1
|
Zhang L, Zhang C, Chen R, Zhang J, Liu Y, Du Y, Gao X, Shang W, Xu R, Zhang X. Dl-3-n-Butylphthalide Promotes Neurogenesis in Ischemic Stroke Mice Through Wnt/β-Catenin Signaling Activation and Neurotrophic Factor Production. Mol Neurobiol 2025:10.1007/s12035-025-04884-8. [PMID: 40172819 DOI: 10.1007/s12035-025-04884-8] [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: 09/10/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
Synchronized neurogenesis and angiogenesis after stroke have been well documented, and inducing neurovascular remodeling may provide a promising strategy to promote tissue repair and functional recovery. Dl-3-n-Butylphthalide (NBP) was reported to exert a potent angiogenic activity in rodent models of stroke. However, little is currently known regarding the effects and mechanisms of NBP on neurogenesis in ischemic stroke. This study aimed to determine whether and how NBP promotes neurogenesis in cerebral ischemic injury. Adult C57BL/6 mice, subjected to distal middle cerebral artery occlusion (dMCAO), were treated with NBP. The efficacy of NBP was assessed using neurologic deficits and infarct volume. Immunofluorescent staining was applied to evaluate neurogenesis. The regulation of the Wnt/β-catenin signaling pathway and the expression of neurotrophic factors were detected by western blotting and qRT-PCR. Administration of NBP reduced infarct volume and ameliorated neurological deficits after stroke. NBP promoted the proliferation of NSCs in the SVZ, migration of neuroblasts along the corpus callosum, and differentiation of neuroblasts toward neurons in the peri-infarct zone, resulting in restored neural function. Moreover, we revealed that NBP-induced neurogenesis was associated with the activation of the Wnt/β-catenin pathway, which was reversed by DKK1. In addition, NBP increased the production of VEGF and BDNF. Our data have unveiled the potentials of NBP to promote neurogenesis and neural functional recovery after stroke, depending on Wnt/β-catenin signaling activation and neurotrophic factor production. Thus, NBP may be a promising candidate for delayed treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
| | - Rong Chen
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
| | - Jian Zhang
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China
| | - Ying Liu
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
| | - Yuanyuan Du
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
| | - Xuan Gao
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
- Department of Geriatrics, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China
| | - Wenyan Shang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, P.R. China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
| | - Renhao Xu
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, P.R. China.
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, 050000, P.R. China.
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, 050000, P.R. China.
| |
Collapse
|
2
|
Zhang Y, Liu Z, Chopp M, Millman M, Li Y, Cepparulo P, Kemper A, Li C, Zhang L, Zhang ZG. Small extracellular vesicles derived from cerebral endothelial cells with elevated microRNA 27a promote ischemic stroke recovery. Neural Regen Res 2025; 20:224-233. [PMID: 38767487 PMCID: PMC11246145 DOI: 10.4103/nrr.nrr-d-22-01292] [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: 12/07/2022] [Revised: 06/14/2023] [Accepted: 01/22/2024] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00030/figure1/v/2024-05-14T021156Z/r/image-tiff Axonal remodeling is a critical aspect of ischemic brain repair processes and contributes to spontaneous functional recovery. Our previous in vitro study demonstrated that exosomes/small extracellular vesicles (sEVs) isolated from cerebral endothelial cells (CEC-sEVs) of ischemic brain promote axonal growth of embryonic cortical neurons and that microRNA 27a (miR-27a) is an elevated miRNA in ischemic CEC-sEVs. In the present study, we investigated whether normal CEC-sEVs engineered to enrich their levels of miR-27a (27a-sEVs) further enhance axonal growth and improve neurological outcomes after ischemic stroke when compared with treatment with non-engineered CEC-sEVs. 27a-sEVs were isolated from the conditioned medium of healthy mouse CECs transfected with a lentiviral miR-27a expression vector. Small EVs isolated from CECs transfected with a scramble vector (Scra-sEVs) were used as a control. Adult male mice were subjected to permanent middle cerebral artery occlusion and then were randomly treated with 27a-sEVs or Scra-sEVs. An array of behavior assays was used to measure neurological function. Compared with treatment of ischemic stroke with Scra-sEVs, treatment with 27a-sEVs significantly augmented axons and spines in the peri-infarct zone and in the corticospinal tract of the spinal grey matter of the denervated side, and significantly improved neurological outcomes. In vitro studies demonstrated that CEC-sEVs carrying reduced miR-27a abolished 27a-sEV-augmented axonal growth. Ultrastructural analysis revealed that 27a-sEVs systemically administered preferentially localized to the pre-synaptic active zone, while quantitative reverse transcription-polymerase chain reaction and Western Blot analysis showed elevated miR-27a, and reduced axonal inhibitory proteins Semaphorin 6A and Ras Homolog Family Member A in the peri-infarct zone. Blockage of the Clathrin-dependent endocytosis pathway substantially reduced neuronal internalization of 27a-sEVs. Our data provide evidence that 27a-sEVs have a therapeutic effect on stroke recovery by promoting axonal remodeling and improving neurological outcomes. Our findings also suggest that suppression of axonal inhibitory proteins such as Semaphorin 6A may contribute to the beneficial effect of 27a-sEVs on axonal remodeling.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | - Michael Millman
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Yanfeng Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | | - Amy Kemper
- Department of Pathology, Henry Ford Hospital, Detroit, MI, USA
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | |
Collapse
|
3
|
Chen Y, Xu Z, Ma Y, Liu T, Tian X, Zhu Z, Zheng W, Wang Y, Zheng R, Xing J, Wang W, Sun F. Deep brain stimulation combined with morroniside promotes neural plasticity and motor functional recovery after ischemic stroke. Front Pharmacol 2024; 15:1457309. [PMID: 39697542 PMCID: PMC11652210 DOI: 10.3389/fphar.2024.1457309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
Background and Objective Until now, there has been an unmet need for treatments promoting chronic-phase post-stroke functional recovery. We previously found that morroniside promoted endogenous neurogenesis in ischemic stroke, but its therapeutic window was limited to the first 48 h. Here, we aimed to explore whether deep brain stimulation (DBS) combined with morroniside could enhance neurogenesis in rats subjected to focal ischemic stroke and contributes to functional recovery. Methods Beginning 2 weeks after the endothelin-1-induced stroke, rats were administered DBS of lateral cerebellar nucleus consecutively for 14 days, followed by morroniside for 7 consecutive days post-stimulation. Behavioral tests were used for assessing motor function. Local field potentials were recorded to evaluate neuronal excitability. Nissl staining was used to assess infarct volume. Immunofluorescence staining and Western blotting were carried out to uncover the stroke recovery mechanisms of DBS combined with morroniside treatment. Results The results showed that this combined treatment improved behavioral outcomes, enhanced cortical local field potentials, and diminished infarct volumes at 35 days post-stroke. Moreover, it notably amplified neurogenic responses post-stroke, evidenced by the proliferation of BrdU/SOX2 and BrdU/DCX in the subventricular zone, and their subsequent differentiation into BrdU/NeuN and BrdU/VgulT1 in the ischemic penumbra. Moreover, the combined treatment also elevated the amount of BrdU/Olig2 and the level of axonal sprouting-related proteins in the perilesional cortex. Conclusion Our results demonstrated that the combined treatment extended the neurorestorative efficacy of morroniside, reduced infarct size, enhanced neuronal excitability and accelerated sensorimotor function recovery. This therapeutic approach may emerge as a potential clinical intervention for chronic ischemic stroke.
Collapse
Affiliation(s)
- Yanxi Chen
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Zhidong Xu
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yifu Ma
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Tingting Liu
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Xin Tian
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Zixin Zhu
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Wenrong Zheng
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Yufeng Wang
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Ruifang Zheng
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Jianguo Xing
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi, China
| | - Wen Wang
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Fangling Sun
- Department of Experimental Animal Center, Xuanwu Hospital of Capital Medical University, Beijing Municipal Geriatric Medical Research Center, Beijing, China
| |
Collapse
|
4
|
Bai S, Zhang G, Chen S, Wu X, Li J, Wang J, Chen D, Liu X, Wang J, Li Y, Tang Y, Tang Z. MicroRNA-451 Regulates Angiogenesis in Intracerebral Hemorrhage by Targeting Macrophage Migration Inhibitory Factor. Mol Neurobiol 2024; 61:10481-10499. [PMID: 38743209 PMCID: PMC11584486 DOI: 10.1007/s12035-024-04207-3] [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: 05/31/2023] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke with the highest fatality and disability rate. Up to now, commonly used first-line therapies have limited value in improving prognosis. Angiogenesis is essential to neurological recovery after ICH. Recent studies have shown that microRNA-451(miR-451) plays an important role in angiogenesis by regulating the function of vascular endothelial cells. We found miR-451 was significantly decreased in the peripheral blood of ICH patients in the acute stage. Based on the clinical findings, we conducted this study to investigate the potential regulatory effect of miR-451 on angiogenesis after ICH. The expression of miR-451 in ICH mouse model and in a hemin toxicity model of human brain microvascular endothelial cells (hBMECs) was decreased the same as in ICH patients. MiR-451 negatively regulated the proliferation, migration, and tube formation of hBMECs in vitro. MiR-451 negatively regulated the microvessel density in the perihematoma tissue and affected neural functional recovery of ICH mouse model. Knockdown of miR-451 could recovered tight junction and protect the integrity of blood-brain barrier after ICH. Based on bioinformatic programs, macrophage migration inhibitory factor (MIF) was predicted to be the target gene and identified to be regulated by miR-451 inhibiting the protein translation. And p-AKT and p-ERK were verified to be downstream of MIF in angiogenesis. These results all suggest that miR-451 will be a potential target for regulating angiogenesis in ICH.
Collapse
Affiliation(s)
- Shuang Bai
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ge Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingxuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Li J, Song J, Jia L, Wang M, Ji X, Meng R, Zhou D. Exosomes in Central Nervous System Diseases: A Comprehensive Review of Emerging Research and Clinical Frontiers. Biomolecules 2024; 14:1519. [PMID: 39766226 PMCID: PMC11673277 DOI: 10.3390/biom14121519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Exosomes, nano-sized lipid bilayer vesicles, have garnered significant attention as mediators of cell communication, particularly within the central nervous system (CNS). Their unique properties, including high stability, low immunogenicity, and the ability to traverse the blood-brain barrier (BBB), position them as promising tools for understanding and addressing CNS diseases. This comprehensive review delves into the biogenesis, properties, composition, functions, and isolation of exosomes, with a particular focus on their roles in cerebrovascular diseases, neurodegenerative disorders, and CNS tumors. Exosomes are involved in key pathophysiological processes in the CNS, including angiogenesis, inflammation, apoptosis, and cellular microenvironment modification. They demonstrate promise in mitigating ischemic injury, regulating inflammatory responses, and providing neuroprotection across various CNS conditions. Furthermore, exosomes carry distinct biomolecules, offering a novel method for the early diagnosis and monitoring of CNS diseases. Despite their potential, challenges such as complex extraction processes, the heterogeneity of exosomal contents, and targeted delivery limitations hinder their clinical application. Nevertheless, exosomes hold significant promise for advancing our understanding of CNS diseases and developing novel therapeutic strategies. This manuscript significantly contributes to the field by highlighting exosomes' potential in advancing our understanding of CNS diseases, underscoring their unique value in developing novel therapeutic strategies and mediating cellular communication.
Collapse
Affiliation(s)
- Jingrun Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jiahao Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lina Jia
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Mengqi Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
6
|
Wu C, Li Y, He X, Sun H, Zhang S, Hou F, Hu M, Lan A, Zhang H, Qi L, Zhang H, Liao H. Chemogenetic activation of astrocytic Gi signaling promotes spinogenesis and motor functional recovery after stroke. Glia 2024; 72:1150-1164. [PMID: 38436489 DOI: 10.1002/glia.24521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
Ischemic stroke is the leading cause of adult disability. The rewiring of surviving neurons is the fundamental process for functional recovery. Accumulating evidence implicates astrocytes in synapses and neural circuits formation, but few studies have further studied how to enhance the effects of astrocytes on synapse and circuits after stroke and its impacts on post-stroke functional recovery. In this study, we made use of chemogenetics to specifically activate astrocytic Gi signaling in the peri-infarcted sensorimotor cortex at different time epochs in a mouse model of photothrombotic stroke. We found that early activation of astrocytic hM4Di after stroke by CNO modulates astrocyte activity and upregulates synaptogenic molecules including thrombospondin-1 (TSP1) as revealed by bulk RNA-sequencing, but no significant improvement was observed in dendritic spine density and behavioral performance in grid walking test. Interestingly, when the manipulation was initiated at the subacute phase of stroke, the recovery of spine density and motor function could be effectively promoted, accompanied by increased TSP1 expression. Our data highlight the important role of astrocytes in synapse remodeling during the repair phase of stroke and suggest astrocytic Gi signaling activation as a potential strategy for synapse regeneration, circuit rewiring, and functional recovery.
Collapse
Affiliation(s)
- Chaoran Wu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yu Li
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xinran He
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hao Sun
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Shiwen Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Fengsheng Hou
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Mengqiu Hu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Aili Lan
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hao Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Long Qi
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Huibin Zhang
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Hong Liao
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
7
|
Rao J, Li H, Zhang H, Xiang X, Ding X, Li L, Geng F, Qi H. Periplaneta Americana (L.) extract activates the ERK/CREB/BDNF pathway to promote post-stroke neuroregeneration and recovery of neurological functions in rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117400. [PMID: 37952730 DOI: 10.1016/j.jep.2023.117400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Periplaneta americana (L.) (PA) has been used in traditional Chinese medicine for thousands of years for the effect of invigorating blood circulation and removing blood stasis. Modern pharmacological research shown that PA extract exhibits promising effects in promoting wound healing and regeneration, as well as in brain diseases such as Parkinson's disease (PD). However, whether it is effective for neuroregeneration and neurological function recovery after stroke still unknown. AIM OF THE STUDY This study aims to investigate the potential effect of PA extract to promote brain remodeling through the activation of endogenous neurogenesis and angiogenesis, in addition, preliminary exploration of its regulatory mechanism. METHODS Firstly, BrdU proliferation assay and immunofluorescence (IF) staining were used to evaluate the effect of PA extract on the neurogenesis and angiogenesis in vitro and in vivo. Subsequently, the effects of PA extract on brain injury in stroke rats were assessed by TTC and HE. While mNSS score, adhesive removal test, rota-rod test, and morris water maze test were used to assess the impact of PA extract on neurological function in post-stroke rats. Finally, the molecular mechanisms of PA extract regulation were explored by RNA-Seq and western blotting. RESULTS The number of BrdU+ cells in C17.2 cells, NSCs and BMECs dramatically increased, as well as the expression of astrocyte marker protein GFAP and neuronal marker protein Tuj-1 in C17.2 and NSCs. Moreover, PA extract also increased the number of BrdU+DCX+, BrdU+GFAP+, BrdU+CD31+ cells in the SGZ area of transient middle cerebral artery occlusion model (tMCAO) rats. TTC and HE staining revealed that PA extract significantly reduced the infarction volume and ameliorated the pathological damage. Behavioral tests demonstrated that treatment with PA extract reduced the mNSS score and the time required to remove adhesive tape, while increasing the time spent on the rotarod. Additionally, in the morris water maze test, the frequency of crossing platform and the time spent in the platform quadrant increased. Finally, RNA-Seq and Western blot revealed that PA extract increased the expression of p-ERK, p-CREB and BDNF. Importantly, PA extract mediated proliferation and differentiation of C17.2 and NSCs reversed by the ERK inhibitor SCH772984 and the BDNF inhibitor ANA-12, respectively. CONCLUSION Our study demonstrated that PA extract promoted neurogenesis and angiogenesis by activating the CREB/ERK signaling pathway and upregulating BDNF expression, thereby recovering neurological dysfunction in post-stroke.
Collapse
Affiliation(s)
- Jiangyan Rao
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Hongpu Li
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Haonan Zhang
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Xiaoxia Xiang
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Xinyu Ding
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Li Li
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Funeng Geng
- Sichuan Key Laboratory of Medical American Cockroach, Chengdu, Sichuan, 610000, China.
| | - Hongyi Qi
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
8
|
Hamanaka G, Hernández IC, Takase H, Ishikawa H, Benboujja F, Kimura S, Fukuda N, Guo S, Lok J, Lo EH, Arai K. Myelination- and migration-associated genes are downregulated after phagocytosis in cultured oligodendrocyte precursor cells. J Neurochem 2023; 167:571-581. [PMID: 37874764 PMCID: PMC10842993 DOI: 10.1111/jnc.15994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023]
Abstract
In the central nervous system, microglia are responsible for removing infectious agents, damaged/dead cells, and amyloid plaques by phagocytosis. Other cell types, such as astrocytes, are also recently recognized to show phagocytotic activity under some conditions. Oligodendrocyte precursor cells (OPCs), which belong to the same glial cell family as microglia and astrocytes, may have similar functions. However, it remains largely unknown whether OPCs exhibit phagocytic activity against foreign materials like microglia. To answer this question, we examined the phagocytosis activity of OPCs using primary rat OPC cultures. Since innate phagocytosis activity could trigger cell death pathways, we also investigated whether participating in phagocytosis activity may lead to OPC cell death. Our data shows that cultured OPCs phagocytosed myelin-debris-rich lysates prepared from rat corpus callosum, without progressing to cell death. In contrast to OPCs, mature oligodendrocytes did not show phagocytotic activity against the bait. OPCs also exhibited phagocytosis towards lysates of rat brain cortex and cell membrane debris from cultured astrocytes, but the percentage of OPCs that phagocytosed beta-amyloid was much lower than the myelin debris. We then conducted RNA-seq experiments to examine the transcriptome profile of OPC cultures and found that myelination- and migration-associated genes were downregulated 24 h after phagocytosis. On the other hand, there were a few upregulated genes in OPCs 24 h after phagocytosis. These data confirm that OPCs play a role in debris removal and suggest that OPCs may remain in a quiescent state after phagocytosis.
Collapse
Affiliation(s)
- Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Iván Coto Hernández
- Surgical Photonics and Engineering Laboratory, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fouzi Benboujja
- Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School
| | - Shintaro Kimura
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Norito Fukuda
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuzhen Guo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eng H. Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Yin D, Wang C, Qi Y, Wang YC, Hagemann N, Mohamud Yusuf A, Dzyubenko E, Kaltwasser B, Tertel T, Giebel B, Gunzer M, Popa-Wagner A, Doeppner TR, Hermann DM. Neural precursor cell delivery induces acute post-ischemic cerebroprotection, but fails to promote long-term stroke recovery in hyperlipidemic mice due to mechanisms that include pro-inflammatory responses associated with brain hemorrhages. J Neuroinflammation 2023; 20:210. [PMID: 37715288 PMCID: PMC10504699 DOI: 10.1186/s12974-023-02894-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND The intravenous delivery of adult neural precursor cells (NPC) has shown promising results in enabling cerebroprotection, brain tissue remodeling, and neurological recovery in young, healthy stroke mice. However, the translation of cell-based therapies to clinical settings has encountered challenges. It remained unclear if adult NPCs could induce brain tissue remodeling and recovery in mice with hyperlipidemia, a prevalent vascular risk factor in stroke patients. METHODS Male mice on a normal (regular) diet or on cholesterol-rich Western diet were exposed to 30 min intraluminal middle cerebral artery occlusion (MCAO). Vehicle or 106 NPCs were intravenously administered immediately after reperfusion, at 3 day and 7 day post-MCAO. Neurological recovery was evaluated using the Clark score, Rotarod and tight rope tests over up to 56 days. Histochemistry and light sheet microscopy were used to examine ischemic injury and brain tissue remodeling. Immunological responses in peripheral blood and brain were analyzed through flow cytometry. RESULTS NPC administration reduced infarct volume, blood-brain barrier permeability and the brain infiltration of neutrophils, monocytes, T cells and NK cells in the acute stroke phase in both normolipidemic and hyperlipidemic mice, but increased brain hemorrhage formation and neutrophil, monocyte and CD4+ and CD8+ T cell counts and activation in the blood of hyperlipidemic mice. While neurological deficits in hyperlipidemic mice were reduced by NPCs at 3 day post-MCAO, NPCs did not improve neurological deficits at later timepoints. Besides, NPCs did not influence microglia/macrophage abundance and activation (assessed by morphology analysis), astroglial scar formation, microvascular length or branching point density (evaluated using light sheet microscopy), long-term neuronal survival or brain atrophy in hyperlipidemic mice. CONCLUSIONS Intravenously administered NPCs did not have persistent effects on post-ischemic neurological recovery and brain remodeling in hyperlipidemic mice. These findings highlight the necessity of rigorous investigations in vascular risk factor models to fully assess the long-term restorative effects of cell-based therapies. Without comprehensive studies in such models, the clinical potential of cell-based therapies cannot be definitely determined.
Collapse
Affiliation(s)
- Dongpei Yin
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Yachao Qi
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Ya-Chao Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
- Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Nina Hagemann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Egor Dzyubenko
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Britta Kaltwasser
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging and Imaging Center Essen (IMCES), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Leibniz-Institut für Analytische Wissenschaften –ISAS– e.V., Dortmund, Germany
| | - Aurel Popa-Wagner
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
- Center of Experimental and Clinical Medicine, University of Medicine and Pharmacy, Craiova, Romania
| | - Thorsten R. Doeppner
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
- Department of Neurology, Justus-Liebig University Gießen, Giessen, Germany
| | - Dirk M. Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
10
|
Zhao H, Wang M, Huang X, Wu X, Xiao H, Jin F, Lv J, Cheng J, Zhao Y, Zhang C. Wasp venom from Vespa magnifica acts as a neuroprotective agent to alleviate neuronal damage after stroke in rats. PHARMACEUTICAL BIOLOGY 2022; 60:334-346. [PMID: 35171059 PMCID: PMC8863380 DOI: 10.1080/13880209.2022.2032207] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 11/24/2021] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT Acute ischaemic stroke (AIS) is a major cause of disability and death, which is a serious threat to human health and life. Wasp venom extracted from Vespa magnifica Smith (Vespidae) could treat major neurological disorders. OBJECTIVE This study investigated the effects of wasp venom on AIS in rats. MATERIAL AND METHODS We used a transient middle cerebral artery occlusion (MCAO) model in Sprague-Dawley rats (260-280 g, n = 8-15) with a sham operation group being treated as negative control. MCAO rats were treated with wasp venom (0.05, 0.2 and 0.6 mg/kg, i.p.) using intraperitoneal injection. After treatment 48 h, behavioural tests, cortical blood flow (CBF), TTC staining, H&E staining, Nissl staining, TUNEL assay, immunohistochemistry (IHC) and ELISA were employed to investigate neuroprotective effects of wasp venom. RESULTS Compared with the MCAO group, wasp venom (0.6 mg/kg) improved neurological impairment, accelerated CBF recovery (205.6 ± 52.92 versus 216.7 ± 34.56), reduced infarct volume (337.1 ± 113.2 versus 140.7 ± 98.03) as well as BBB permeability as evidenced by changes in claudin-5 and AQP4. In addition, function recovery of stroke by wasp venom treatment was associated with a decrease in TNF-α, IL-1β, IL-6 and inhibition activated microglia as well as apoptosis. Simultaneously, the wasp venom regulated the angiogenesis factors VEGF and b-FGF in the brain. CONCLUSIONS Wasp venom exhibited a potential neuroprotective effect for AIS. In the future, we will focus on determining whether the observed actions were due to a single compound or the interaction of multiple components of the venom.
Collapse
Affiliation(s)
- Hairong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- School of Medicine, Xiamen University, Xiamen, PR China
| | - Mei Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
| | - Xi Huang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
| | - Xiumei Wu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Huai Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Fanmao Jin
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
| | - Jiaming Lv
- School of Medicine, Xiamen University, Xiamen, PR China
| | - Jidong Cheng
- School of Medicine, Xiamen University, Xiamen, PR China
| | - Yu Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| |
Collapse
|
11
|
Kugler C, Blank N, Matuskova H, Thielscher C, Reichenbach N, Lin TC, Bradke F, Petzold GC. Pregabalin improves axon regeneration and motor outcome in a rodent stroke model. Brain Commun 2022; 4:fcac170. [PMID: 36072905 PMCID: PMC9443992 DOI: 10.1093/braincomms/fcac170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/24/2022] [Accepted: 06/24/2022] [Indexed: 12/03/2022] Open
Abstract
Ischaemic stroke remains a leading cause of death and disability worldwide. Surviving neurons in the peri-infarct area are able to establish novel axonal projections to juxtalesional regions, but this regeneration is curtailed by a growth-inhibitory environment induced by cells such as reactive astrocytes in the glial scar. Here, we found that the astroglial synaptogenic cue thrombospondin-1 is upregulated in the peri-infarct area, and hence tested the effects of the anticonvulsant pregabalin, a blocker of the neuronal thrombospondin-1 receptor Alpha2delta1/2, in a mouse model of cortical stroke. Studying axonal projections after cortical stroke in mice by three-dimensional imaging of cleared whole-brain preparations, we found that pregabalin, when administered systemically for 5 weeks after stroke, augments novel peri-infarct motor cortex projections and improves skilled forelimb motor function. Thus, the promotion of axon elongation across the glial scar by pregabalin represents a promising target beyond the acute phase after stroke to improve structural and functional recovery.
Collapse
Affiliation(s)
- Christof Kugler
- Vascular Neurology Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
| | - Nelli Blank
- Vascular Neurology Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
| | - Hana Matuskova
- Vascular Neurology Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
| | - Christian Thielscher
- Vascular Neurology Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
| | - Nicole Reichenbach
- Vascular Neurology Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
| | - Tien-Chen Lin
- Axon Growth and Regeneration Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
| | - Frank Bradke
- Axon Growth and Regeneration Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
| | - Gabor C Petzold
- Vascular Neurology Laboratory, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn , Germany
- Division of Vascular Neurology, University Hospital Bonn , 53127 Bonn , Germany
| |
Collapse
|
12
|
Mostajeran M, Edvinsson L, Ahnstedt H, Arkelius K, Ansar S. Repair-related molecular changes during recovery phase of ischemic stroke in female rats. BMC Neurosci 2022; 23:23. [PMID: 35413803 PMCID: PMC9004052 DOI: 10.1186/s12868-022-00696-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Some degree of spontaneous recovery is usually observed after stroke. Experimental studies have provided information about molecular mechanisms underlying this recovery. However, the majority of pre-clinical stroke studies are performed in male rodents, and females are not well studied. This is a clear discrepancy when considering the clinical situation. Thus, it is important to include females in the evaluation of recovery mechanisms for future therapeutic strategies. This study aimed to evaluate spontaneous recovery and molecular mechanisms involved in the recovery phase two weeks after stroke in female rats. METHODS Transient middle cerebral artery occlusion was induced in female Wistar rats using a filament model. Neurological functions were assessed up to day 14 after stroke. Protein expression of interleukin 10 (IL-10), transforming growth factor (TGF)-β, neuronal specific nuclei protein (NeuN), nestin, tyrosine-protein kinase receptor Tie-2, extracellular signal-regulated kinase (ERK) 1/2, and Akt were evaluated in the peri-infarct and ischemic core compared to contralateral side of the brain at day 14 by western blot. Expression of TGF-β in middle cerebral arteries was evaluated by immunohistochemistry. RESULTS Spontaneous recovery after stroke was observed from day 2 to day 14 and was accompanied by a significantly higher expression of nestin, p-Akt, p-ERK1/2 and TGF-β in ischemic regions compared to contralateral side at day 14. In addition, a significantly higher expression of TGF-β was observed in occluded versus non-occluded middle cerebral arteries. The expression of Tie-2 and IL-10 did not differ between the ischemic and contralateral sides. CONCLUSION Spontaneous recovery after ischemic stroke in female rats was coincided by a difference observed in the expression of molecular markers. The alteration of these markers might be of importance to address future therapeutic strategies.
Collapse
Affiliation(s)
- Maryam Mostajeran
- Division of Experimental Vascular Research, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lars Edvinsson
- Division of Experimental Vascular Research, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Hilda Ahnstedt
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kajsa Arkelius
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Saema Ansar
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
13
|
Ma S, Zhang M, Qu H, Cheng Y, Du S, Fan J, Yao Q, Zhang X, Chen M, Zhang N, Shi K, Huang Y, Zhan S. Combination of High-Density Lipoprotein Cholesterol and Lipoprotein(a) as a Predictor of Collateral Circulation in Patients With Severe Unilateral Internal Carotid Artery Stenosis or Occlusion. J Clin Neurol 2022; 18:14-23. [PMID: 35021272 PMCID: PMC8762497 DOI: 10.3988/jcn.2022.18.1.14] [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: 06/28/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Collateral circulation is considered an important factor affecting the risk of stroke, but the factors that affect collateral circulation remain unclear. This study was performed to identify the factors associated with collateral circulation, especially blood lipids. METHODS The study involved patients who had undergone digital subtraction angiography and were confirmed as having severe unilateral stenosis or occlusion of the internal carotid artery (ICA). We classified the collateral circulation status of each patient as good (Grade 3 or 4) or poor (Grade 0, 1, or 2) according to the grading system of the American Society of Interventional and Therapeutic Neuroradiology/American Society of Interventional Radiology. We collected data on patients' characteristics and identified the factors that affect collateral circulation. RESULTS This study included 212 patients. The multivariate logistic regression analysis showed that the high-density lipoprotein cholesterol (HDL-C) concentration and a complete anterior half of the circle of Willis were independent protective factors for good collateral circulation, whereas elevated lipoprotein(a) [Lp(a)] and serum creatinine concentrations were independent risk factors for good collateral circulation. The area under the receiver operating characteristics curve (AUC) was 0.68 (95% confidence interval [CI], 0.61-0.76) for HDL-C and 0.69 (95% CI, 0.62-0.76) for Lp(a). A binary logistic regression model analysis of the joint factor of HDL-C and Lp(a) yielded an AUC of 0.77 (95% CI, 0.71-0.84). CONCLUSIONS In patients with severe unilateral ICA stenosis or occlusion, the combination of HDL-C and Lp(a) is a useful predictor of collateral circulation.
Collapse
Affiliation(s)
- Shuyin Ma
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meijuan Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huiyang Qu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuxuan Cheng
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuang Du
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaxin Fan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qingling Yao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaodong Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengying Chen
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Nan Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kaili Shi
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yizhou Huang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuqin Zhan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
14
|
Jiang L, Chen W, Ye J, Wang Y. Potential Role of Exosomes in Ischemic Stroke Treatment. Biomolecules 2022; 12:115. [PMID: 35053263 PMCID: PMC8773818 DOI: 10.3390/biom12010115] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/01/2022] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is a life-threatening cerebral vascular disease and accounts for high disability and mortality worldwide. Currently, no efficient therapeutic strategies are available for promoting neurological recovery in clinical practice, except rehabilitation. The majority of neuroprotective drugs showed positive impact in pre-clinical studies but failed in clinical trials. Therefore, there is an urgent demand for new promising therapeutic approaches for ischemic stroke treatment. Emerging evidence suggests that exosomes mediate communication between cells in both physiological and pathological conditions. Exosomes have received extensive attention for therapy following a stroke, because of their unique characteristics, such as the ability to cross the blood brain-barrier, low immunogenicity, and low toxicity. An increasing number of studies have demonstrated positively neurorestorative effects of exosome-based therapy, which are largely mediated by the microRNA cargo. Herein, we review the current knowledge of exosomes, the relationships between exosomes and stroke, and the therapeutic effects of exosome-based treatments in neurovascular remodeling processes after stroke. Exosomes provide a viable and prospective treatment strategy for ischemic stroke patients.
Collapse
Affiliation(s)
- Lingling Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Weiqi Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Jinyi Ye
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| |
Collapse
|
15
|
Acid sphingomyelinase deactivation post-ischemia promotes brain angiogenesis and remodeling by small extracellular vesicles. Basic Res Cardiol 2022; 117:43. [PMID: 36038749 PMCID: PMC9424180 DOI: 10.1007/s00395-022-00950-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/17/2022] [Accepted: 08/08/2022] [Indexed: 01/31/2023]
Abstract
Antidepressants have been reported to enhance stroke recovery independent of the presence of depressive symptoms. They have recently been proposed to exert their mood-stabilizing actions by inhibition of acid sphingomyelinase (ASM), which catalyzes the hydrolysis of sphingomyelin to ceramide. Their restorative action post-ischemia/reperfusion (I/R) still had to be defined. Mice subjected to middle cerebral artery occlusion or cerebral microvascular endothelial cells exposed to oxygen-glucose deprivation were treated with vehicle or with the chemically and pharmacologically distinct antidepressants amitriptyline, fluoxetine or desipramine. Brain ASM activity significantly increased post-I/R, in line with elevated ceramide levels in microvessels. ASM inhibition by amitriptyline reduced ceramide levels, and increased microvascular length and branching point density in wildtype, but not sphingomyelinase phosphodiesterase-1 ([Smpd1]-/-) (i.e., ASM-deficient) mice, as assessed by 3D light sheet microscopy. In cell culture, amitriptyline, fluoxetine, and desipramine increased endothelial tube formation, migration, VEGFR2 abundance and VEGF release. This effect was abolished by Smpd1 knockdown. Mechanistically, the promotion of angiogenesis by ASM inhibitors was mediated by small extracellular vesicles (sEVs) released from endothelial cells, which exhibited enhanced uptake in target cells. Proteomic analysis of sEVs revealed that ASM deactivation differentially regulated proteins implicated in protein export, focal adhesion, and extracellular matrix interaction. In vivo, the increased angiogenesis was accompanied by a profound brain remodeling response with increased blood-brain barrier integrity, reduced leukocyte infiltrates and increased neuronal survival. Antidepressive drugs potently boost angiogenesis in an ASM-dependent way. The release of sEVs by ASM inhibitors disclosed an elegant target, via which brain remodeling post-I/R can be amplified.
Collapse
|
16
|
Chavda V, Chaurasia B, Deora H, Umana GE. Chronic Kidney disease and stroke: A Bi-directional risk cascade and therapeutic update. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
17
|
Zhang Y, Zhang Y, Chopp M, Pang H, Zhang ZG, Mahmood A, Xiong Y. MiR-17-92 Cluster-Enriched Exosomes Derived from Human Bone Marrow Mesenchymal Stromal Cells Improve Tissue and Functional Recovery in Rats after Traumatic Brain Injury. J Neurotrauma 2021; 38:1535-1550. [PMID: 33787364 DOI: 10.1089/neu.2020.7575] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exosomes play an important role in intercellular communication by delivering microribonucleic acids (miRNAs) to recipient cells. Previous studies have demonstrated that multi-potent mesenchymal stromal cell (MSC)-derived exosomes improve functional recovery after experimental traumatic brain injury (TBI). This study was performed to determine efficacy of miR-17-92 cluster-enriched exosomes (Exo-17-92) harvested from human bone marrow MSCs transfected with a miR-17-92 cluster plasmid in enhancing tissue and neurological recovery compared with exosomes derived from MSCs transfected with an empty plasmid vector (Exo-empty) for treatment of TBI. Adult male rats underwent a unilateral moderate cortical contusion. Animals received a single intravenous injection of miR-17-92 cluster-enriched exosomes (100 μg/rat, approximately 3.75x1011 particles, Exo-17-92) or control exosomes (100 μg/rat, Exo-empty) or Vehicle (phosphate-buffered solution) one day after injury. A battery of neurological functional tests was performed weekly after TBI for five weeks. Spatial learning and memory were measured on days 31-35 after TBI using the Morris water maze test. All animals were sacrificed five weeks after injury. Their brains were processed for histopathological and immunohistochemical analyses of lesion volume, cell loss, angiogenesis, neurogenesis, and neuroinflammation. Compared with Vehicle, both Exo-17-92 and Exo-empty treatments significantly improved sensorimotor and cognitive function, reduced neuroinflammation and hippocampal neuronal cell loss, promoted angiogenesis and neurogenesis without altering the lesion volume. Moreover, Exo-17-92 treatment exhibited a significantly more robust therapeutic effect on improvement in functional recovery by reducing neuroinflammation and cell loss, enhancing angiogenesis and neurogenesis than did Exo-empty treatment. Exosomes enriched with miR-17-92 cluster have a significantly better effect on improving functional recovery after TBI compared with Exo-empty, likely by reducing neuroinflammation and enhancing endogenous angiogenesis and neurogenesis. Engineering specific miRNA in exosomes may provide a novel therapeutic strategy for management of unilateral moderate cortical contusion TBI.
Collapse
Affiliation(s)
- Yanlu Zhang
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Haiyan Pang
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Asim Mahmood
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| | - Ye Xiong
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
18
|
Prospects of Therapeutic Target and Directions for Ischemic Stroke. Pharmaceuticals (Basel) 2021; 14:ph14040321. [PMID: 33916253 PMCID: PMC8065883 DOI: 10.3390/ph14040321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is a serious, adverse neurological event and the third leading cause of death and disability worldwide. Most strokes are caused by a block in cerebral blood flow, resulting in neurological deficits through the death of brain tissue. Recombinant tissue plasminogen activator (rt-PA) is currently the only immediate treatment medication for stroke. The goal of rt-PA administration is to reduce the thrombus and/or embolism via thrombolysis; however, the administration of rt-PA must occur within a very short therapeutic timeframe (3 h to 6 h) after symptom onset. Components of the pathological mechanisms involved in ischemic stroke can be used as potential biomarkers in current treatment. However, none are currently under investigation in clinical trials; thus, further studies investigating biomarkers are needed. After ischemic stroke, microglial cells can be activated and release inflammatory cytokines. These cytokines lead to severe neurotoxicity via the overactivation of microglia in prolonged and lasting insults such as stroke. Thus, the balanced regulation of microglial activation may be necessary for therapy. Stem cell therapy is a promising clinical treatment strategy for ischemic stroke. Stem cells can increase the functional recovery of damaged tissue after post-ischemic stroke through various mechanisms including the secretion of neurotrophic factors, immunomodulation, the stimulation of endogenous neurogenesis, and neovascularization. To investigate the use of stem cell therapy for neurological diseases in preclinical studies, however, it is important to develop imaging technologies that are able to evaluate disease progression and to “chase” (i.e., track or monitor) transplanted stem cells in recipients. Imaging technology development is rapidly advancing, and more sensitive techniques, such as the invasive and non-invasive multimodal techniques, are under development. Here, we summarize the potential risk factors and biomarker treatment strategies, stem cell-based therapy and emerging multimodal imaging techniques in the context of stroke. This current review provides a conceptual framework for considering the therapeutic targets and directions for the treatment of brain dysfunctions, with a particular focus on ischemic stroke.
Collapse
|
19
|
Chavda V, Madhwani K. Coding and non-coding nucleotides': The future of stroke gene therapeutics. Genomics 2021; 113:1291-1307. [PMID: 33677059 DOI: 10.1016/j.ygeno.2021.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/01/2020] [Accepted: 03/02/2021] [Indexed: 01/05/2023]
Abstract
Stroke is the foremost cause of death ranked after heart disease and cancer. It is the fatal life-threatening event that requires immediate medical admissions to overcome following morbidity and mortality. The therapeutic advances in stroke therapy have been manipulated with diverse paths for last 5 years. Recent research and clinical trials have investigated a variety of anti-stroke agents including anti-coagulants, cerebro-protective agents, antiplatelet therapy, stem-cell therapy, and specified gene therapy. In recent advanced studies, genetic therapies including noncoding RNAs (ncRNAs), long non-coding RNAs (LncRNAs), small interfering RNAs (siRNAs), microRNAs (miRNAs), Piwi interacting RNAs (PiWi RNAs) have shown better potential as targeted future therapeutics with a better outcome than conventional stroke therapeutics. The potential of targeted gene therapy is much more advanced in not only the induction of neuroprotection but also safer non-toxic targeted therapeutics. In the current state of the art review, we have focused on the recent advancements made towards the stroke with RNA modifications and targeted gene therapeutics.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, India.
| | - Kajal Madhwani
- Department of Microbiology, Nirma University, Ahmadabad, Gujarat, India
| |
Collapse
|
20
|
Sommer CJ, Schäbitz WR. Principles and requirements for stroke recovery science. J Cereb Blood Flow Metab 2021; 41:471-485. [PMID: 33175596 PMCID: PMC7907998 DOI: 10.1177/0271678x20970048] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022]
Abstract
The disappointing results in bench-to-bedside translation of neuroprotective strategies caused a certain shift in stroke research towards enhancing the endogenous recovery potential of the brain. One reason for this focus on recovery is the much wider time window for therapeutic interventions which is open for at least several months. Since recently two large clinical studies using d-amphetamine or fluoxetine, respectively, to enhance post-stroke neurological outcome failed again it is a good time for a critical reflection on principles and requirements for stroke recovery science. In principal, stroke recovery science deals with all events from the molecular up to the functional and behavioral level occurring after brain ischemia eventually ending up with any measurable improvement of various clinical parameters. A detailed knowledge of the spontaneously occurring post-ischemic regeneration processes is the indispensable prerequisite for any therapeutic approaches aiming to modify these responses to enhance post-stroke recovery. This review will briefly illuminate the molecular mechanisms of post-ischemic regeneration and the principle possibilities to foster post-stroke recovery. In this context, recent translational approaches are analyzed. Finally, the principal and specific requirements and pitfalls in stroke recovery research as well as potential explanations for translational failures will be discussed.
Collapse
Affiliation(s)
- Clemens J Sommer
- Institute of Neuropathology, University Medical Center of the
Johannes Gutenberg-University Mainz, Mainz, Germany
| | | |
Collapse
|
21
|
Brain-Derived Extracellular Vesicles in Health and Disease: A Methodological Perspective. Int J Mol Sci 2021; 22:ijms22031365. [PMID: 33573018 PMCID: PMC7866382 DOI: 10.3390/ijms22031365] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are double membrane structures released by presumably all cell types that transport and deliver lipids, proteins, and genetic material to near or distant recipient cells, thereby affecting their phenotype. The basic knowledge of their functions in healthy and diseased brain is still murky and many questions about their biology are unsolved. In neurological diseases, EVs are regarded as attractive biomarkers and as therapeutic tools due to their ability to cross the blood–brain barrier (BBB). EVs have been successfully isolated from conditioned media of primary brain cells and cerebrospinal fluid (CSF), but protocols allowing for the direct study of pathophysiological events mediated or influenced by EVs isolated from brain have only recently been published. This review aims to give a brief overview of the current knowledge of EVs’ functions in the central nervous system (CNS) and the current protocols to isolate brain-derived EVs (BDEVs) used in different publications. By comparing the proteomic analysis of some of these publications, we also assess the influence of the isolation method on the protein content of BDEVs.
Collapse
|
22
|
Du L, Zhao Z, Liu X, Chen Y, Gao W, Wang Y, Liu J, Liu B, Ma G. Alterations of Iron Level in the Bilateral Basal Ganglia Region in Patients With Middle Cerebral Artery Occlusion. Front Neurosci 2021; 14:608058. [PMID: 33551726 PMCID: PMC7859276 DOI: 10.3389/fnins.2020.608058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/17/2020] [Indexed: 11/27/2022] Open
Abstract
Background and Purpose: The purpose of this study was to explore the changes of iron level using quantitative susceptibility mapping (QSM) in the bilateral basal ganglia region in middle cerebral artery occlusion (MCAO) patients with long-term ischemia. Methods: Twenty-seven healthy controls and nine patients with MCAO were recruited, and their QSM images were obtained. The bilateral caudate nucleus (Cd), putamen (Pt), and globus pallidus (Gp) were selected as the regions of interest (ROIs). Susceptibility values of bilateral ROIs were calculated and compared between the affected side and unaffected side in patients with MCAO and between patients with MCAO and healthy controls. In addition, receiver operating characteristic (ROC) curves were performed to evaluate the diagnostic capability of susceptibility values in differentiating healthy controls and patients with MCAO by the area under the curve (AUC). Results: The susceptibility values of bilateral Cd were asymmetric in healthy controls; however, this asymmetry disappeared in patients with MCAO. In addition, compared with healthy controls, the average susceptibility values of the bilateral Pt in patients with MCAO were increased (P < 0.05), and the average susceptibility value of the bilateral Gp was decreased (P < 0.05). ROC curves showed that the susceptibility values of the Pt and Gp had a larger AUC (AUC = 0.700 and 0.889, respectively). Conclusion: As measured by QSM, the iron levels of the bilateral basal ganglia region were significantly changed in patients with MCAO. Iron dyshomeostasis in the basal ganglia region might be involved in the pathophysiological process of middle cerebral artery stenosis and occlusion. These findings may provide a novel insight to profoundly address the pathophysiological mechanisms of MCAO.
Collapse
Affiliation(s)
- Lei Du
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zifang Zhao
- Department of Anesthesiology, Peking University First Hospital, Peking University, Beijing, China
| | - Xiuxiu Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yue Chen
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Wenwen Gao
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yige Wang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Jian Liu
- Department of Ultrasound Diagnosis, China-Japan Friendship Hospital, Beijing, China
| | - Bing Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Li L, Gan H, Jin H, Fang Y, Yang Y, Zhang J, Hu X, Chu L. Astragaloside IV promotes microglia/macrophages M2 polarization and enhances neurogenesis and angiogenesis through PPARγ pathway after cerebral ischemia/reperfusion injury in rats. Int Immunopharmacol 2021; 92:107335. [PMID: 33429332 DOI: 10.1016/j.intimp.2020.107335] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022]
Abstract
Microglia/macrophages play a dual role in brain injury and repair following cerebral ischemia/reperfusion. Promoting microglia/macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2 phenotype has been considered as a potential treatment for ischemic stroke. Astragaloside IV (AS-IV) is a primary active ingredient of Chinese herb Radix Astragali, which protects against acute cerebral ischemic/reperfusion injury through its antioxidant, anti-inflammatory, and anti-apoptotic properties. However, it remains unknown whether AS-IV improves ischemic brain tissue repair and its underlying mechanism. A transient middle cerebral artery occlusion (tMCAO) rat model was used in this study. The results showed that AS-IV significantly improved long-term brain injury, reduced the expression of M1 microglia/macrophage markers and increased the expression of M2 microglia/macrophage markers 14 days after cerebral ischemia/reperfusion. AS-IV also increased peroxisome proliferator-activated receptor γ (PPARγ) mRNA and protein expression. Moreover, AS-IV promoted neurogenesis and angiogenesis, and increased the protein expression of brain-derived growth factor (BDNF), insulin-like growth factor-1 (IGF-1) and vascular endothelial growth factor (VEGF). However, these beneficial effects were greatly blocked by PPARγ antagonist T0070907. These results together suggest that AS-IV could enhance neurogenesis, angiogenesis and neurological functional recovery, which may be partially through transforming microglia/macrophage from M1 to M2 phenotype in a PPARγ-dependent manner after cerebral ischemia/reperfusion injury. Therefore, AS-IV can be considered as a promising therapeutic agent for ischemic stroke.
Collapse
Affiliation(s)
- Lin Li
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haiyan Gan
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huaqian Jin
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Fang
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Yang
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianping Zhang
- Department of Anatomy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaowei Hu
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lisheng Chu
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
24
|
Shindo A, Takase H, Hamanaka G, Chung KK, Mandeville ET, Egawa N, Maki T, Borlongan M, Takahashi R, Lok J, Tomimoto H, Lo EH, Arai K. Biphasic roles of pentraxin 3 in cerebrovascular function after white matter stroke. CNS Neurosci Ther 2020; 27:60-70. [PMID: 33314664 PMCID: PMC7804900 DOI: 10.1111/cns.13510] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/24/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022] Open
Abstract
Recent clinical studies suggest that pentraxin 3 (PTX3), which is known as an acute-phase protein that is produced rapidly at local sites of inflammation, may be a new biomarker of disease risk for central nervous system disorders, including stroke. However, the effects of PTX3 on cerebrovascular function in the neurovascular unit (NVU) after stroke are mostly unknown, and the basic research regarding the roles of PTX3 in NVU function is still limited. In this reverse translational study, we prepared mouse models of white matter stroke by vasoconstrictor (ET-1 or L-Nio) injection into the corpus callosum region to examine the roles of PTX3 in the pathology of cerebral white matter stroke. PTX3 expression was upregulated in GFAP-positive astrocytes around the affected region in white matter for at least 21 days after vasoconstrictor injection. When PTX3 expression was reduced by PTX3 siRNA, blood-brain barrier (BBB) damage at day 3 after white matter stroke was exacerbated. In contrast, when PTX3 siRNA was administered at day 7 after white matter stroke, compensatory angiogenesis at day 21 was promoted. In vitro cell culture experiments confirmed the inhibitory effect of PTX3 in angiogenesis, that is, recombinant PTX3 suppressed the tube formation of cultured endothelial cells in a Matrigel-based in vitro angiogenesis assay. Taken together, our findings may support a novel concept that astrocyte-derived PTX3 plays biphasic roles in cerebrovascular function after white matter stroke; additionally, it may also provide a proof-of-concept that PTX3 could be a therapeutic target for white matter-related diseases, including stroke.
Collapse
Affiliation(s)
- Akihiro Shindo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hajime Takase
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kelly K Chung
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Naohiro Egawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mia Borlongan
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Pediatric Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
25
|
Zhang J, Sun P, Zhou C, Zhang X, Ma F, Xu Y, Hamblin MH, Yin K. Regulatory microRNAs and vascular cognitive impairment and dementia. CNS Neurosci Ther 2020; 26:1207-1218. [PMID: 33459504 PMCID: PMC7702235 DOI: 10.1111/cns.13472] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is defined as a progressive dementia disease related to cerebrovascular injury and often occurs in aged populations. Despite decades of research, effective treatment for VCID is still absent. The pathological processes of VCID are mediated by the molecular mechanisms that are partly modulated at the post-transcriptional level. As small endogenous non-coding RNAs, microRNAs (miRs) can regulate target gene expression through post-transcriptional gene silencing. miRs have been reported to play an important role in the pathology of VCID and have recently been suggested as potential novel pharmacological targets for the development of new diagnosis and treatment strategies in VCID. In this review, we summarize the current understanding of VCID, the possible role of miRs in the regulation of VCID and attempt to envision future therapeutic strategies. Since manipulation of miR levels by either pharmacological or genetic approaches has shown therapeutic effects in experimental VCID models, we also emphasize the potential therapeutic value of miRs in clinical settings.
Collapse
Affiliation(s)
- Jing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Ping Sun
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Chao Zhou
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Xuejing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Feifei Ma
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Yang Xu
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Milton H. Hamblin
- Department of PharmacologyTulane University School of MedicineNew OrleansLAUSA
| | - Ke‐Jie Yin
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Geriatric ResearchEducation and Clinical CenterVeterans Affairs Pittsburgh Healthcare SystemPittsburghPAUSA
| |
Collapse
|
26
|
Zhang Y, Qin Y, Chopp M, Li C, Kemper A, Liu X, Wang X, Zhang L, Zhang ZG. Ischemic Cerebral Endothelial Cell-Derived Exosomes Promote Axonal Growth. Stroke 2020; 51:3701-3712. [PMID: 33138691 PMCID: PMC7686085 DOI: 10.1161/strokeaha.120.031728] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/07/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral endothelial cells (CECs) and axons of neurons interact to maintain vascular and neuronal homeostasis and axonal remodeling in normal and ischemic brain, respectively. However, the role of exosomes in the interaction of CECs and axons in brain under normal conditions and after stroke is unknown. METHODS Exosomes were isolated from CECs of nonischemic rats and is chemic rats (nCEC-exos and isCEC-exos), respectively. A multicompartmental cell culture system was used to separate axons from neuronal cell bodies. RESULTS Axonal application of nCEC-exos promotes axonal growth of cortical neurons, whereas isCEC-exos further enhance axonal growth than nCEC-exos. Ultrastructural analysis revealed that CEC-exos applied into distal axons were internalized by axons and reached to their parent somata. Bioinformatic analysis revealed that both nCEC-exos and isCEC-exos contain abundant mature miRNAs; however, isCEC-exos exhibit more robust elevation of select miRNAs than nCEC-exos. Mechanistically, axonal application of nCEC-exos and isCEC-exos significantly elevated miRNAs and reduced proteins in distal axons and their parent somata that are involved in inhibiting axonal outgrowth. Blockage of axonal transport suppressed isCEC-exo-altered miRNAs and proteins in somata but not in distal axons. CONCLUSIONS nCEC-exos and isCEC-exos facilitate axonal growth by altering miRNAs and their target protein profiles in recipient neurons.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
| | - Yi Qin
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
- Department of Physics, Oakland University, Rochester, Michigan, 48309
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
| | - Amy Kemper
- Department of Pathology, Henry Ford Hospital, Detroit, Michigan, 48202
| | - Xianshuang Liu
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
| | - Xinli Wang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, 48202
| |
Collapse
|
27
|
Enhanced repair processes and iron uptake by ischemic preconditioning in the brain during the recovery phase after ischemic stroke. Brain Res 2020; 1750:147172. [PMID: 33141066 DOI: 10.1016/j.brainres.2020.147172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022]
Abstract
Ischemic preconditioning (IP) reduces brain damage after subsequent ischemic strokes by activating endogenous protective mechanisms in rodents. Transient ischemic attack (TIA) induces tolerance in the human brain after ischemic strokes; defining mechanisms of IP effects may provide therapeutic targets to improve recovery of patients with ischemic strokes. Iron transported across the blood-brain barrier (BBB) is required for brain functions, including myelination, and its levels should be finely regulated to avoid harmful effects. This study aimed to determine whether IP enhances repair processes by modulating iron metabolism during the post-stroke chronic phase. Male mice were divided into sham and IP groups, and IP was induced 24 h before a transient focal ischemic stroke. Sensorimotor recovery was observed over 8 weeks after the stroke, and brain volumes and levels of proteins related to repair processes and iron metabolism in the ischemic brains were examined 8 weeks after the stroke. There was significantly less ischemic brain atrophy in the IP group than in the sham group, with no differences in sensorimotor recovery between the groups. Levels of tight junction proteins of BBB, neurites outgrowth markers, and myelin sheath proteins and markers for mature oligodendrocytes were significantly increased in the IP group. Iron import proteins, transferrin receptor 1 and DMT1, were also increased in the IP group. These results indicate that IP increases brain repair processes and iron uptake during the chronic phase after an ischemic stroke, and provide new insights to understand the molecular mechanisms of TIA effects on post-stroke recovery.
Collapse
|
28
|
Wang C, Chopp M, Huang R, Li C, Zhang Y, Golembieski W, Lu M, Hazan Z, Zhang ZG, Zhang L. Delayed (21 Days) Post Stroke Treatment With RPh201, a Botany-Derived Compound, Improves Neurological Functional Recovery in a Rat Model of Embolic Stroke. Front Neurosci 2020; 14:813. [PMID: 32848574 PMCID: PMC7412960 DOI: 10.3389/fnins.2020.00813] [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: 04/14/2020] [Accepted: 07/10/2020] [Indexed: 11/13/2022] Open
Abstract
Background Despite the recent advances in the acute stroke care, treatment options for long-term disability are limited. RPh201 is a botany-derived bioactive compound that has been shown to exert beneficial effects in various experimental models of neural injury. The present study evaluated the effect of delayed RPh201 treatment on long term functional recovery after stroke. Methods Adult male Wistar rats subjected to embolic middle cerebral artery occlusion (MCAO) were randomized into the following experimental groups (n = 20/group): (1) RPh201 treatment, and (2) Vehicle (cottonseed oil). RPh201 (20 μl) or Vehicle were subcutaneously administered twice a week for 16 consecutive weeks starting at 21 days after MCAO. An array of behavioral tests was performed up to120 days after MCAO. Results Ischemic rats treated with RPh201 exhibited significant (p < 0.05) improvement of neurological function measured by adhesive removal test, foot-fault test, and modified neurological severity score at 90 and 120 days after MCAO. Immunohistochemistry analysis showed that RPh201 treatment robustly increased neurofilament heavy chain positive axons and myelin basic protein densities in the peri-infarct area by 61% and 31%, respectively, when compared to the Vehicle treatment, which were further confirmed by Western blot analysis. The RPh201 treatment did not reduce infarct volume. Conclusion Our data demonstrated that RPh201 has a therapeutic effect on improvement of functional recovery in male ischemic rats even when the treatment was initiated 21 days post stroke. Enhanced axonal and myelination densities by RPh201 in ischemic brain may contribute to improved stroke recovery.
Collapse
Affiliation(s)
- Chunyang Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, MI, United States
| | - Rui Huang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
29
|
Wang MM, Feng YS, Tan ZX, Xing Y, Dong F, Zhang F. The role of exosomes in stroke. Mol Biol Rep 2020; 47:6217-6228. [PMID: 32514999 DOI: 10.1007/s11033-020-05569-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/05/2020] [Indexed: 12/15/2022]
Abstract
Stroke is induced by a partial disruption of cerebral blood flow to the brain and is related to high morbidity and mortality. In the central nervous system, exosomes have been proven to exert neuroprotective effects, reducing brain damage following a stroke. This review was performed by searching the relevant articles in the SCIENCEDIRECT, PUBMED, and Web of Science databases from respective inception to November 2018. We review the relationship between exosomes and angiogenesis, neurogenesis, antiapoptosis, autophagy, and the blood-brain barrier in stroke. Moreover, exosomes are found to be a promising tool for the diagnosis and treatment of stroke. In summary, exosomes provide a novel way to alleviate brain damage following a stroke.
Collapse
Affiliation(s)
- Man-Man Wang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Ying Xing
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China. .,Hebei Provincial Orthopedic Biomechanics Key Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
30
|
Zhang Z, Cao X, Bao X, Zhang Y, Xu Y, Sha D. Cocaine- and amphetamine-regulated transcript protects synaptic structures in neurons after ischemic cerebral injury. Neuropeptides 2020; 81:102023. [PMID: 32005500 DOI: 10.1016/j.npep.2020.102023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/05/2019] [Accepted: 01/23/2020] [Indexed: 01/26/2023]
Abstract
Cocaine-regulated and amphetamine-regulated transcript (CART) is a neuropeptide with reported neuroprotective effects in ischemic cerebral injury. However, its mechanism has not yet been elucidated. Herein, we investigated the role and mechanism of CART in synaptic plasticity in neurons after ischemic cerebral stroke. We found that the survival rate of the oxygen-glucose deprivation (OGD) neurons was increased after CART treatment. Moreover, CART treatment significantly attenuated ischemia-induced neuronal synaptic damage and increased synaptophysin expression. In addition, the number of presynaptic vesicles was increased and the postsynaptic density (PSD) was thickened after CART treatment. Mechanistically, CART treatment enhanced the expression of Arc mRNA in a cAMP response element binding protein (CREB) dependent manner in OGD neurons, and blockade of CREB by KG-501 eliminated the protective effect of CART. Collectively, CART protected the synaptic structure in neurons after ischemic cerebral injury by increasing the Arc expression via upregulating p-CREB.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yan Zhang
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China; Institute of Functional Biomolecules, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Dujuan Sha
- Department of Emergency, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China; Institute of Functional Biomolecules, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
31
|
Effects of mesencephalic astrocyte-derived neurotrophic factor on cerebral angiogenesis in a rat model of cerebral ischemia. Neurosci Lett 2019; 715:134657. [PMID: 31785307 DOI: 10.1016/j.neulet.2019.134657] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum stress-related protein that exhibits neuroprotective effects. Recent studies have shown that MANF promotes poststroke functional recovery in rats. However, the underlying mechanisms have not yet been fully understood. Here, we examined the effects of MANF on cerebral angiogenesis in a permanent middle cerebral artery occlusion model in rats. Recombinant human MANF was administered intracerebroventricularly 24 h after stroke. We performed neurobehavioral tests and assessed microvessel density, functional microvessels, and regional cerebral blood flow (rCBF), as well as detected angiogenic factors in the peri-infarct cerebral cortex. Results showed that MANF ameliorated neurobehavioral scores, promoted rCBF, upregulated the expression of CD34, as well as the total vessel surface area and the number of microvessel branch points, and activated the vascular endothelial growth factor (VEGF) pathway. In conclusion, our findings provide insight into the mechanisms of MANF in promoting functional recovery from ischemic stroke. Our results suggest that MANF improves neurobehavioral recovery from cerebral ischemic injury, and that this effect is mediated partly by its proangiogenic effects and augmentation of rCBF, which are possibly associated with VEGF.
Collapse
|
32
|
Functions of subventricular zone neural precursor cells in stroke recovery. Behav Brain Res 2019; 376:112209. [PMID: 31493429 DOI: 10.1016/j.bbr.2019.112209] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/11/2019] [Accepted: 09/03/2019] [Indexed: 12/16/2022]
Abstract
The proliferation and ectopic migration of neural precursor cells (NPCs) in response to ischemic brain injury was first reported two decades ago. Since then, studies of brain injury-induced subventricular zone cytogenesis, primarily in rodent models, have provided insight into the cellular and molecular determinants of this phenomenon and its modulation by various factors. However, despite considerable correlational evidence-and some direct evidence-to support contributions of NPCs to behavioral recovery after stroke, the causal mechanisms have not been identified. Here we discuss the subventricular zone cytogenic response and its possible roles in brain injury and disease, focusing on rodent models of stroke. Emerging evidence suggests that NPCs can modulate harmful responses and enhance reparative responses to neurologic diseases. We speculatively identify four broad functions of NPCs in the context of stroke: cell replacement, cytoprotection, remodeling of residual tissue, and immunomodulation. Thus, NPCs may have pleiotropic functions in supporting behavioral recovery after stroke.
Collapse
|
33
|
Thrombin promotes pericyte coverage by Tie2 activation in a rat model of intracerebral hemorrhage. Brain Res 2019; 1708:58-68. [DOI: 10.1016/j.brainres.2018.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 11/18/2022]
|
34
|
Pines AR, Sattur MG, Abi-Aad KR, Bendok BR. Edonerpic Maleate: A Promising Pharmacological Agent for Stroke Recovery. Neurosurgery 2019; 84:E3-E4. [PMID: 30551191 DOI: 10.1093/neuros/nyy397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Andrew R Pines
- Mayo Clinic School of Medicine Mayo Clinic Scottsdale, Arizona
| | - Mithun G Sattur
- Department of Neurological Surgery Mayo Clinic Phoenix, Arizona
- Precision Neuro-therapeutics Innovation Lab Mayo Clinic Phoenix, Arizona
- Neurosurgery Simulation and Innovation Lab Mayo Clinic Phoenix, Arizona
| | - Karl R Abi-Aad
- Department of Neurological Surgery Mayo Clinic Phoenix, Arizona
- Precision Neuro-therapeutics Innovation Lab Mayo Clinic Phoenix, Arizona
- Neurosurgery Simulation and Innovation Lab Mayo Clinic Phoenix, Arizona
| | - Bernard R Bendok
- Department of Neurological Surgery Mayo Clinic Phoenix, Arizona
- Precision Neuro-therapeutics Innovation Lab Mayo Clinic Phoenix, Arizona
- Neurosurgery Simulation and Innovation Lab Mayo Clinic Phoenix, Arizona
- Department of Otolaryngology Mayo Clinic Phoenix, Arizona
- Department of Radiology Mayo Clinic Phoenix, Arizona
| |
Collapse
|
35
|
Yu P, Chen W. Advances in the diagnosis of exosomal miRNAs in ischemic stroke. Neuropsychiatr Dis Treat 2019; 15:2339-2343. [PMID: 31695378 PMCID: PMC6707376 DOI: 10.2147/ndt.s216784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/12/2019] [Indexed: 01/13/2023] Open
Abstract
Early diagnosis, early treatment, and improved prognosis in patients with ischemic stroke are vital requirements. Current clinical practices for the diagnosis of stroke include computed tomography, magnetic resonance imaging, and other traditional imaging methods to quickly check the location, volume, etc, in the hospital; however, diagnosis of the underlying cause of infarction is not effective with these practices. Owing to the coexistence of various etiologies, accurate and timely diagnosis using routine hematology and biochemical tests remains a challenge. Exosomes are membrane vesicles, approximately 30-150 nm in diameter, which fuse with cell membrane and are released into the extracellular space. As one of the research hotspots in the field of medicine in recent years, exosomes can participate in immune response, antigen presentation, cell migration, tumor invasion, and so on. Owing to the important role played by the miRNAs contained in exosomes, the latter have shown great potential in the diagnosis and treatment of ischemic stroke. This article reviews the progress made regarding the exosomal miRNAs as ischemic stroke biomarkers.
Collapse
Affiliation(s)
- Pei Yu
- Department of Clinical Laboratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, People's Republic of China
| | - Wencheng Chen
- Department of Clinical Laboratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, People's Republic of China
| |
Collapse
|
36
|
Sanchez-Rojas L, Gómez-Pinedo U, Benito-Martin MS, León-Espinosa G, Rascón-Ramirez F, Lendinez C, Martínez-Ramos C, Matías-Guiu J, Pradas MM, Barcia JA. Biohybrids of scaffolding hyaluronic acid biomaterials plus adipose stem cells home local neural stem and endothelial cells: Implications for reconstruction of brain lesions after stroke. J Biomed Mater Res B Appl Biomater 2018; 107:1598-1606. [PMID: 30307108 DOI: 10.1002/jbm.b.34252] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/23/2018] [Accepted: 09/08/2018] [Indexed: 12/25/2022]
Abstract
Endogenous neurogenesis in stroke is insufficient to replace the lost brain tissue, largely due to the lack of a proper biological structure to let new cells dwell in the damaged area. We hypothesized that scaffolds made of hyaluronic acid (HA) biomaterials (BM) could provide a suitable environment to home not only new neurons, but also vessels, glia and neurofilaments. Further, the addition of exogenous cells, such as adipose stem cells (ASC) could increase this effect. Athymic mice were randomly assigned to a one of four group: stroke alone, stroke and implantation of BM, stroke and implantation of BM with ASC, and sham operated animals. Stroke model consisted of middle cerebral artery thrombosis with FeCl3 . After 30 days, animals underwent magnetic resonance imaging (MRI) and were sacrificed. Proliferation and neurogenesis increased at the subventricular zone ipsilateral to the ventricle and neuroblasts, glial, and endothelial cells forming capillaries were seen inside the BM. Those effects increased when ASC were added, while there was less inflammatory reaction. Three-dimensional scaffolds made of HA are able to home newly formed neurons, glia, and endothelial cells permitting the growth neurofilaments inside them. The addition of ASC increase these effects and decrease the inflammatory reaction to the implant. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1598-1606, 2019.
Collapse
Affiliation(s)
- Leyre Sanchez-Rojas
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Ulises Gómez-Pinedo
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - María Soledad Benito-Martin
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Gonzalo León-Espinosa
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain.,Instituto Cajal, CSIC; Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid; Facultad de Farmacia, Universidad San Pablo CEU, Madrid, Spain
| | - Fernando Rascón-Ramirez
- Servicio de Neurocirugía, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Lendinez
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politécnica de Valencia, C. de Vera s/n, Valencia, Spain
| | - Jorge Matías-Guiu
- Servicio de Neurología. Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politécnica de Valencia, C. de Vera s/n, Valencia, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Valencia, Spain
| | - Juan A Barcia
- Servicio de Neurocirugía, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
37
|
Sheikh AM, Yano S, Mitaki S, Haque MA, Yamaguchi S, Nagai A. A Mesenchymal stem cell line (B10) increases angiogenesis in a rat MCAO model. Exp Neurol 2018; 311:182-193. [PMID: 30291853 DOI: 10.1016/j.expneurol.2018.10.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 08/29/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022]
Abstract
A human mesenchymal stem cell line (B10) transplantation has been shown to improve ischemia-induced neurological deficits in animal stroke models. To understand the underlying mechanism, we have investigated the effects of B10 transplantation on cerebral angiogenesis in a rat middle cerebral artery occlusion (MCAO) model. B10 cells were transplanted intravenously 24 h after MCAO. Immunofluorescence staining results showed that compared to PBS-groups, vWF positive vessel and endoglin positive new vessels were increased in B10-transplanted MCAO groups in the lesion areas. The mRNA of angiogenesis factors including placental growth factor and hypoxia inducible factor (HIF)-1α were increased 3 days after MCAO in the core and IBZ areas of B10-transplanted group. Angiopoetin1 mRNA was increased only in the IBZ. Western blotting results showed that HIF-1α and vascular endothelial growth factor (VEGF) proteins were increased in B10-transplanted group. Both HIF-1α and VEGF were expressed in macrophage/microglia in the core area. In the IBZ, however, HIF-1α was expressed both in astrocytes and macrophage/microglia, while VEGF was expressed only in macrophage/microglia. Moreover, TGFβ protein levels were found to be increased in B10-transplanted group in the core and IBZ regions. Cell culture experiments using a human microglia cell line (HMO6) and B10 showed that IL-1β induced VEGF mRNA expression in both cell types. IL-1β was found to be highly expressed in B10 cells, and its co-culture with HMO6 further increased that in B10. Co-culture increased VEGF mRNA in both B10 and HMO6. In the rat brains, IL-1β was expressed in macrophage/microglia and transplanted-B10 cells in the core. IL-1β positive cell number was increased slightly, but significantly in B10-transplanted rats. To explore further, IL-1β expression was silenced in B10 cells by transfecting mRNA specific siRNA, and then transplanted in MCAO rats. Immunostaining result showed that endoglin positive area was decreased in IL-1β-silenced B10 transplanted groups compared to nonsilenced-B10 transplanted groups. Interestingly, vessel-like structure appeared as early as 3 days after MCAO in IL-1β-silenced B10-transplanted group. Thus our results demonstrated that B10 cells increased angiogenesis in MCAO rat model, through the regulation of HIF-1α and VEGF expression, where IL-1β might play a role.
Collapse
Affiliation(s)
- Abdullah Md Sheikh
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shozo Yano
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shingo Mitaki
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Md Ahsanul Haque
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Shuhei Yamaguchi
- Department of Neurology, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan
| | - Atsushi Nagai
- Department of Laboratory Medicine, Shimane University School of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan.
| |
Collapse
|
38
|
Reza-Zaldivar EE, Hernández-Sapiéns MA, Minjarez B, Gutiérrez-Mercado YK, Márquez-Aguirre AL, Canales-Aguirre AA. Potential Effects of MSC-Derived Exosomes in Neuroplasticity in Alzheimer's Disease. Front Cell Neurosci 2018; 12:317. [PMID: 30319358 PMCID: PMC6165870 DOI: 10.3389/fncel.2018.00317] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/30/2018] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia affecting regions of the central nervous system that exhibit synaptic plasticity and are involved in higher brain functions such as learning and memory. AD is characterized by progressive cognitive dysfunction, memory loss and behavioral disturbances of synaptic plasticity and energy metabolism. Cell therapy has emerged as an alternative treatment of AD. The use of adult stem cells, such as neural stem cells and Mesenchymal Stem Cells (MSCs) from bone marrow and adipose tissue, have the potential to decrease cognitive deficits, possibly by reducing neuronal loss through blocking apoptosis, increasing neurogenesis, synaptogenesis and angiogenesis. These processes are mediated primarily by the secretion of many growth factors, anti-inflammatory proteins, membrane receptors, microRNAs (miRNA) and exosomes. Exosomes encapsulate and transfer several functional molecules like proteins, lipids and regulatory RNA which can modify cell metabolism. In the proteomic characterization of the content of MSC-derived exosomes, more than 730 proteins have been identified, some of which are specific cell type markers and others are involved in the regulation of binding and fusion of exosomes with adjacent cells. Furthermore, some factors were found that promote the recruitment, proliferation and differentiation of other cells like neural stem cells. Moreover, within exosomal cargo, a wide range of miRNAs were found, which can control functions related to neural remodeling as well as angiogenic and neurogenic processes. Taking this into consideration, the use of exosomes could be part of a strategy to promote neuroplasticity, improve cognitive impairment and neural replacement in AD. In this review, we describe how exosomes are involved in AD pathology and discuss the therapeutic potential of MSC-derived exosomes mediated by miRNA and protein cargo.
Collapse
Affiliation(s)
- Edwin E Reza-Zaldivar
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Mercedes A Hernández-Sapiéns
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Benito Minjarez
- Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| | - Yanet K Gutiérrez-Mercado
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Ana L Márquez-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Alejandro A Canales-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico.,Profesor del programa de Maestría en Ciencias de la Salud Ambiental, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
39
|
HDAC4 in ischemic stroke: mechanisms and therapeutic potential. Clin Epigenetics 2018; 10:117. [PMID: 30208931 PMCID: PMC6136233 DOI: 10.1186/s13148-018-0549-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/28/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide, and the majority of the cases are ischemic stroke. However, it still lacks effective treatment except for thrombolytic therapy in an extremely narrow time window. Increased evidence suggests that histone deacetylase 4 (HDAC4) was dysregulated in ischemic stroke, which plays a key role in the pathogenesis of ischemic stroke and post-stroke recovery by affecting neuronal death, angiogenesis, and neurogenesis. Therefore, we aim to review the dysregulation of HDAC4 in ischemic stroke and the role of dysregulated HDAC4 in the pathogenesis of ischemic stroke. Furthermore, the therapeutic potential of modulating HDAC4 in ischemic stroke is discussed.
Collapse
|
40
|
Lu Y, Huang Z, Hua Y, Xiao G. Minocycline Promotes BDNF Expression of N2a Cells via Inhibition of miR-155-Mediated Repression After Oxygen-Glucose Deprivation and Reoxygenation. Cell Mol Neurobiol 2018; 38:1305-1313. [PMID: 29951932 PMCID: PMC11481882 DOI: 10.1007/s10571-018-0599-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
Minocycline, an anti-infective agent of a tetracycline derivative, is reported to improve behavioral functional recovery after cerebral ischemia via enhancing the levels of brain-derived neurotrophic factor (BDNF). However, the precise mechanisms that minocycline targets to enhance the expression of BDNF are not fully defined. In the present study, we observed the neuroprotective effect and its potential mechanisms of minocycline using oxygen-glucose deprivation/reoxygenation (OGD/R)-treated N2a cells. We found that 50 µM minocycline protected against neuronal apoptosis induced by OGD/R injury, with increased expression ratio of Bcl-2/Bax and reduced expression of caspase-3. Interestingly, minocycline resulted in the up-regulation of only BDNF protein, not BDNF mRNA in N2a cells treated with OGD/R. Furthermore, we found that minocycline inhibited OGD/R-induced up-regulation of miR-155 targeted BDNF transcripts. Moreover, miR-155 mimic could partially abolish the neuroprotective effects of minocycline via inhibiting the levels of BDNF protein. These findings suggest that minocycline is neuroprotective against ischemic brain injury through their modulation of miR-155-mediated BDNF repression.
Collapse
Affiliation(s)
- Yunnan Lu
- Department of Neurology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, 214000, Jiangsu, China
| | - Zhichao Huang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Rd, Gusu District, Suzhou, 215004, Jiangsu, China
| | - Ye Hua
- Department of Neurology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, 214000, Jiangsu, China
| | - Guodong Xiao
- Department of Neurology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Rd, Gusu District, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
41
|
Lin Z, Lin H, Li W, Huang Y, Dai H. Complement Component C3 Promotes Cerebral Ischemia/Reperfusion Injury Mediated by TLR2/NFκB Activation in Diabetic Mice. Neurochem Res 2018; 43:1599-1607. [PMID: 29948726 DOI: 10.1007/s11064-018-2574-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 05/16/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Complement component C3 (C3), a key factor in the complement system, is heavily involved in various inflammation-associated diseases. However, it remains obscure for its role in the pathogenesis of cerebral ischemia/reperfusion (I/R) injury in diabetes. A transient middle cerebral artery occlusion (tMCAO) model was used for cerebral I/R injury in streptozotocin-induced diabetic mice. Cerebral infarct volume and neurological function were measured at different times of reperfusion. Complement C3 was measured by ELISA and western blotting. It was observed that complement C3 expression was increased in cerebral I/R injury of diabetic mice, whereas complement C3 deficiency abrogated the activation and injury. Furthermore, activating complement C3 promotes TLR2/NFκB activation after I/R injury in diabetic mice, which is inhibited by of the silencing of TLR2. Taken together, our data demonstrate that complement C3 promotes cerebral I/R injury via the TLR2/NFκB pathway in diabetic mice, and regulating the complement C3/TLR2/NFκB pathway may be a novel target for therapeutic intervention in diabetic stroke.
Collapse
Affiliation(s)
- Zheng Lin
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Haoran Lin
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Wenlu Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yuwen Huang
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Haibin Dai
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
42
|
Chen M, Zou W, Chen M, Cao L, Ding J, Xiao W, Hu G. Ginkgolide K promotes angiogenesis in a middle cerebral artery occlusion mouse model via activating JAK2/STAT3 pathway. Eur J Pharmacol 2018; 833:221-229. [PMID: 29890157 DOI: 10.1016/j.ejphar.2018.06.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 11/26/2022]
Abstract
Ginkgolide K (GK) is a new compound extracted from the leaves of Ginkgo biloba, which has been recognized to exert anti-oxidative stress and neuroprotective effect on ischemic stroke. While whether it plays an enhanced effect on angiogenesis during ischemic stroke remains unknown. The aim of this study was to investigate the effect of ginkgolide K on promoting angiogenesis as well as the protective mechanism after cerebral ischemia-reperfusion. Using the transient middle cerebral artery occlusion (tMCAO) mouse model, we found that GK (3.5, 7.0, 14.0 mg/kg, i.p., bid., 2 weeks) attenuated neurological impairments, and promoted angiogenesis of injured ipsilateral cortex and striatum after 14 days of cerebral ischemia-reperfusion in mice. Further, GK (3.5 mg/kg in vivo, 10 μM in vitro) significantly up-regulated the expressions of HIF-1α and VEGF in tMCAO mouse brains and in b End3 cells after OGD/R, and GK-induced upregulation of HIF-1α and VEGF in b End3 cells could be abolished by JAK2/STAT3 inhibitor AG490. Our results demonstrate that GK promotes angiogenesis after ischemia stroke through increasing the expression of HIF-1α/VEGF via JAK2/STAT3 pathway, which provide an insight into the novel clinical application of GK and its analogs in ischemic stroke therapy in future.
Collapse
Affiliation(s)
- Meng Chen
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Wenyan Zou
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Miaomiao Chen
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Liang Cao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical CO. LTD., Lianyungang, Jiangsu 222001, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical CO. LTD., Lianyungang, Jiangsu 222001, China.
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China; Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
43
|
Chang YH, Wu KC, Harn HJ, Lin SZ, Ding DC. Exosomes and Stem Cells in Degenerative Disease Diagnosis and Therapy. Cell Transplant 2018; 27:349-363. [PMID: 29692195 PMCID: PMC6038041 DOI: 10.1177/0963689717723636] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stroke can cause death and disability, resulting in a huge burden on society. Parkinson’s disease (PD) is a chronic neurodegenerative disorder characterized by motor dysfunction. Osteoarthritis (OA) is a progressive degenerative joint disease characterized by cartilage destruction and osteophyte formation in the joints. Stem cell therapy may provide a biological treatment alternative to traditional pharmacological therapy. Mesenchymal stem cells (MSCs) are preferred because of their differentiation ability and possible derivation from many adult tissues. In addition, the paracrine effects of MSCs play crucial anti-inflammatory and immunosuppressive roles in immune cells. Extracellular vesicles (EVs) are vital mediators of cell-to-cell communication. Exosomes contain various molecules such as microRNA (miRNA), which mediates biological functions through gene regulation. Therefore, exosomes carrying miRNA or other molecules can enhance the therapeutic effects of MSC transplantation. MSC-derived exosomes have been investigated in various animal models representing stroke, PD, and OA. Exosomes are a subtype of EVs. This review article focuses on the mechanism and therapeutic potential of MSC-derived exosomes in stroke, PD, and OA in basic and clinical aspects.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- 1 Department of Pediatrics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.,2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Kung-Chi Wu
- 3 Department of Orthopedics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Horng-Jyh Harn
- 4 Department of Pathology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Shinn-Zong Lin
- 5 Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Dah-Ching Ding
- 2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,6 Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| |
Collapse
|
44
|
Tahsili-Fahadan P, Farrokh S, Geocadin RG. Hypothermia and brain inflammation after cardiac arrest. Brain Circ 2018; 4:1-13. [PMID: 30276330 PMCID: PMC6057700 DOI: 10.4103/bc.bc_4_18] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/17/2018] [Accepted: 03/18/2018] [Indexed: 12/14/2022] Open
Abstract
The cessation (ischemia) and restoration (reperfusion) of cerebral blood flow after cardiac arrest (CA) induce inflammatory processes that can result in additional brain injury. Therapeutic hypothermia (TH) has been proven as a brain protective strategy after CA. In this article, the underlying pathophysiology of ischemia-reperfusion brain injury with emphasis on the role of inflammatory mechanisms is reviewed. Potential targets for immunomodulatory treatments and relevant effects of TH are also discussed. Further studies are needed to delineate the complex pathophysiology and interactions among different components of immune response after CA and identify appropriate targets for clinical investigations.
Collapse
Affiliation(s)
- Pouya Tahsili-Fahadan
- Department of Medicine, Virginia Commonwealth University, Falls Church, Virginia, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Salia Farrokh
- Department of Pharmacy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Romergryko G Geocadin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
|
46
|
Maki T, Choi YK, Miyamoto N, Shindo A, Liang AC, Ahn BJ, Mandeville ET, Kaji S, Itoh K, Seo JH, Gelman IH, Lok J, Takahashi R, Kim KW, Lo EH, Arai K. A-Kinase Anchor Protein 12 Is Required for Oligodendrocyte Differentiation in Adult White Matter. Stem Cells 2018; 36:751-760. [PMID: 29314444 DOI: 10.1002/stem.2771] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/20/2017] [Accepted: 12/26/2017] [Indexed: 12/14/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes in cerebral white matter. However, the underlying mechanisms that regulate this process remain to be fully defined, especially in adult brains. Recently, it has been suggested that signaling via A-kinase anchor protein 12 (AKAP12), a scaffolding protein that associates with intracellular molecules such as protein kinase A, may be involved in Schwann cell homeostasis and peripheral myelination. Here, we asked whether AKAP12 also regulates the mechanisms of myelination in the CNS. AKAP12 knockout mice were compared against wild-type (WT) mice in a series of neurochemical and behavioral assays. Compared with WTs, 2-months old AKAP12 knockout mice exhibited loss of myelin in white matter of the corpus callosum, along with perturbations in working memory as measured by a standard Y-maze test. Unexpectedly, very few OPCs expressed AKAP12 in the corpus callosum region. Instead, pericytes appeared to be one of the major AKAP12-expressing cells. In a cell culture model system, conditioned culture media from normal pericytes promoted in-vitro OPC maturation. However, conditioned media from AKAP12-deficient pericytes did not support the OPC function. These findings suggest that AKAP12 signaling in pericytes may be required for OPC-to-oligodendrocyte renewal to maintain the white matter homeostasis in adult brain. Stem Cells 2018;36:751-760.
Collapse
Affiliation(s)
- Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yoon Kyung Choi
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Republic of Korea
| | - Nobukazu Miyamoto
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Akihiro Shindo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anna C Liang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Bum Ju Ahn
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Seiji Kaji
- Department of Neurology, Graduate School of Medicine, Kyoto University, Japan
| | - Kanako Itoh
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ji Hae Seo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences
- Department of Biochemistry, Keimyung University School of Medicine, Daegu, 42601, Korea
| | - Irwin H Gelman
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Japan
| | - Kyu-Won Kim
- NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 151-742, Korea
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Long non-coding RNA RMST silencing protects against middle cerebral artery occlusion (MCAO)-induced ischemic stroke. Biochem Biophys Res Commun 2018; 495:2602-2608. [DOI: 10.1016/j.bbrc.2017.12.087] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 12/15/2017] [Indexed: 01/05/2023]
|
48
|
The emerging field of epigenetics in neurodegeneration and neuroprotection. Nat Rev Neurosci 2017; 18:347-361. [PMID: 28515491 DOI: 10.1038/nrn.2017.46] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic mechanisms - including DNA methylation, histone post-translational modifications and changes in nucleosome positioning - regulate gene expression, cellular differentiation and development in almost all tissues, including the brain. In adulthood, changes in the epigenome are crucial for higher cognitive functions such as learning and memory. Striking new evidence implicates the dysregulation of epigenetic mechanisms in neurodegenerative disorders and diseases. Although these disorders differ in their underlying causes and pathophysiologies, many involve the dysregulation of restrictive element 1-silencing transcription factor (REST), which acts via epigenetic mechanisms to regulate gene expression. Although not somatically heritable, epigenetic modifications in neurons are dynamic and reversible, which makes them good targets for therapeutic intervention.
Collapse
|
49
|
Osawa S, Kurachi M, Yamamoto H, Yoshimoto Y, Ishizaki Y. Fibronectin on extracellular vesicles from microvascular endothelial cells is involved in the vesicle uptake into oligodendrocyte precursor cells. Biochem Biophys Res Commun 2017; 488:232-238. [PMID: 28499870 DOI: 10.1016/j.bbrc.2017.05.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 11/25/2022]
Abstract
We previously reported transplantation of brain microvascular endothelial cells (MVECs) into cerebral white matter infarction model improved the animal's behavioral outcome by increasing the number of oligodendrocyte precursor cells (OPCs). We also revealed extracellular vesicles (EVs) derived from MVECs promoted survival and proliferation of OPCs in vitro. In this study, we investigated the mechanism how EVs derived from MVECs contribute to OPC survival and proliferation. Protein mass spectrometry and enzyme-linked immunosorbent assay revealed fibronectin was abundant on the surface of EVs from MVECs. As fibronectin has been reported to promote OPC survival and proliferation via integrin signaling pathway, we blocked the binding between fibronectin and integrins using RGD sequence mimics. Blocking the binding, however, did not attenuate the survival and proliferation promoting effect of EVs on OPCs. Flow cytometric and imaging analyses revealed fibronectin on EVs mediates their internalization into OPCs by its binding to heparan sulfate proteoglycan on OPCs. OPC survival and proliferation promoted by EVs were attenuated by blocking the internalization of EVs into OPCs. These lines of evidence suggest that fibronectin on EVs mediates their internalization into OPCs, and the cargo of EVs promotes survival and proliferation of OPCs, independent of integrin signaling pathway.
Collapse
Affiliation(s)
- Sho Osawa
- Department of Neurosurgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hanako Yamamoto
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yuhei Yoshimoto
- Department of Neurosurgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
50
|
Tajiri N, Quach DM, Kaneko Y, Wu S, Lee D, Lam T, Hayama KL, Hazel TG, Johe K, Wu MC, Borlongan CV. NSI-189, a small molecule with neurogenic properties, exerts behavioral, and neurostructural benefits in stroke rats. J Cell Physiol 2017; 232:2731-2740. [PMID: 28181668 PMCID: PMC5518191 DOI: 10.1002/jcp.25847] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 02/07/2017] [Indexed: 12/26/2022]
Abstract
Enhancing neurogenesis may be a powerful stroke therapy. Here, we tested in a rat model of ischemic stroke the beneficial effects of NSI-189, an orally active, new molecular entity (mol. wt. 366) with enhanced neurogenic activity, and indicated as an anti-depressant drug in a clinical trial (Fava et al., , Molecular Psychiatry, DOI: 10.1038/mp.2015.178) and being tested in a Phase 2 efficacy trial (ClinicalTrials.gov, , ClinicalTrials.gov Identifier: NCT02695472) for treatment of major depression. Oral administration of NSI-189 in adult Sprague-Dawley rats starting at 6 hr after middle cerebral artery occlusion, and daily thereafter over the next 12 weeks resulted in significant amelioration of stroke-induced motor and neurological deficits, which was maintained up to 24 weeks post-stroke. Histopathological assessment of stroke brains from NSI-189-treated animals revealed significant increments in neurite outgrowth as evidenced by MAP2 immunoreactivity that was prominently detected in the hippocampus and partially in the cortex. These results suggest NSI-189 actively stimulated remodeling of the stroke brain. Parallel in vitro studies further probed this remodeling process and demonstrated that oxygen glucose deprivation and reperfusion (OGD/R) initiated typical cell death processes, which were reversed by NSI-189 treatment characterized by significant attenuation of OGD/R-mediated hippocampal cell death and increased Ki67 and MAP2 expression, coupled with upregulation of neurogenic factors such as BDNF and SCF. These findings support the use of oral NSI-189 as a therapeutic agent well beyond the initial 6-hr time window to accelerate and enhance the overall functional improvement in the initial 6 months post stroke.
Collapse
Affiliation(s)
- Naoki Tajiri
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| | | | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| | | | - David Lee
- Neuralstem, Inc., Rockville, Maryland
| | - Tina Lam
- Neuralstem, Inc., Rockville, Maryland
| | | | | | - Karl Johe
- Neuralstem, Inc., Rockville, Maryland
| | | | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University South Florida College of Medicine, Tampa, Florida
| |
Collapse
|