1
|
Dai M, Zhao Y, Jia Z, Xu S, Xu N, Wu X, Liu J, Wu L, Yu K, Lin X. Effect of Specific mode electroacupuncture stimulation combined with NGF during the ischaemic stroke: Study protocol for a randomized controlled trial. Clinics (Sao Paulo) 2024; 79:100451. [PMID: 39033586 PMCID: PMC11325668 DOI: 10.1016/j.clinsp.2024.100451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND The emergence of the Nerve Growth Factor (NGF) has promoted the development of neuroprotective therapy; however, it has little effect on cerebral ischemia because of its poor Blood-Brain Barrier (BBB) permeability. Specific Mode Electroacupuncture Stimulation (SMES) can open BBB safely and effectively; however, it has shown inconclusive clinical effects and indirect clinical evidence in the recovery phase. Hence, the authors conducted a multicentre, randomized, placebo-controlled, assessor-blinded clinical trial to assess the effectiveness and safety of SMES combined with NGF treatment used during ischaemic stroke recovery. METHODS A total of 288 stroke patients from three hospitals will be recruited and randomly allocated to four groups: acupuncture + placebo, acupuncture + NGF, SMES + placebo, and SMES + NGF, in a 1:1:1:1 ratio. Assessment data will be collected at baseline, 2-weeks, and 4-weeks during the treatment period, as well as at the 4-week and 8-week follow-up after treatment completion. The primary outcome measure will be the basic cure rate. The secondary outcome measures include the simplified Modified Barthel Index, Timed Up and Go Test, Fugl-Meyer Assessment of Motor Function Score, Tinetti Performance Oriented Mobility Assessment, Montreal Cognitive Assessment, and Loewenstein Occupational Therapy Cognitive Assessment. Moreover, resting-state functional magnetic resonance imaging and Functional near-infrared spectroscopy can detect changes in cerebral blood flow and brain function and investigate the relationship between the clinical efficacy and mechanism of the prescribed interventions. CONCLUSION This study will provide clinical evidence for the efficacy and safety of SMES combined with NGF in the treatment of stroke patients.
Collapse
Affiliation(s)
- Mengyuan Dai
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China; Department of Rehabilitation, The Second People's Hospital of Lishui, LiShui, ZheJiang Province, China
| | - Yibin Zhao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China; Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhaoxing Jia
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Shiting Xu
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Nuo Xu
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xuewen Wu
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jianxun Liu
- Department of Rehabilitation, Ningbo No.2 Hospital, Ningbo, Zhejiang Province, China
| | - Lixiu Wu
- Department of Rehabilitation, The Second People's Hospital of Lishui, LiShui, ZheJiang Province, China
| | - Kunqiang Yu
- Department of Rehabilitation, The Second People's Hospital of Lishui, LiShui, ZheJiang Province, China
| | - Xianming Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China; The Third Affiliated Hospital of Zhejiang Chinese Medical University, Xihu District, Zhejiang Province, China; Department of Rehabilitation, Zhejiang Rehabilitation Medical Center, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
2
|
Bernoud-Hubac N, Lo Van A, Lazar AN, Lagarde M. Ischemic Brain Injury: Involvement of Lipids in the Pathophysiology of Stroke and Therapeutic Strategies. Antioxidants (Basel) 2024; 13:634. [PMID: 38929073 PMCID: PMC11200865 DOI: 10.3390/antiox13060634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke is a devastating neurological disorder that is characterized by the sudden disruption of blood flow to the brain. Lipids are essential components of brain structure and function and play pivotal roles in stroke pathophysiology. Dysregulation of lipid signaling pathways modulates key cellular processes such as apoptosis, inflammation, and oxidative stress, exacerbating ischemic brain injury. In the present review, we summarize the roles of lipids in stroke pathology in different models (cell cultures, animal, and human studies). Additionally, the potential of lipids, especially polyunsaturated fatty acids, to promote neuroprotection and their use as biomarkers in stroke are discussed.
Collapse
Affiliation(s)
- Nathalie Bernoud-Hubac
- Univ Lyon, INSA Lyon, CNRS, LAMCOS, UMR5259, 69621 Villeurbanne, France; (A.L.V.); (A.-N.L.); (M.L.)
| | | | | | | |
Collapse
|
3
|
Wen J, Satyanarayanan SK, Li A, Yan L, Zhao Z, Yuan Q, Su KP, Su H. Unraveling the impact of Omega-3 polyunsaturated fatty acids on blood-brain barrier (BBB) integrity and glymphatic function. Brain Behav Immun 2024; 115:335-355. [PMID: 37914102 DOI: 10.1016/j.bbi.2023.10.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/05/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023] Open
Abstract
Alzheimer's disease (AD) and other forms of dementia represent major public health challenges but effective therapeutic options are limited. Pathological brain aging is associated with microvascular changes and impaired clearance systems. The application of omega-3 polyunsaturated fatty acids (n-3 or omega-3 PUFAs) is one of the most promising nutritional interventions in neurodegenerative disorders from epidemiological data, clinical and pre-clinical studies. As essential components of neuronal membranes, n-3 PUFAs have shown neuroprotection and anti-inflammatory effects, as well as modulatory effects through microvascular pathophysiology, amyloid-beta (Aβ) clearance and glymphatic pathways. This review meticulously explores these underlying mechanisms that contribute to the beneficial effects of n-3 PUFAs against AD and dementia, synthesizing evidence from both animal and interventional studies.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong
| | - Ang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Ziai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong
| | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan; Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan; Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan.
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| |
Collapse
|
4
|
Gong L, Liang J, Xie L, Zhang Z, Mei Z, Zhang W. Metabolic Reprogramming in Gliocyte Post-cerebral Ischemia/ Reperfusion: From Pathophysiology to Therapeutic Potential. Curr Neuropharmacol 2024; 22:1672-1696. [PMID: 38362904 PMCID: PMC11284719 DOI: 10.2174/1570159x22666240131121032] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/17/2024] Open
Abstract
Ischemic stroke is a leading cause of disability and death worldwide. However, the clinical efficacy of recanalization therapy as a preferred option is significantly hindered by reperfusion injury. The transformation between different phenotypes of gliocytes is closely associated with cerebral ischemia/ reperfusion injury (CI/RI). Moreover, gliocyte polarization induces metabolic reprogramming, which refers to the shift in gliocyte phenotype and the overall transformation of the metabolic network to compensate for energy demand and building block requirements during CI/RI caused by hypoxia, energy deficiency, and oxidative stress. Within microglia, the pro-inflammatory phenotype exhibits upregulated glycolysis, pentose phosphate pathway, fatty acid synthesis, and glutamine synthesis, whereas the anti-inflammatory phenotype demonstrates enhanced mitochondrial oxidative phosphorylation and fatty acid oxidation. Reactive astrocytes display increased glycolysis but impaired glycogenolysis and reduced glutamate uptake after CI/RI. There is mounting evidence suggesting that manipulation of energy metabolism homeostasis can induce microglial cells and astrocytes to switch from neurotoxic to neuroprotective phenotypes. A comprehensive understanding of underlying mechanisms and manipulation strategies targeting metabolic pathways could potentially enable gliocytes to be reprogrammed toward beneficial functions while opening new therapeutic avenues for CI/RI treatment. This review provides an overview of current insights into metabolic reprogramming mechanisms in microglia and astrocytes within the pathophysiological context of CI/RI, along with potential pharmacological targets. Herein, we emphasize the potential of metabolic reprogramming of gliocytes as a therapeutic target for CI/RI and aim to offer a novel perspective in the treatment of CI/RI.
Collapse
Affiliation(s)
- Lipeng Gong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Junjie Liang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Letian Xie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhanwei Zhang
- Department of Neurosurgery, First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410007, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei 443002, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
5
|
Zang J, Tang X, Su X, Zhang T, Lu D, Xu A. Systematic Analysis of RNA Expression Profiles in Different Ischemic Cortices in MCAO Mice. Cell Mol Neurobiol 2023; 43:859-878. [PMID: 35449428 PMCID: PMC11415153 DOI: 10.1007/s10571-022-01220-9] [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: 12/24/2021] [Accepted: 03/29/2022] [Indexed: 11/03/2022]
Abstract
The prognosis of ischemic stroke patients is highly associated with the collateral circulation. And the competing endogenous RNAs (ceRNAs) generated from different compensatory supply regions may also involve in the regulation of ischemic tissues prognosis. In this study, we found the apoptosis progress of ischemic neurons in posterior circulation-supplied regions (close to PCA, cortex2) was much slower than that in anterior circulation-supplied territory (close to ACA, cortex1) in MCAO-3-h mice. Using the RNA sequencing and functional enrichment analysis, we analyzed the difference between RNA expression profile in cortex1 and cortex2 and the related biological processes. The results indicated that the differential expressed ceRNAs in cortex1 were involved in cell process under acute injury, while the differential expressed ceRNAs in cortex2 was more likely to participate in long-term injury and repair process. Besides, by establishing the miRNA-ceRNA interaction network we further sorted out two specifically distributed miRNAs, namely mmu-miR446i-3p (in cortex1) and mmu-miR3473d (in cortex2). And the specifically increased mmu-miR3473d in cortex2 mainly involved the angiogenesis and cell proliferation after ischemic stroke, which may be the critical reason for the longer therapeutic time window in cortex2. In conclusion, the present study reported the specific changes of ceRNAs in distinct compensatory regions potentially involved in the evolution of cerebral ischemic tissues and the unbalance prognosis after stroke. It provided more evidence for the collateral compensatory effects on patients' prognosis and carried out the new targets for the ischemic stroke therapy.
Collapse
Affiliation(s)
- Jiankun Zang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xionglin Tang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xuanlin Su
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tianyuan Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dan Lu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China.
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China.
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
6
|
Jiang G, Li X, Liu M, Li H, Shen H, liao J, You W, Fang Q, Chen G. Remote ischemic postconditioning ameliorates stroke injury via the SDF-1α/CXCR4 signaling axis in rats. Brain Res Bull 2023; 197:31-41. [PMID: 36990325 DOI: 10.1016/j.brainresbull.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/14/2023] [Accepted: 03/26/2023] [Indexed: 03/29/2023]
Abstract
Remote Ischemic Postconditioning (RIPostC) has become a research hotspot due to its protective effect on the brain in clinical studies related to ischemic stroke. The purpose of this study is to investigate the protective effect of RIPostC after ischemic stroke in rats. The middle cerebral artery occlusion/reperfusion (MCAO/R) model was established by the wire embolization method. RIPostC was obtained by inducing temporary ischemia in the hind limbs of rats. First, based on the results of short-term behavioral measures and long-term neurological function experiments, RIPostC was found to have a protective effect on the MCAO/R model and to improve neurological recovery in rats. Compared to the sham group, RIPostC upregulated the expression levels of C-X-C motif chemokine receptor 4(CXCR4) in the brain and stromal cell-derived factor-1(SDF-1α) in peripheral blood. In addition, RIPostC upregulated CXCR4 expression on CD34+ stem cells in peripheral blood in flow cytometric assays. Meanwhile, according to the results of EdU/DCX co-staining and CD31 staining, it was found that the effect of RIPostC on ameliorating brain injury via SDF-1α/CXCR4 signaling axis may be associated with vascular neogenesis. Finally, after inhibiting the SDF-1α/CXCR4 signaling axis using AMD3100(Plerixafor), we found that the neuroprotective effect of RIPostC was diminished. Taken together, RIPostC can improve neurobehavioral damage induced by MCAO/R in rats, and its mechanism may be related to SDF-1α/CXCR4 signaling axis. Therefore, RIPostC can be used as an intervention strategy for stroke. SDF-1α/CXCR4 signaling axis can also be a potential target for intervention.
Collapse
|
7
|
Shi J, Li X, Ding J, Lian J, Zhong Y, Li H, Shen H, You W, Fu X, Chen G. Transient Receptor Potential Mucolipin-1 Participates in Intracerebral Hemorrhage-Induced Secondary Brain Injury by Inducing Neuroinflammation and Neuronal Cell Death. Neuromolecular Med 2023:10.1007/s12017-023-08734-5. [PMID: 36737508 DOI: 10.1007/s12017-023-08734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/05/2023] [Indexed: 02/05/2023]
Abstract
Transient receptor potential mucolipin-1 (TRPML1) is the most abundantly and widely expressed channel protein in the TRP family. While numerous studies have been conducted involving many aspects of TRPML1, such as its role in cell biology, oncology, and neurodegenerative diseases, there are limited reports about what role it plays in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI). Here we examined the function of TRPML1 in ICH-induced SBI. The caudal arterial blood of rats was injected into the caudate nucleus of basal ganglia to establish an experimental ICH model. We observed that lentivirus downregulated the expression level of TRPML1 and chemical agonist promoted the enzyme activity of TRPML1. The results indicated that the protein levels of TRPML1 in brain tissues increased 24 h after ICH. These results suggested that downregulated TRPML1 could significantly reduce inflammatory cytokines, and ICH induced the production of LDH and ROS. Furthermore, TRPML1 knockout relieved ICH-induced neuronal cell death and degeneration, and declines in learning and memory after ICH could be improved by downregulating the expression of TRPML1. In addition, chemical agonist-expressed TRPML1 showed the opposite effect and exacerbated SBI after ICH. In summary, this study demonstrated that TRPML1 contributed to brain injury after ICH, and downregulating TRPML1 could improve ICH-induced SBI, suggesting a potential target for ICH therapy.
Collapse
Affiliation(s)
- Jinzhao Shi
- Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, 215008, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Jiasheng Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Jinrong Lian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Yi Zhong
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Wanchun You
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China. .,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China.
| | - Xi'an Fu
- Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, 215008, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.,Institute of Stroke Research, Soochow University, Suzhou, 215006, Jiangsu Province, China
| |
Collapse
|
8
|
Li Q, Fadoul G, Ikonomovic M, Yang T, Zhang F. Sulforaphane promotes white matter plasticity and improves long-term neurological outcomes after ischemic stroke via the Nrf2 pathway. Free Radic Biol Med 2022; 193:292-303. [PMID: 36244590 DOI: 10.1016/j.freeradbiomed.2022.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 01/11/2023]
Abstract
AIMS Post-stroke cognitive impairment (PSCI) is a common condition following ischemic stroke. Neuronal loss and white matter injury are among the most common neuropathological characteristics in patients with PSCI. The present study tested our hypothesis that activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) reduces neuronal loss, white matter injury, and neurobehavioral deficits in a mouse model of PSCI and investigated the underlying protective mechanisms. METHODS PSCI was modeled in wildtype (WT) and Nrf2 knockout (KO), male and female mice, by distal middle cerebral artery occlusion (dMCAO), with intraperitoneal injections of the Nrf2 activator sulforaphane (Sfn) or vehicle. Long-term (35 days) sensorimotor and cognitive performances, white matter integrity, oligodendrogenesis by BrdU incorporation, and neurite sprouting using anterograde tract-tracing were evaluated up to 35 days after dMCAO. Neuronal apoptosis was evaluated three days after dMCAO. In vitro, primary neuronal cultures were applied to validate the in vivo findings. RESULTS Compared to vehicle-injected controls, Sfn treatment improved long-term sensorimotor and cognitive deficits after dMCAO in WT male and female mice. Sfn-treated WT mice also had less myelin loss/axonal injury and showed evidence of Nrf2 activation. Sfn treatment failed to provide the same level of protection in Nrf2 KO mice. Mechanistically, the ability of Sfn to reduce neuronal death after ischemia in vitro and in vivo, augment axonal sprouting and enhance oligodendrogenesis after dMCAO was dependent on Nrf2 activation. CONCLUSION Our results support that Nrf2 is critical for Sfn-afforded neuroprotection after ischemic stroke. Thus, targeting Nrf2 may be a promising strategy for the treatment of PSCI.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - George Fadoul
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Milos Ikonomovic
- Departments of Neurology and Psychiatry, University of Pittsburgh, USA; Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Feng Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Li Q, Lou J, Yang T, Wei Z, Li S, Zhang F. Ischemic Preconditioning Induces Oligodendrogenesis in Mouse Brain: Effects of Nrf2 Deficiency. Cell Mol Neurobiol 2022; 42:1859-1873. [PMID: 33666795 PMCID: PMC11421701 DOI: 10.1007/s10571-021-01068-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/23/2021] [Indexed: 10/22/2022]
Abstract
Ischemic preconditioning (IPC) is an approach of protection against cerebral ischemia by inducing endogenous cytoprotective machinery. However, few studies in neurogenesis and oligodendrogenesis after IPC have been reported, especially the latter. The purpose of this study is to test our hypothesis that IPC may also induce cell proliferation and oligodendrogenesis in the subventricular zone and striatum, as well as to investigate the effect of nuclear factor erythroid 2-related factor 2 (Nrf2) on oligodendrogenesis. IPC was induced in mice by 12-min ischemia through the occlusion of the middle cerebral artery. Newly generated cells were labeled with 5-bromo-2'-deoxyuridine. Our findings demonstrated that IPC stimulated the proliferation of neural stem cells in the subventricular zone, promoted the generation of oligodendrocyte precursor cells in the striatum and corpus callosum/external capsule (CC/EC), and stimulated oligodendrocyte precursor cells differentiation into oligodendrocytes in the striatum and the CC/EC. Furthermore, we describe a crucial role for Nrf2 in IPC-induced oligodendrogenesis in the subventricular zone, striatum, and CC/EC and show for the first time that Nrf2 promoted the migration and differentiation of oligodendrocyte precursor cells into oligodendrocytes in the striatum and CC/EC. Our data imply that IPC stimulates the oligodendrogenesis in the brain and that Nrf2 signaling may contribute to the oligodendrogenesis.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiyu Lou
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zhishuo Wei
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Senmiao Li
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Feng Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Chumak T, Lecuyer MJ, Nilsson AK, Faustino J, Ardalan M, Svedin P, Sjöbom U, Ek J, Obenaus A, Vexler ZS, Mallard C. Maternal n-3 Polyunsaturated Fatty Acid Enriched Diet Commands Fatty Acid Composition in Postnatal Brain and Protects from Neonatal Arterial Focal Stroke. Transl Stroke Res 2021; 13:449-461. [PMID: 34674145 PMCID: PMC9046339 DOI: 10.1007/s12975-021-00947-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/19/2021] [Accepted: 09/13/2021] [Indexed: 01/20/2023]
Abstract
The fetus is strongly dependent on nutrients from the mother, including polyunsaturated fatty acids (PUFA). In adult animals, n-3 PUFA ameliorates stroke-mediated brain injury, but the modulatory effects of different PUFA content in maternal diet on focal arterial stroke in neonates are unknown. This study explored effects of maternal n-3 or n-6 enriched PUFA diets on neonatal stroke outcomes. Pregnant mice were assigned three isocaloric diets until offspring reached postnatal day (P) 10–13: standard, long-chain n-3 PUFA (n-3) or n-6 PUFA (n-6) enriched. Fatty acid profiles in plasma and brain of mothers and pups were determined by gas chromatography–mass spectrometry and cytokines/chemokines by multiplex protein analysis. Transient middle cerebral artery occlusion (tMCAO) was induced in P9-10 pups and cytokine and chemokine accumulation, caspase-3 and calpain-dependent spectrin cleavage and brain infarct volume were analyzed. The n-3 diet uniquely altered brain lipid profile in naïve pups. In contrast, cytokine and chemokine levels did not differ between n-3 and n-6 diet in naïve pups. tMCAO triggered accumulation of inflammatory cytokines and caspase-3-dependent and -independent cell death in ischemic-reperfused regions in pups regardless of diet, but magnitude of neuroinflammation and caspase-3 activation were attenuated in pups on n-3 diet, leading to protection against neonatal stroke. In conclusion, maternal/postnatal n-3 enriched diet markedly rearranges neonatal brain lipid composition and modulates the response to ischemia. While standard diet is sufficient to maintain low levels of inflammatory cytokines and chemokines under physiological conditions, n-3 PUFA enriched diet, but not standard diet, attenuates increases of inflammatory cytokines and chemokines in ischemic-reperfused regions and protects from neonatal stroke.
Collapse
Affiliation(s)
- Tetyana Chumak
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | | | - Anders K Nilsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Faustino
- Department of Neurology, UCSF, San Francisco, CA, USA
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Pernilla Svedin
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Ulrika Sjöbom
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden
| | - Andre Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | | | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30, Gothenburg, Sweden. .,Department of Pediatrics, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
11
|
Rayasam A, Fukuzaki Y, Vexler ZS. Microglia-leucocyte axis in cerebral ischaemia and inflammation in the developing brain. Acta Physiol (Oxf) 2021; 233:e13674. [PMID: 33991400 DOI: 10.1111/apha.13674] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022]
Abstract
Development of the Central Nervous System (CNS) is reliant on the proper function of numerous intricately orchestrated mechanisms that mature independently, including constant communication between the CNS and the peripheral immune system. This review summarizes experimental knowledge of how cerebral ischaemia in infants and children alters physiological communication between leucocytes, brain immune cells, microglia and the neurovascular unit (NVU)-the "microglia-leucocyte axis"-and contributes to acute and long-term brain injury. We outline physiological development of CNS barriers in relation to microglial and leucocyte maturation and the plethora of mechanisms by which microglia and peripheral leucocytes communicate during postnatal period, including receptor-mediated and intracellular inflammatory signalling, lipids, soluble factors and extracellular vesicles. We focus on the "microglia-leucocyte axis" in rodent models of most common ischaemic brain diseases in the at-term infants, hypoxic-ischaemic encephalopathy (HIE) and focal arterial stroke and discuss commonalities and distinctions of immune-neurovascular mechanisms in neonatal and childhood stroke compared to stroke in adults. Given that hypoxic and ischaemic brain damage involve Toll-like receptor (TLR) activation, we discuss the modulatory role of viral and bacterial TLR2/3/4-mediated infection in HIE, perinatal and childhood stroke. Furthermore, we provide perspective of the dynamics and contribution of the axis in cerebral ischaemia depending on the CNS maturational stage at the time of insult, and modulation independently and in consort by individual axis components and in a sex dependent ways. Improved understanding on how to modify crosstalk between microglia and leucocytes will aid in developing age-appropriate therapies for infants and children who suffered cerebral ischaemia.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Neurology University of California San Francisco San Francisco CA USA
| | - Yumi Fukuzaki
- Department of Neurology University of California San Francisco San Francisco CA USA
| | - Zinaida S. Vexler
- Department of Neurology University of California San Francisco San Francisco CA USA
| |
Collapse
|
12
|
Xue T, Sun Q, Zhang Y, Wu X, Shen H, Li X, Wu J, Li H, Wang Z, Chen G. Phosphorylation at S548 as a Functional Switch of Sterile Alpha and TIR Motif-Containing 1 in Cerebral Ischemia/Reperfusion Injury in Rats. Mol Neurobiol 2021; 58:453-469. [PMID: 32968873 DOI: 10.1007/s12035-020-02132-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022]
Abstract
Sterile alpha and Toll/interleukin-1 receptor motif-containing 1 (SARM1) is a pro-degenerative molecule in Wallerian degeneration, which is mainly expressed in brain/neurons and colocalized with mitochondria and microtubules. The aim of this study was to determine the role of SARM1 in cerebral ischemia/reperfusion (I/R) injury and the underlying mechanisms. In vivo, a middle cerebral artery occlusion/reperfusion (MCAO/R) model in adult male Sprague Dawley rats (250-300 g) was established, and in vitro, cultured primary neurons were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to imitate I/R injury. Overexpression lentiviruses encoding wild-type SARM1 and SARM1 with serine 548 alanine mutation (S548A) were constructed and administered to rats by intra-penumbral injection. First, the potential role of SARM1 in cerebral I/R injury was confirmed by the increased protein levels of SARM1 within penumbra tissue, especially in neurons. Second, there was an increase in the phosphorylation ratio of p-SARM1(S548)/SARM1 at 2 h after MCAO/R. Third, on the basis of site-specific mutagenesis, we identified S548 as a key site for SARM1 phosphorylation in I/R conditions. Fourth, SARM1 (S548A) overexpression reduced infarct size, neuronal death, and neurobehavioral dysfunction, while wild-type SARM1 overexpression had the opposite effects. Finally, we found that SARM1 phosphorylation at the S548 site switched SARM1 function from promoting mitochondrial transport to inhibiting mitochondrial transport along axons after I/R injury. Restriction of SARM1 phosphorylation at S548 may be a promising intervention target for I/R injury; thus, exogenous administration of SARM1 (S548A) may be a novel strategy for improving neurological outcomes.
Collapse
Affiliation(s)
- Tao Xue
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yijie Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xin Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
13
|
Role of polyunsaturated fatty acids in ischemic stroke - A perspective of specialized pro-resolving mediators. Clin Nutr 2021; 40:2974-2987. [PMID: 33509668 DOI: 10.1016/j.clnu.2020.12.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) have been proposed as beneficial for cardiovascular health. However, results from both epidemiological studies and clinical trials have been inconsistent, whereas most of the animal studies showed promising benefits of PUFAs in the prevention and treatment of ischemic stroke. In recent years, it has become clear that PUFAs are metabolized into various types of bioactive derivatives, including the specialized pro-resolving mediators (SPMs). SPMs exert multiple biofunctions, such as to limit excessive inflammatory responses, regulate lipid metabolism and immune cell functions, decrease production of pro-inflammatory factors, increase anti-inflammatory mediators, as well as to promote tissue repair and homeostasis. Inflammation has been recognised as a key contributor to the pathophysiology of acute ischemic stroke. Owing to their potent pro-resolving actions, SPMs are potential for development of novel anti-stroke therapy. In this review, we will summarize current knowledge of epidemiological studies, basic research and clinical trials concerning PUFAs in stroke prevention and treatment, with special attention to SPMs as the unsung heroes behind PUFAs.
Collapse
|
14
|
Leal-Martínez F, Franco D, Peña-Ruiz A, Castro-Silva F, Escudero-Espinosa AA, Rolón-Lacarrier OG, López-Alarcón M, De León X, Linares-Eslava M, Ibarra A. Effect of a Nutritional Support System (Diet and Supplements) for Improving Gross Motor Function in Cerebral Palsy: An Exploratory Randomized Controlled Clinical Trial. Foods 2020; 9:E1449. [PMID: 33066040 PMCID: PMC7601161 DOI: 10.3390/foods9101449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Most patients with cerebral palsy (CP) do not respond to physical therapy due to deterioration in their nutritional status, secondary to gastrointestinal disorders and the catabolic state of the disease itself. However, basic treatments only contemplate the energy requirements and do not consider supplementation with glutamine, zinc, selenium, colecalciferol, spirulina, omega 3 or even vegetal proteins. OBJECTIVE In this study, we determined the effect of using a nutritional support system (NSS): diet and supplements, on the gross motor function in children with CP with spastic diparesic and Gross Motor Function Classification System III (GMFCS III). METHODS An exploratory study was performed. Thirty patients (from 4 to 12 years old) were randomly assigned to: (1) dietary surveillance (FG), (2) deworming and WHO diet (CG), or (3) deworming and the NSS (IG). Gross motor function was evaluated using the gross motor function measure (GMFM) scale. RESULTS The IG-treated group presented a significant improvement in standing and walking parameters analyzed in the GMFM compared with FG and CG groups. Fifty percent of the IG-treated patients managed to walk, while in the other groups, no patients were able to walk. CONCLUSIONS The NSS used in the present work improves gross motor function and promotes walking in patients with CP.
Collapse
Affiliation(s)
- Fernando Leal-Martínez
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan 52786, Mexico; (F.L.-M.); (D.F.); (A.P.-R.); (X.D.L.); (M.L.-E.)
| | - Denise Franco
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan 52786, Mexico; (F.L.-M.); (D.F.); (A.P.-R.); (X.D.L.); (M.L.-E.)
| | - Andrea Peña-Ruiz
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan 52786, Mexico; (F.L.-M.); (D.F.); (A.P.-R.); (X.D.L.); (M.L.-E.)
| | - Fabiola Castro-Silva
- Departamento de Terapia Física, Centro de Rehabilitación e Inclusión Infantil Teletón (CRIT), Tlalnepantla de Baz 54010, Mexico; (F.C.-S.); (A.A.E.-E.)
| | - Andrea A. Escudero-Espinosa
- Departamento de Terapia Física, Centro de Rehabilitación e Inclusión Infantil Teletón (CRIT), Tlalnepantla de Baz 54010, Mexico; (F.C.-S.); (A.A.E.-E.)
| | - Oscar G. Rolón-Lacarrier
- Departamento de Investigación y Enseñanza, Centro de Rehabilitación e Inclusión Infantil Teletón (CRIT), Tlalnepantla de Baz 54010, Mexico;
| | - Mardia López-Alarcón
- Unidad de Investigación Médica en Nutrición, Hospital de Pediatría CMN siglo XXI, Ciudad de Mexico 06720, Mexico;
| | - Ximena De León
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan 52786, Mexico; (F.L.-M.); (D.F.); (A.P.-R.); (X.D.L.); (M.L.-E.)
| | - Mariana Linares-Eslava
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan 52786, Mexico; (F.L.-M.); (D.F.); (A.P.-R.); (X.D.L.); (M.L.-E.)
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan 52786, Mexico; (F.L.-M.); (D.F.); (A.P.-R.); (X.D.L.); (M.L.-E.)
| |
Collapse
|
15
|
Zhào H, Wang R, Zhang Y, Liu Y, Huang Y. Neuroprotective effects of troxerutin and cerebroprotein hydrolysate injection on the neurovascular unit in a rat model of Middle cerebral artery occlusion. Int J Neurosci 2020; 131:264-278. [PMID: 32125198 DOI: 10.1080/00207454.2020.1738431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Purpose: Cerebral ischemic stroke, caused by obstruction of the blood flow to the brain, initiates a complex cascade of pathophysiological changes. The aim of the present study was to assess the protective role and the underlying mechanism of troxerutin and cerebroprotein hydrolysate (TCH) injections for five days in rats subjected to middle cerebral artery occlusion (MCAO).Materials and Methods: Male Sprague-Dawley rats treated with either TCH or a vehicle (0.9% saline) via intraperitoneal injection were examined one or three days after MCAO.Results: TCH alleviated neurological deficits and reduced infarct volume, innate immune response, blood-brain barrier destruction, and suppressed cell apoptosis. The therapeutic effects of TCH were achieved by diminished neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS), and increased endothelial nitric oxide synthase (eNOS). Furthermore, L-NAME showed an inhibitory effect against TCH after MCAO on eNOS expression, NO and peroxynitrite production, neurobehavioral score, and infarct volume.Conclusions: The results indicate that injection of TCH has multifaceted neuroprotective effects against MCAO via regulation of the various NOS isoforms.
Collapse
Affiliation(s)
- Hóngyi Zhào
- Department of Neurology, The Seventh Medical Centre of PLA General Hospital, Beijing, China.,Department of Neurology, NO 984 Hospital of the PLA, Beijing, China
| | - Ru Wang
- Department of Neurology, 2nd Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yan Zhang
- Affiliated Bayi Brain Hospital, the Seventh Medical Centre of PLA General Hospital, Beijing, China
| | - Yu Liu
- Department of Neurology, NO 984 Hospital of the PLA, Beijing, China
| | - Yonghua Huang
- Department of Neurology, The Seventh Medical Centre of PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Yu Y, Fang H, Qiu Z, Xia Z, Zhou B. DHA Attenuates Hypoxia/Reoxygenation Injury by Activating SSeCKS in Human Cerebrovascular Pericytes. Neurochem Res 2019; 45:310-321. [PMID: 31776970 PMCID: PMC6985071 DOI: 10.1007/s11064-019-02915-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/07/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022]
Abstract
Docosahexaenoic acid (DHA) can alleviate cerebral ischemia/reperfusion injury by reducing blood–brain barrier permeability and maintaining its integrity, accompanied by an increased Ang-1/Ang-2 ratio; however, the underlying mechanisms of these effects remain unclear. Src-suppressed C kinase substrates (SSeCKS), a substrate of protein kinase C, plays an important role in maintaining cell junctions and cell morphology and regulating cell permeability. However, whether DHA can increase SSeCKS expression and then mediate the Ang-1/Ang-2 ratio still needs to be studied. Human cerebrovascular pericytes (HBVPs) cultured in vitro were divided into groups, treated with or without DHA along with SSeCKS siRNA to knockdown SSeCKS expression, and then subjected to 24 h of hypoxia followed by 6 h of reoxygenation. Cell viability; lactate dehydrogenase (LDH) release; and Ang-1, Ang-2 and VEGF activity were detected by using ELISA kits. The apoptosis rate was assessed by TUNEL flow cytometry. Expression of the SSeCKS, Ang-1, Ang-2 and VEGF proteins was evaluated by western blotting. Pretreatment with 10 μM or 40 μM DHA efficiently attenuated hypoxia/reoxygenation (H/R) injury by activating SSeCKS to increase the Ang-1/Ang-2 ratio and downregulate VEGF expression in HBVPs, as evidenced by decreased LDH release and apoptotic rates and increased HBVPs viability. Meanwhile, after we used SSeCKS siRNA to knock down SSeCKS protein expression, the protective effect of DHA on HBVPs following H/R injury was reversed. In conclusion, DHA can activate SSeCKS to increase the Ang-1/Ang-2 ratio and downregulate VEGF expression in HBVPs, thus reducing H/R injury.
Collapse
Affiliation(s)
- Yanli Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Haibin Fang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bin Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
17
|
Bu F, Min JW, Munshi Y, Lai YJ, Qi L, Urayama A, McCullough LD, Li J. Activation of endothelial ras-related C3 botulinum toxin substrate 1 (Rac1) improves post-stroke recovery and angiogenesis via activating Pak1 in mice. Exp Neurol 2019; 322:113059. [PMID: 31499064 DOI: 10.1016/j.expneurol.2019.113059] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE Long-term disability after stroke is common yet the mechanisms of post-stroke recovery are far from clear. It has been suggested that Ras-related C3 botulinum toxin substrate 1 (Rac1) contributes to functional recovery after ischemic stroke in mice. As Rac1 activation plays diverse roles in multiple cell types after central nervous system (CNS) injury, we herein examined the functional role of endothelial Rac1 in post-stroke recovery and angiogenesis. METHODS Transient middle cerebral artery occlusion (MCAO) in mice and oxygen-glucose deprivation (OGD) in human brain endothelial cell line-5i (HEBC 5i) were performed to mimic ischemic stroke. Lentivirus vectors encoding Rac1 with GFP and endothelial promotor ENG were injected into the animal's brain after stroke to overexpress Rac1. After injection, stroke recovery was tested by multiple behavioral tests including novel object recognition, adhesive removal and single pellet reaching tests. Endothelial regeneration in the peri-infarct zone was detected by immunohistochemistry (IHC). In the vitro model, the effect of Rac1 and Pak1 inhibitors to cell proliferation and migration was examined by CCK-8 and wound healing assays after OGD. The cellular protein level of brain-derived neurotrophic factor (BDNF), phosphorylated cAMP response element-binding protein (CREB), extracellular signal-regulated kinase (ERK) 1/2 and mitogen-activated protein kinase kinase (MEK) 1/2 were detected by western blots. RESULTS Delayed overexpression of endothelial Rac1 after MCAO improved cognitive and sensorimotor recovery from day 14 to 21 after stroke, increased vascular density and the protein level of pericytes in the peri-infarct zone without altering tissue loss in mice. Consistently, inhibition of Rac1 prevented endothelial proliferation and migration after OGD. Pak1 inhibition exerted a similar effect on endothelial cells. However, co-incubation of Rac1 and Pak1 inhibitors with cells did not lead to additive effects when compared with either inhibitor alone. Moreover, individual inhibition of Rac1 or Pak1 suppressed OGD-induced activation of pro-regenerative molecules, including CREB, MEK1/2 and ERK1/2, as well as the production of BDNF in vitro. The level of these proteins did not further decrease if both Rac1 and Pak1 were simultaneously inhibited. CONCLUSIONS We conclude that activation of endothelial Rac1 improves functional recovery and angiogenesis after stroke, and this process is mediated by Pak1 signaling. This study provides novel insight for Rac1 in the mechanism of long-term stroke recovery.
Collapse
Affiliation(s)
- Fan Bu
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Jia-Wei Min
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Yashasvee Munshi
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Yun-Ju Lai
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Li Qi
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | - Jun Li
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
18
|
Zheng W, Matei N, Pang J, Luo X, Song Z, Tang J, Zhang JH. Delayed recanalization at 3 days after permanent MCAO attenuates neuronal apoptosis through FGF21/FGFR1/PI3K/Caspase-3 pathway in rats. Exp Neurol 2019; 320:113007. [PMID: 31295445 DOI: 10.1016/j.expneurol.2019.113007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/06/2019] [Accepted: 07/04/2019] [Indexed: 12/19/2022]
Abstract
Reperfusion exceeded time window may induce ischemia/reperfusion injury, increase hemorrhagic transformation, and deteriorate neurological outcomes in ischemic stroke models. However, the increasing clinical evidences supported that reperfusion even within 6-24 h may salvage ischemic tissue and improve neurological outcomes in selected large vessel occlusion patients, without inducing serious ischemia/reperfusion injury and hemorrhagic transformation. The underlying molecular mechanisms are less clear. In present study, we demonstrated that delayed recanalization at 3 days after permanent middle cerebral artery occlusion (MCAO) decreased infarct volumes and improved neurobehavioral deficits in rats, with no increasing animal mortality and intracerebral hemorrhage. Meanwhile, we observed that endogenous neuroprotective agent fibroblast growth factor 21 (FGF21) significantly increased in serum after MCAO, but which did not synchronously increase in penumbra due to permanent MCAO. Recanalization dramatically increased the endogenous FGF21 expression on neurons in penumbra after MCAO. We confirmed that FGF21 activated the FGFR1/PI3K/Caspase-3 signaling pathway, which attenuated neuronal apoptosis in penumbra. Conversely, knockdown of FGFR1 via FGFR1 siRNA abolished the anti-apoptotic effects of FGF21, and in part abrogated beneficial effects of recanalization on neurological outcomes. These findings suggested that delayed recanalization at 3 days after MCAO improved neurological outcomes in rats via increasing endogenous FGF21 expression and activating FGFR1/PI3K/Caspase-3 pathway to attenuate neuronal apoptosis in penumbra. Delayed recanalization at 3 days after ischemic stroke onset may be a promising treatment strategy in selected patients.
Collapse
Affiliation(s)
- Wen Zheng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Department of Anesthesiology, Neurosurgery and Neurology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Nathanael Matei
- Department of Anesthesiology, Neurosurgery and Neurology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jinwei Pang
- Department of Anesthesiology, Neurosurgery and Neurology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Xu Luo
- Department of Anesthesiology, Neurosurgery and Neurology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Zhi Song
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - John H Zhang
- Department of Anesthesiology, Neurosurgery and Neurology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA.
| |
Collapse
|
19
|
Cannabinoid signalling in embryonic and adult neurogenesis: possible implications for psychiatric and neurological disorders. Acta Neuropsychiatr 2019; 31:1-16. [PMID: 29764526 DOI: 10.1017/neu.2018.11] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cannabinoid signalling modulates several aspects of brain function, including the generation and survival of neurons during embryonic and adult periods. The present review intended to summarise evidence supporting a role for the endocannabinoid system on the control of neurogenesis and neurogenesis-dependent functions. Studies reporting participation of cannabinoids on the regulation of any step of neurogenesis and the effects of cannabinoid compounds on animal models possessing neurogenesis-dependent features were selected from Medline. Qualitative evaluation of the selected studies indicated that activation of cannabinoid receptors may change neurogenesis in embryonic or adult nervous systems alongside rescue of phenotypes in animal models of different psychiatric and neurological disorders. The text offers an overview on the effects of cannabinoids on central nervous system development and the possible links with psychiatric and neurological disorders such as anxiety, depression, schizophrenia, brain ischaemia/stroke and Alzheimer's disease. An understanding of the mechanisms by which cannabinoid signalling influences developmental and adult neurogenesis will help foster the development of new therapeutic strategies for neurodevelopmental, psychiatric and neurological disorders.
Collapse
|
20
|
Use of Nutraceuticals in Angiogenesis-Dependent Disorders. Molecules 2018; 23:molecules23102676. [PMID: 30340320 PMCID: PMC6222874 DOI: 10.3390/molecules23102676] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022] Open
Abstract
The term of angiogenesis refers to the growth of new vessels from pre-existing capillaries. The phenomenon is necessary for physiological growth, repair and functioning of our organs. When occurring in a not regulated manner, it concurs to pathological conditions as tumors, eye diseases, chronic degenerative disorders. On the contrary insufficient neovascularization or endothelial disfunction accompanies ischemic and metabolic disorders. In both the cases an inflammatory and oxidative condition exists in supporting angiogenesis deregulation and endothelial dysfunction. The use of nutraceuticals with antioxidant and anti-inflammatory activities can be a therapeutic option to maintain an adequate vascularization and endothelial cell proper functioning or to blunt aberrant angiogenesis. A revision of the updated literature reports on nutraceuticals to guide endothelial cell wellness and to restore physiological tissue vascularization is the objective of this paper. The critical aspects as well as lacking data for human use will be explored from a pharmacological perspective.
Collapse
|
21
|
Zhang J, Chen S, Shi W, Li M, Zhan Y, Yang L, Zou H, Lei J, Chai X, Gao K, Liu J, Wang W, Wang Y, Zhao H. Effects of Xiaoshuan Enteric-Coated Capsule on White and Gray Matter Injury Evaluated by Diffusion Tensor Imaging in Ischemic Stroke. Cell Transplant 2018; 28:671-683. [PMID: 30284459 PMCID: PMC6686435 DOI: 10.1177/0963689718802755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Xiaoshuan enteric-coated capsule (XSECC) is a drug approved by the Chinese State Food and Drug Administration for the treatment of stroke. This study was to investigate the effects of XSECC on white and gray matter injury in a rat model of ischemic stroke by diffusion tensor imaging (DTI) and histopathological analyses. The ischemia was induced by middle cerebral artery occlusion (MCAO). The cerebral blood flow measured by arterial spin labeling was improved by treatment with XSECC on the 3rd, 7th, 14th and 30th days after MCAO. Spatiotemporal white and gray matter changes in MCAO rats were examined with DTI-derived parameters (fractional anisotropy, FA; apparent diffusion coefficient, ADC; axial diffusivity, λ//; radial diffusivity, λ⊥). The increased FA was found in the XSECC treatment group in the corpus callosum, external capsule and internal capsule, linked with the decreased λ//, λ⊥ and ADC on the 3rd day and reduced ADC on the 30th day in the external capsule, suggesting XSECC reduced the axon and myelin damage in white matter after stroke. The relative FA in the striatum, cortex and thalamus in XSECC treatment group was significantly increased on the 3rd, 7th, 14th and 30th days accompanied by the increased λ// on the 3rd day and reduced relative ADC and λ⊥ on the 30th day, indicating that XSECC attenuated cell swelling and membrane damage in the early stage and tissue liquefaction necrosis in the late stage in gray matter after stroke. Additionally, XSECC-treated rats exhibited increased mean fiber length assessed by diffusion tensor tractography. Moreover, histopathological analyses provided evidence that XSECC relieved nerve cell and myelin damage in white and gray matter after stroke. Our research reveals that XSECC could alleviate white and gray matter injury, especially reducing nerve cell damage and promoting the repair of axon and myelin after ischemic stroke.
Collapse
Affiliation(s)
- Jian Zhang
- 1 School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,2 School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shengpan Chen
- 3 Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| | - Weilong Shi
- 4 Pharmacy Department, Peking University Third Hospital, Beijing, China
| | - Manzhong Li
- 1 School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yu Zhan
- 1 School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Le Yang
- 1 School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Haiyan Zou
- 1 School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jianfeng Lei
- 1 School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xinlou Chai
- 2 School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kuo Gao
- 2 School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junjie Liu
- 5 Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- 2 School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Wang
- 2 School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,6 School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Zhao
- 1 School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Post-stroke administration of omega-3 polyunsaturated fatty acids promotes neurovascular restoration after ischemic stroke in mice: Efficacy declines with aging. Neurobiol Dis 2018; 126:62-75. [PMID: 30218758 DOI: 10.1016/j.nbd.2018.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 01/11/2023] Open
Abstract
Post-stroke treatment with omega-3 polyunsaturated fatty acids (n-3 PUFAs) may be a promising therapy in young animals but this has not been tested in aged subjects, a population at most risk of ischemic stroke. Herein we examined the therapeutic efficacy of n-3 PUFAs after distal middle cerebral artery occlusion (dMCAO) in young (10-12 weeks old) and aged (18 months old) mice. Post-ischemic mice were randomly assigned to 4 groups that received: 1) regular food with low content of n-3 PUFAs, 2) intraperitoneal docosahexaenoic acid (DHA, a major component of n-3 PUFAs) injections, 3) Fish oil (FO, containing high concentration of n-3 PUFAs) dietary supplement, or 4) combined treatment with DHA and FO dietary supplement. Long-term neurorestoration induced by n-3 PUFA post-stroke administration and its underlying mechanism(s) were analyzed up to 35 days after dMCAO. Aged mice showed more severe neurological deficits than young mice after dMCAO with histological lesions extended to the striatum. Notably, post-stroke treatment with combined DHA injections and FO dietary supplementation was more effective in reducing brain injury and improving sensorimotor function in aged mice than either treatment alone, albeit to a lesser extent than in the young mice. Unlike the improvement in spatial cognitive function observed in young mice, the combined treatment regimen failed to improve cognitive function in aged mice. The reduction in stroke-induced neurological deficits with n-3 PUFA post-treatment was associated with enhanced angiogenesis, oligodendrogenesis, neuron survival and white matter restoration. Together, these results indicate that the neurological benefits of n-3 PUFA administration after stroke extend to older animals and are associated with improved neuronal survival and brain remodeling, therefore suggesting that post-stroke administration of n-3 PUFAs is a viable clinically relevant treatment option against stroke.
Collapse
|
23
|
Post-stroke DHA Treatment Protects Against Acute Ischemic Brain Injury by Skewing Macrophage Polarity Toward the M2 Phenotype. Transl Stroke Res 2018; 9:669-680. [PMID: 30203370 DOI: 10.1007/s12975-018-0662-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
Systemic docosahexaenoic acid (DHA) has been explored as a clinically feasible protectant in stroke models. However, the mechanism for DHA-afforded neuroprotection remains elusive. Transient middle cerebral artery occlusion (tMCAO) was induced for 1 h. DHA (i.p., 10 mg/kg) was administered immediately after reperfusion and repeated daily for 3 days. Stroke outcomes, systemic inflammatory status, and microglia/macrophage phenotypic alterations were assessed 3 days after stroke. Macrophage depletion was induced by clodronate liposomes injection. Primary macrophage cultures were used to evaluate the direct effect of DHA on macrophages. We demonstrated that post-stroke DHA injection efficiently reduced brain infarct and ameliorated neurological deficits 3 days after tMCAO. Systemic DHA treatment significantly inhibited immune cell infiltration (macrophages, neutrophils, T lymphocytes, and B lymphocytes) and promoted macrophage polarization toward an anti-inflammatory M2 phenotype in the ischemic brain. Meanwhile, systemic DHA administration inhibited the otherwise elevated pro-inflammatory factors in blood and shifted circulating macrophage polarity toward M2 phenotype after ischemic stroke. The numbers of circulating immune cells in blood and spleen, however, were equivalent between DHA- and vehicle-treated groups. The protective effects of DHA were macrophage-dependent, as macrophage depletion abolished DHA-afforded neuroprotection. In vitro studies confirmed that DHA suppressed production of chemokines and pro-inflammatory cytokines from macrophages under inflammatory stimulation. These data indicate that post-stroke DHA treatment ameliorated acute ischemic brain injury in a macrophage-dependent manner and DHA enhanced macrophage phenotypic shift toward an anti-inflammatory phenotype to reduced central and peripheral inflammation after stroke.
Collapse
|
24
|
Turcato F, Kim P, Barnett A, Jin Y, Scerba M, Casey A, Selman W, Greig NH, Luo Y. Sequential combined Treatment of Pifithrin-α and Posiphen Enhances Neurogenesis and Functional Recovery After Stroke. Cell Transplant 2018; 27:607-621. [PMID: 29871513 PMCID: PMC6041885 DOI: 10.1177/0963689718766328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective: Although cerebral ischemia can activate endogenous reparative processes, such as
proliferation of endogenous neural stem cells (NSCs) in the subventricular zone (SVZ)
and subgranular zone (SGZ), the majority of these new cells die shortly after injury and
do not appropriately differentiate into neurons, or migrate and functionally integrate
into the brain. The purpose of this study was to examine a novel strategy for treatment
of stroke after injury by optimizing the survival of ischemia-induced endogenous NSCs in
the SVZ and SGZ. Methods: Adult SVZ and SGZ NSCs were grown as neurospheres in culture and treated with a p53
inactivator, pifithrin-α (PFT-α), and an amyloid precursor protein (APP)-lowering drug,
posiphen, and effects on neurosphere number, size and neuronal differentiation were
evaluated. This combined sequential treatment approach was then evaluated in mice
challenged with middle cerebral artery occlusion (MCAo). Locomotor behavior and
cognition were evaluated at 4 weeks, and the number of new surviving neurons was
quantified in nestin creERT2-YFP mice. Results: PFT-α and posiphen enhanced the self-renewal, proliferation rate and neuronal
differentiation of adult SVZ and SGZ NSCs in culture. Their sequential combination in
mice challenged with MCAo-induced stroke mitigated locomotor and cognitive impairments
and increased the survival of SVZ and SGZ NSCs cells. PFT-α and the combined
posiphen+PFT-α treatment similarly improved locomotion behavior in stroke challenged
mice. Notably, however, the combined treatment provided significantly more potent
cognitive function enhancement in stroke mice, as compared with PFT-α single
treatment. Interpretation: Delayed combined sequential treatment with an inhibitor of p53 dependent apoptosis
(PFT-α) and APP synthesis (posiphen) proved able to enhance stroke-induced endogenous
neurogenesis and improve the functional recovery in stroke animals. Whereas the combined
sequential treatment provided no further improvement in locomotor function, as compared
with PFT-α alone treatment, suggesting a potential ceiling in the locomotion behavioral
outcome in stroke animals, combined treatment more potently augmented cognitive function
recovery after stroke.
Collapse
Affiliation(s)
- Flavia Turcato
- 1 Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA.,2 Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Paul Kim
- 1 Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Austin Barnett
- 1 Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Yongming Jin
- 1 Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Mike Scerba
- 3 National Institute of Aging, Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, Baltimore, USA
| | - Anthony Casey
- 1 Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Warren Selman
- 1 Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| | - Nigel H Greig
- 3 National Institute of Aging, Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, Baltimore, USA
| | - Yu Luo
- 1 Department of Neurological Surgery, Case Western Reserve University, Cleveland, USA
| |
Collapse
|
25
|
Docosanoids Promote Neurogenesis and Angiogenesis, Blood-Brain Barrier Integrity, Penumbra Protection, and Neurobehavioral Recovery After Experimental Ischemic Stroke. Mol Neurobiol 2018; 55:7090-7106. [PMID: 29858774 DOI: 10.1007/s12035-018-1136-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/18/2018] [Indexed: 02/07/2023]
Abstract
Docosahexaenoic acid (DHA) and neuroprotectin D1 (NPD1) are neuroprotective after experimental ischemic stroke. To explore underlying mechanisms, SD rats underwent 2 h of middle cerebral artery occlusion (MCAo) and treated with DHA (5 mg/kg, IV) or NPD1 (5 μg/per rat, ICV) and vehicles 1 h after. Neuro-behavioral assessments was conducted on days 1, 2, and 3, and on week 1, 2, 3, or 4. BrdU was injected on days 4, 5, and 6, immunohistochemistry was performed on week 2 or 4, MRI on day 7, and lipidomic analysis at 4 and 5 h after onset of stroke. DHA improved short- and long-term behavioral functions and reduced cortical, subcortical, and total infarct volumes (by 42, 47, and 31%, respectively) after 2 weeks and reduced tissue loss by 50% after 4 weeks. DHA increased the number of BrdU+/Ki-67+, BrdU+/DCX+, and BrdU+/NeuN+ cells in the cortex, subventricular zone, and dentate gyrus and potentiated NPD1 synthesis in the penumbra at 5 h after MCAo. NPD1 improved behavior, reduced lesion volumes, protected ischemic penumbra, increased NeuN, GFAP, SMI-71-positive cells and vessels, axonal regeneration in the penumbra, and attenuated blood-brain barrier (BBB) after MCAo. We conclude that docosanoid administration increases neurogenesis and angiogenesis, activates NPD1 synthesis in the penumbra, and diminishes BBB permeability, which correlates to long-term neurobehavioral recovery after experimental ischemic stroke.
Collapse
|
26
|
Ras-Related C3 Botulinum Toxin Substrate 1 Promotes Axonal Regeneration after Stroke in Mice. Transl Stroke Res 2018; 9:506-514. [PMID: 29476448 DOI: 10.1007/s12975-018-0611-5] [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: 11/02/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/18/2022]
Abstract
Neurite plasticity is a critical aspect of brain functional recovery after stroke. Emerging data suggest that Ras-related C3 botulinum toxin substrate 1 (Rac1) plays a central role in axonal regeneration in the injured brain, specifically by stimulating neuronal intrinsic growth and counteracting the growth inhibitory signaling that leads to growth cone collapse. Therefore, we investigated the functional role of Rac1 in axonal regeneration after stroke.Delayed treatment with a specific Rac1 inhibitor, NSC 23766, worsened functional recovery, which was assessed by the pellet reaching test from day 14 to day 28 after stroke. It additionally reduced axonal density in the peri-infarct zone, assessed 28 days after stroke, with no effect on brain cavity size or on the number of newly formed cells. Accordingly, Rac1 overexpression using lentivirus promoted axonal regeneration and functional recovery after stroke from day 14 to day 28. Rac1 inhibition led to inactivation of pro-regenerative molecules, including mitogen-activated protein kinase kinase (p-MEK)1/2, LIM domain kinase (LIMK)1, and extracellular signal-regulated kinase (p-ERK)1/2 at 14 days after stroke. Inhibition of Rac1 reduced axonal length and number in cultured primary mouse cortical neurons using microfluidic chambers after oxygen-glucose deprivation (OGD) without affecting cell viability. In contrast, inhibition of Rac1 increased levels of glial fibrillary acidic protein, an extrinsic inhibitory signal for axonal growth, after stroke in vivo and in primary astrocytes after OGD.In conclusion, Rac1 signaling enhances axonal regeneration and improve post-stroke functional recovery in experimental models of stroke.
Collapse
|
27
|
Zhang M, Wu X, Xu Y, He M, Yang J, Li J, Li Y, Ao G, Cheng J, Jia J. The cystathionine β-synthase/hydrogen sulfide pathway contributes to microglia-mediated neuroinflammation following cerebral ischemia. Brain Behav Immun 2017; 66:332-346. [PMID: 28751019 DOI: 10.1016/j.bbi.2017.07.156] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/09/2017] [Accepted: 07/24/2017] [Indexed: 11/16/2022] Open
Abstract
The mechanisms underlying neuroinflammation following cerebral ischemia remain unclear. Hydrogen sulfide (H2S), a newly identified gasotransmitter, has been reported to regulate inflammation. In the current study, we investigated whether the endogenous H2S production pathway contributed to microglia-mediated neuroinflammation following stroke. We used a mouse middle cerebral artery occlusion (MCAO) model and an in vitro cellular model to mimic ischemia-induced microglial neuroinflammation. Expression of the H2S synthase cystathionine β-synthase (CBS) and H2S synthetic activity were rapidly decreased in the ischemic brain tissue following MCAO. Consistently, when cultured microglia were polarized toward a pro-inflammatory phenotype with conditioned medium collected from neurons that had been subjected to oxygen-glucose deprivation (OGD neuron CM), they displayed reduced CBS expression and H2S production. Enhancing H2S bioavailability either by overexpressing CBS or by supplementing with exogenous H2S donors promoted a shift in microglial polarization from ischemia-induced pro-inflammatory phenotypes toward anti-inflammatory phenotypes. Mechanistically, microglia that were exposed to OGD neuron CM displayed reduced activation of AMP-activated protein kinase (AMPK), which was rescued by overexpressing CBS or by supplementing with H2S donors. Moreover, the promoting effects of H2S donors on microglial anti-inflammatory polarization were abolished by an AMPK inhibitor or CaMKKβ inhibitor. Our results suggested that reduced CBS-H2S-AMPK cascade activity contributed to microglia-mediated neuroinflammation following stroke. Targeting the CBS-H2S pathway is a promising therapeutic approach for ischemic stroke.
Collapse
Affiliation(s)
- Minjie Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xiaowei Wu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yingxiu Xu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Meijun He
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jiaying Yang
- College of Medicine, Soochow University, Suzhou, China
| | - Jie Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yuyao Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Guizhen Ao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jian Cheng
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.
| | - Jia Jia
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017; 163-164:144-171. [PMID: 28987927 DOI: 10.1016/j.pneurobio.2017.10.001] [Citation(s) in RCA: 639] [Impact Index Per Article: 79.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/30/2017] [Accepted: 10/02/2017] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in regulating the trafficking of fluid, solutes and cells at the blood-brain interface and maintaining the homeostatic microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the BBB can be disrupted, followed by the extravasation of blood components into the brain and compromise of normal neuronal function. This article reviews recent advances in our knowledge of the mechanisms underlying BBB dysfunction and recovery after ischemic stroke. CNS cells in the neurovascular unit, as well as blood-borne peripheral cells constantly modulate the BBB and influence its breakdown and repair after ischemic stroke. The involvement of stroke risk factors and comorbid conditions further complicate the pathogenesis of neurovascular injury by predisposing the BBB to anatomical and functional changes that can exacerbate BBB dysfunction. Emphasis is also given to the process of long-term structural and functional restoration of the BBB after ischemic injury. With the development of novel research tools, future research on the BBB is likely to reveal promising potential therapeutic targets for protecting the BBB and improving patient outcome after ischemic stroke.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | | | - Ling Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael V L Bennett
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
29
|
Cai M, Zhang W, Weng Z, Stetler RA, Jiang X, Shi Y, Gao Y, Chen J. Promoting Neurovascular Recovery in Aged Mice after Ischemic Stroke - Prophylactic Effect of Omega-3 Polyunsaturated Fatty Acids. Aging Dis 2017; 8:531-545. [PMID: 28966799 PMCID: PMC5614319 DOI: 10.14336/ad.2017.0520] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/20/2017] [Indexed: 12/17/2022] Open
Abstract
The aged population is among the highest at risk for ischemic stroke, yet most stroke patients of advanced ages (>80 years) are excluded from access to thrombolytic treatment by tissue plasminogen activator, the only FDA approved pharmacological therapy for stroke victims. Omega-3 polyunsaturated fatty acids (n-3 PUFAs) robustly alleviate ischemic brain injury in young adult rodents, but have not yet been studied in aged animals. This study investigated whether chronic dietary supplementation of n-3 PUFAs protects aging brain against cerebral ischemia and improves long-term neurological outcomes. Aged (18-month-old) mice were administered n-3 PUFA-enriched fish oil in daily chow for 3 months before and up to 8 weeks after 45 minutes of transient middle cerebral artery occlusion (tMCAO). Sensorimotor outcomes were assessed by cylinder test and corner test up to 35 days and brain repair dynamics evaluated immunohistologically up to 56 days after tMCAO. Mice receiving dietary supplementation of n-3 PUFAs for 3 months showed significant increases in brain ratio of n-3/n-6 PUFA contents, and markedly reduced long-term sensorimotor deficits and chronic ischemic brain tissue loss after tMCAO. Mechanistically, n-3 PUFAs robustly promoted post-ischemic angiogenesis and neurogenesis, and enhanced white matter integrity after tMCAO. The Pearson linear regression analysis revealed that the enhancement of neurogenesis and white matter integrity both correlated positively with improved sensorimotor activities after tMCAO. This study demonstrates that prophylactic dietary supplementation of n-3 PUFAs effectively improves long-term stroke outcomes in aged mice, perhaps by promoting post-stroke brain repair processes such as angiogenesis, neurogenesis, and white matter restoration.
Collapse
Affiliation(s)
- Mengfei Cai
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Wenting Zhang
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Zhongfang Weng
- 2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - R Anne Stetler
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China.,2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,3Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Xiaoyan Jiang
- 2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yejie Shi
- 2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,3Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Yanqin Gao
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China.,2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jun Chen
- 1State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, and Collaborative Innovation Center, Fudan University, Shanghai 200032, China.,2Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,3Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| |
Collapse
|
30
|
Three-Dimensional Organoid System Transplantation Technologies in Future Treatment of Central Nervous System Diseases. Stem Cells Int 2017; 2017:5682354. [PMID: 28904534 PMCID: PMC5585580 DOI: 10.1155/2017/5682354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/24/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
In recent years, scientists have made great achievements in understanding the development of human brain and elucidating critical elements of stepwise spatiotemporal control strategies in neural stem cell specification lineage, which facilitates successful induction of neural organoid in vitro including the cerebral cortex, cerebellar, neural tube, hippocampus cortex, pituitary, and optic cup. Besides, emerging researches on neural organogenesis promote the application of 3D organoid system transplantation in treating central nervous system (CNS) diseases. Present review will categorize current researches on organogenesis into three approaches: (a) stepwise, direct organization of region-specific or population-enriched neural organoid; (b) assemble and direct distinct organ-specific progenitor cells or stem cells to form specific morphogenesis organoid; and (c) assemble embryoid bodies for induction of multilayer organoid. However, the majority of these researches focus on elucidating cellular and molecular mechanisms involving in brain organogenesis or disease development and only a few of them conducted for treating diseases. In this work, we will compare three approaches and also analyze their possible indications for diseases in future treatment on the basis of their distinct characteristics.
Collapse
|
31
|
McBride DW, Zhang JH. Precision Stroke Animal Models: the Permanent MCAO Model Should Be the Primary Model, Not Transient MCAO. Transl Stroke Res 2017; 8:10.1007/s12975-017-0554-2. [PMID: 28718030 PMCID: PMC5772000 DOI: 10.1007/s12975-017-0554-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 06/28/2017] [Accepted: 07/05/2017] [Indexed: 12/16/2022]
Abstract
An argument for preclinical stroke research to make more use of the permanent middle cerebral artery occlusion (MCAO) model, rather than transient MCAO, is presented. Despite STAIR recommending permanent MCAO as the primary model, preclinical stroke research has not been listened. In 2012, Hossmann reported that 64% of the treatment studies for MCAO used prompt transient MCAO models and only 36% of the studies used permanent MCAO or gradual transient MCAO (i.e., embolic stroke model). Then, in 2014 and 2015, 88% of published basic science studies on large vessel occlusion used the transient MCAO model. However, this model only represents 2.5-11.3% of large vessel stroke patients. Therefore, the transient MCAO model, which mimics stroke with reperfusion, does not accurately reflect the majority of clinical stroke cases. Thus, once again, the argument for studying permanent MCAO as a primary model is made and supported.
Collapse
Affiliation(s)
- Devin W McBride
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
32
|
DRα1-MOG-35-55 Reduces Permanent Ischemic Brain Injury. Transl Stroke Res 2016; 8:284-293. [PMID: 27988839 DOI: 10.1007/s12975-016-0514-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/21/2016] [Accepted: 12/06/2016] [Indexed: 01/07/2023]
Abstract
Stroke induces a catastrophic immune response that involves the global activation of peripheral leukocytes, especially T cells. The human leukocyte antigen-DRα1 domain linked to MOG-35-55 peptide (DRα1-MOG-35-55) is a partial major histocompatibility complex (MHC) class II construct which can inhibit neuroantigen-specific T cells and block binding of the cytokine/chemokine macrophage migration inhibitory factor (MIF) to its CD74 receptor on monocytes and macrophages. Here, we evaluated the therapeutic effect of DRα1-MOG-35-55 in a mouse model of permanent distal middle cerebral artery occlusion (dMCAO). DRα1-MOG-35-55 was administered to WT C57BL/6 mice by subcutaneous injection starting 4 h after the onset of ischemia followed by three daily injections. We demonstrated that DRα1-MOG-35-55 post treatment significantly reduced brain infarct volume, improved functional outcomes, and inhibited the accumulation of CD4+ and CD8+ T cells and expression of pro-inflammatory cytokines in the ischemic brain 96 h after dMCAO. In addition, DRα1-MOG-35-55 treatment shifted microglia/macrophages in the ischemic brain to a beneficial M2 phenotype without changing their total numbers in the brain or blood. This study demonstrates for the first time the therapeutic efficacy of the DRα1-MOG-35-55 construct in dMCAO across MHC class II barriers in C57BL/6 mice. This MHC-independent effect obviates the need for tissue typing and will thus greatly expedite treatment with DRα1-MOG-35-55 in human stroke subjects. Taken together, our findings suggest that DRα1-MOG-35-55 treatment may reduce ischemic brain injury by regulating post-stroke immune responses in the brain and the periphery.
Collapse
|
33
|
Pu H, Jiang X, Wei Z, Hong D, Hassan S, Zhang W, Liu J, Meng H, Shi Y, Chen L, Chen J. Repetitive and Prolonged Omega-3 Fatty Acid Treatment After Traumatic Brain Injury Enhances Long-Term Tissue Restoration and Cognitive Recovery. Cell Transplant 2016; 26:555-569. [PMID: 27938482 DOI: 10.3727/096368916x693842] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most disabling clinical conditions that could lead to neurocognitive disorders in survivors. Our group and others previously reported that prophylactic enrichment of dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) markedly ameliorate cognitive deficits after TBI. However, it remains unclear whether a clinically relevant therapeutic regimen with n-3 PUFAs administered after TBI would still offer significant improvement of long-term cognitive recovery. In the present study, we employed the decline of spatial cognitive function as a main outcome after TBI to investigate the therapeutic efficacy of post-TBI n-3 PUFA treatment and the underlying mechanisms. Mice were subjected to sham operation or controlled cortical impact, followed by random assignment to receive the following four treatments: (1) vehicle control; (2) daily intraperitoneal injections of n-3 PUFAs for 2 weeks, beginning 2 h after TBI; (3) fish oil dietary supplementation throughout the study, beginning 1 day after TBI; or (4) combination of treatments (2) and (3). Spatial cognitive deficits and chronic brain tissue loss, as well as endogenous brain repair processes such as neurogenesis, angiogenesis, and oligodendrogenesis, were evaluated up to 35 days after TBI. The results revealed prominent spatial cognitive deficits and massive tissue loss caused by TBI. Among all mice receiving post-TBI n-3 PUFA treatments, the combined treatment of fish oil dietary supplement and n-3 PUFA injections demonstrated a reproducible beneficial effect in attenuating cognitive deficits although without reducing gross tissue loss. Mechanistically, the combined treatment promoted post-TBI restorative processes in the brain, including generation of immature neurons, microvessels, and oligodendrocytes, each of which was significantly correlated with the improved cognitive recovery. These results indicated that repetitive and prolonged n-3 PUFA treatments after TBI are capable of enhancing brain remodeling and could be developed as a potential therapy to treat TBI victims in the clinic.
Collapse
|
34
|
Jiang X, Pu H, Hu X, Wei Z, Hong D, Zhang W, Gao Y, Chen J, Shi Y. A Post-stroke Therapeutic Regimen with Omega-3 Polyunsaturated Fatty Acids that Promotes White Matter Integrity and Beneficial Microglial Responses after Cerebral Ischemia. Transl Stroke Res 2016; 7:548-561. [PMID: 27714669 DOI: 10.1007/s12975-016-0502-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/24/2016] [Accepted: 09/27/2016] [Indexed: 02/03/2023]
Abstract
White matter injury induced by ischemic stroke elicits sensorimotor impairments, which can be further deteriorated by persistent proinflammatory responses. We previously reported that delayed and repeated treatments with omega-3 polyunsaturated fatty acids (n-3 PUFAs) improve spatial cognitive functions and hippocampal integrity after ischemic stroke. In the present study, we report a post-stroke n-3 PUFA therapeutic regimen that not only confers protection against neuronal loss in the gray matter but also promotes white matter integrity. Beginning 2 h after 60 min of middle cerebral artery occlusion (MCAO), mice were randomly assigned to receive intraperitoneal docosahexaenoic acid (DHA) injections (10 mg/kg, daily for 14 days), alone or in combination with dietary fish oil (FO) supplements starting 5 days after MCAO. Sensorimotor functions, gray and white matter injury, and microglial responses were examined up to 28 days after MCAO. Our results showed that DHA and FO combined treatment-facilitated long-term sensorimotor recovery and demonstrated greater beneficial effect than DHA injections alone. Mechanistically, n-3 PUFAs not only offered direct protection on white matter components, such as oligodendrocytes, but also potentiated microglial M2 polarization, which may be important for white matter repair. Notably, the improved white matter integrity and increased M2 microglia were strongly linked to the mitigation of sensorimotor deficits after stroke upon n-3 PUFA treatments. Together, our results suggest that post-stroke DHA injections in combination with FO dietary supplement benefit white matter restoration and microglial responses, thereby dictating long-term functional improvements.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hongjian Pu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiaoming Hu
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Zhishuo Wei
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dandan Hong
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Wenting Zhang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| |
Collapse
|