regional constrictions and impaired relaxation in hyperglycemic animals. Furthermore, hemorrhagic transformation was also increased in the hyperglycemic rats compared to the control (P < 0.05). In fasudil-treated rats, the internal carotid artery narrowing was ameliorated and L-NAME-induced regional constriction was abolished. Importantly, stroke prognosis was improved in fasudil-treated rats compared to the control (P < 0.05). CONCLUSIONS Our dynamic angiographic data demonstrated that diabetes could impair the cerebral arterial reactivity. Prolonged fasudil treatment could attenuate arterial dysfunction and improve the prognosis of ischemic stroke by affecting both the large and small vasculature.
Collapse
Affiliation(s)
- Zhi-Hao Mu
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Jiang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Jie Lin
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Ping Wang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Xi
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Center for Biotechnology and Interdisciplinary Studies, Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Zhi-Jun Zhang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Ting Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
Lin H, Jing J, Lu YF, Xie C, Lin XJ, Yang GY. Effect of iodine contrast agent concentration on cerebrovascular dose for synchrotron radiation microangiography based on a simple mouse head model and a voxel mouse head phantom by Monte Carlo simulation. JOURNAL OF SYNCHROTRON RADIATION 2016; 23:304-311. [PMID: 26698078 DOI: 10.1107/s1600577515019025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/09/2015] [Indexed: 06/05/2023]
Abstract
Effective setting strategies using Monte Carlo simulation are presented to mitigate the irradiation damage in synchrotron radiation microangiography (SRA). A one-dimensional mouse head model and a segmented voxel phantom mouse head were simulated using the EGSnrc/DOSXYZnrc code to investigate the dose enhancement effect of an iodine contrast agent irradiated by a monochromatic synchrotron radiation source. The influence of the iodine concentration, vessel width and depth, protection with and without the skull layer, and various incident X-ray energies were all simulated. The dose enhancement effect and the absolute dose based on the segmented voxel mouse head phantom were evaluated. The dose enhancement ratio depended little on the irradiation depth, but strongly and linearly increasing on iodine concentration. The protection given by the skull layer cannot be ignored in SRA because a 700 µm-thick skull can decrease the dose by 10%. The incident X-ray energy can affect the dose significantly. Compared with a dose of 33.2 keV for 50 mgI ml(-1), a dose of 32.7 keV decreased by 38%, whereas a dose of 33.7 keV increased by 69.2% and the variation strengthened more with enhanced iodine concentration. The segmented voxel mouse head phantom also showed that the average dose enhancement effect and the maximal voxel dose per photon depended little on the iodine voxel volume ratio but strongly on the iodine concentration. To decrease the damage caused by the dose in SRA, a high-Z contrast agent should be used as little as possible and irradiation of the injection site of the contrast agent should be avoided immediately after the injection. The fragile vessel containing iodine should avoid being closely irradiated. Avoiding irradiating through a thin (or no) skull region, or attaching a thin equivalent material on the outside for protection are better methods. An incident X-ray energy as low as possible should be used as long as the SRA image quality is ensured. The use of the synergetic and synchronous shuttering technique in SRA is also very critical in order to effectively shorten the accumulative irradiation time in in vivo animal irradiation experiments.
Collapse
Affiliation(s)
- Hui Lin
- School of Electronic Science and Application Physics, Hefei University of Technology, 230009 Hefei, People's Republic of China
| | - Jia Jing
- School of Electronic Science and Application Physics, Hefei University of Technology, 230009 Hefei, People's Republic of China
| | - Yi-Fan Lu
- Neuroscience and Neuroengineering Centre, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Cong Xie
- School of Electronic Science and Application Physics, Hefei University of Technology, 230009 Hefei, People's Republic of China
| | - Xiao-Jie Lin
- Neuroscience and Neuroengineering Centre, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Centre, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| |
Collapse
|
26
|
El Amki M, Clavier T, Perzo N, Bernard R, Guichet PO, Castel H. Hypothalamic, thalamic and hippocampal lesions in the mouse MCAO model: Potential involvement of deep cerebral arteries? J Neurosci Methods 2015. [PMID: 26213218 DOI: 10.1016/j.jneumeth.2015.07.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Intraluminal monofilament occlusion of the middle cerebral artery (MCAO) in mice is the most used rodent model to study the pathophysiology of stroke. However, this model often shows brain damage in regions not supplied by the MCA such as the hypothalamus, hippocampus and thalamus. Several studies have suggested some explanations on these localized infarcts. We aim to provide an alternative explanation which could allow each experimenter to better grasp the MCAO model. We propose that the MCA occlusion by the monofilament also occludes deep and small cerebral arteries arising directly from the internal carotid artery, proximally to the origin of MCA. Then, drawbacks and pitfalls of the MCAO model must be appreciated and the almost systematic risk of inducing lesions in some unwanted territories for neuroanatomical reasons, i.e. vascular connections between deep arteries and hypothalamic, thalamic and hippocampal areas in rodents has to be integrated.
Collapse
Affiliation(s)
- Mohamad El Amki
- Institut National de la Santé et de la Recherche Médicale (Inserm), U982, Rouen University, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Mont-Saint-Aignan, France.
| | - Thomas Clavier
- Institut National de la Santé et de la Recherche Médicale (Inserm), U982, Rouen University, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Mont-Saint-Aignan, France; Department of Anesthesiology and Critical Care, Rouen University Hospital, Rouen, France
| | - Nicolas Perzo
- Institut National de la Santé et de la Recherche Médicale (Inserm), U982, Rouen University, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Mont-Saint-Aignan, France
| | - René Bernard
- Department of Experimental Neurology, Charité University Medicine, Berlin, Germany
| | - Pierre-Olivier Guichet
- Institut National de la Santé et de la Recherche Médicale (Inserm), U982, Rouen University, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Mont-Saint-Aignan, France
| | - Hélène Castel
- Institut National de la Santé et de la Recherche Médicale (Inserm), U982, Rouen University, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Mont-Saint-Aignan, France
| |
Collapse
|
27
|
Poinsatte K, Selvaraj UM, Ortega SB, Plautz EJ, Kong X, Gidday JM, Stowe AM. Quantification of neurovascular protection following repetitive hypoxic preconditioning and transient middle cerebral artery occlusion in mice. J Vis Exp 2015:e52675. [PMID: 25993394 DOI: 10.3791/52675] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Experimental animal models of stroke are invaluable tools for understanding stroke pathology and developing more effective treatment strategies. A 2 week protocol for repetitive hypoxic preconditioning (RHP) induces long-term protection against central nervous system (CNS) injury in a mouse model of focal ischemic stroke. RHP consists of 9 stochastic exposures to hypoxia that vary in both duration (2 or 4 hr) and intensity (8% and 11% O2). RHP reduces infarct volumes, blood-brain barrier (BBB) disruption, and the post-stroke inflammatory response for weeks following the last exposure to hypoxia, suggesting a long-term induction of an endogenous CNS-protective phenotype. The methodology for the dual quantification of infarct volume and BBB disruption is effective in assessing neurovascular protection in mice with RHP or other putative neuroprotectants. Adult male Swiss Webster mice were preconditioned by RHP or duration-equivalent exposures to 21% O2 (i.e. room air). A 60 min transient middle cerebral artery occlusion (tMCAo) was induced 2 weeks following the last hypoxic exposure. Both the occlusion and reperfusion were confirmed by transcranial laser Doppler flowmetry. Twenty-two hr after reperfusion, Evans Blue (EB) was intravenously administered through a tail vein injection. 2 hr later, animals were sacrificed by isoflurane overdose and brain sections were stained with 2,3,5- triphenyltetrazolium chloride (TTC). Infarcts volumes were then quantified. Next, EB was extracted from the tissue over 48 hr to determine BBB disruption after tMCAo. In summary, RHP is a simple protocol that can be replicated, with minimal cost, to induce long-term endogenous neurovascular protection from stroke injury in mice, with the translational potential for other CNS-based and systemic pro-inflammatory disease states.
Collapse
Affiliation(s)
- Katherine Poinsatte
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center
| | - Uma Maheswari Selvaraj
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center
| | - Sterling B Ortega
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center
| | - Erik J Plautz
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center
| | - Xiangmei Kong
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center
| | - Jeffrey M Gidday
- Department of Neurological Surgery, Washington University School of Medicine
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center;
| |
Collapse
|
28
|
Lin X, Miao P, Mu Z, Jiang Z, Lu Y, Guan Y, Chen X, Xiao T, Wang Y, Yang GY. Development of functional in vivo imaging of cerebral lenticulostriate artery using novel synchrotron radiation angiography. Phys Med Biol 2015; 60:1655-65. [PMID: 25632958 DOI: 10.1088/0031-9155/60/4/1655] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lenticulostriate artery plays a vital role in the onset and development of cerebral ischemia. However, current imaging techniques cannot assess the in vivo functioning of small arteries such as the lenticulostriate artery in the brain of rats. Here, we report a novel method to achieve a high resolution multi-functional imaging of the cerebrovascular system using synchrotron radiation angiography, which is based on spatio-temporal analysis of contrast density in the arterial cross section. This method provides a unique tool for studying the sub-cortical vascular elasticity after cerebral ischemia in rats. Using this technique, we demonstrated that the vascular elasticity of the lenticulostriate artery decreased from day 1 to day 7 after transient middle cerebral artery occlusion in rats and recovered from day 7 to day 28 compared to the controls (p < 0.001), which paralleled with brain edema formation and inversely correlated with blood flow velocity (p < 0.05). Our results demonstrated that the change of vascular elasticity was related to the levels of brain edema and the velocity of focal blood flow, suggesting that reducing brain edema is important for the improvement of the function of the lenticulostriate artery in the ischemic brain.
Collapse
Affiliation(s)
- Xiaojie Lin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yuan F, Lin X, Guan Y, Mu Z, Chen K, Wang Y, Yang GY. Collateral circulation prevents masticatory muscle impairment in rat middle cerebral artery occlusion model. JOURNAL OF SYNCHROTRON RADIATION 2014; 21:1314-1318. [PMID: 25343800 DOI: 10.1107/s1600577514016130] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/11/2014] [Indexed: 06/04/2023]
Abstract
The rat suture middle cerebral artery occlusion (MCAO) is a frequently used animal model for investigating the mechanisms of ischemic brain injury. During suture MCAO, transection of the external carotid artery (ECA) potentially restrains blood flow and impairs masticatory muscle and other ECA-supported territories, consequently influencing post-operation animal survival. This study was aimed at investigating the effect of ECA transection on the hemodynamic alterations using a novel synchrotron radiation (SR) angiography technique and magnetic resonance imaging in live animals. Fifteen male adult Sprague-Dawley rats were used in this study. Animals underwent MCAO, in which the ECA was transected. SR angiography was performed before and after MCAO. Rats then underwent magnetic resonance imaging (MRI) to detect the tissue lesion both intra- and extra-cranially. Animals with SR angiography without other manipulations were used as control. High-resolution cerebrovascular morphology was analyzed using a novel technique of SR angiography. The masticatory muscle lesion was further examined by hematoxylin and eosin staining. MRI and histological results showed that there was no masticatory muscle lesion at 1, 7 and 28 days following MCAO with ECA transection. In normal condition, the ECA and its branch external maxillary artery were clearly detected. Following ECA transection, the external maxillary artery was still observed and the blood supply appeared from the anastomotic branch from the pterygopalatine artery. SR angiography further revealed the inter-relationship of hemisphere extra- and intra-cranial vasculature in the rat following MCAO. Transection of the ECA did not impair masticatory muscles in rat suture MCAO. Interrupted blood flow could be compensated by the collateral circulation from the pterygopalatine artery.
Collapse
Affiliation(s)
- Falei Yuan
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Xiaojie Lin
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Yongjing Guan
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, People's Republic of China
| | - Zhihao Mu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Kemin Chen
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, People's Republic of China
| | - Yongting Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Guo-Yuan Yang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| |
Collapse
|
30
|
Guo F, Han X, Zhang J, Zhao X, Lou J, Chen H, Huang X. Repetitive transcranial magnetic stimulation promotes neural stem cell proliferation via the regulation of MiR-25 in a rat model of focal cerebral ischemia. PLoS One 2014; 9:e109267. [PMID: 25302788 PMCID: PMC4193773 DOI: 10.1371/journal.pone.0109267] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 09/08/2014] [Indexed: 01/03/2023] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) has increasingly been studied over the past decade to determine whether it has a therapeutic benefit on focal cerebral ischemia. However, the underlying mechanism of rTMS in this process remains unclear. In the current study, we investigated the effects of rTMS on the proliferation of adult neural stem cells (NSCs) and explored microRNAs (miRNAs) that were affected by rTMS. Our data showed that 10 Hz rTMS significantly increased the proliferation of adult NSCs after focal cerebral ischemia in the subventricular zone (SVZ), and the expression of miR-25 was obviously up-regulated in the ischemic cortex after rTMS. p57, an identified miR-25 target gene that regulates factors linked to NSC proliferation, was also evaluated, and it exhibited down-regulation. To further verify the role of miR-25, rats were injected with a single dose of antagomir-25 and were subjected to focal cerebral ischemia followed by rTMS treatment. The results confirmed that miR-25 could be repressed specifically and could drive the up-regulation of its target gene (p57), which resulted in the inhibition of adult NSC proliferation in the SVZ after rTMS. Thus, our studies strongly indicated that 10 Hz rTMS can promote the proliferation of adult NSCs in the SVZ after focal cerebral ischemia by regulating the miR-25/p57 pathway.
Collapse
Affiliation(s)
- Feng Guo
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Han
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghui Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiuxiu Zhao
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jicheng Lou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolin Huang
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| |
Collapse
|