151
|
Zhang H, Gao L, Zhang W, Li K. Differentiation of rat bone marrow mesenchymal stem cells into neurons induced by bone morphogenetic protein 7 in vitro. Neurol Res 2022; 45:440-448. [PMID: 36542543 DOI: 10.1080/01616412.2022.2154487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Spinal cord injury (SCI) is caused by external direct or indirect factors with high disability rate, which may even endanger the life of patients. To explore the role of bone morphogenetic protein 7 (BMP-7) in the differentiation of rat bone marrow mesenchymal stem cells (BMSCs) into neurons in vitro. METHODS BMSCs were isolated and cultured by whole bone marrow adherence method. Adipogenic induction and osteogenic differentiation were used to test the multi⁃directional differentiation ability of BMSCs. RESULTS After 28 days of adipogenic induction, BMSCs showed lipid droplets in the cytoplasm. After osteogenic induction, there were opaque lumps of mineral nodules in BMSCs. There were also orange-red or red mineral nodules in the extracellular matrix. The BMSCs in the 75 ng/ml BMP-7 group were morphologically similar to the neurons. After induction with BMP-7 for 2 h, the NF200 mRNA expression was higher, mRNA expression levels of SYN1, MAP2 and GFAP were higher. Positive rate of immunofluorescence staining in the BMP-7 group was notably increased. The positive rate of NSE immunofluorescence staining in the BMP-7 group was higher. CONCLUSION BMP-7 can induce rat BMSCs to differentiate into neurons in vitro.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Orthopaedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Lei Gao
- Department of Orthopaedics, the Second Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Wen Zhang
- Department of Orthopaedics, the Second Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| | - Kuanxin Li
- Department of Orthopaedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Department of Orthopaedics, the Second Affiliated Hospital of Medical College, Shihezi University, Shihezi 832000, China
| |
Collapse
|
152
|
Li X, Zhu H, Wen J, Huang J, Chen Y, Tian M, Ren J, Zhou L, Yang Q. Inhibition of BRD4 decreases fibrous scarring after ischemic stroke in rats by inhibiting the phosphorylation of Smad2/3. Brain Res 2022; 1797:148126. [PMID: 36244457 DOI: 10.1016/j.brainres.2022.148126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/17/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022]
Abstract
AIMS Fibrous scarring may play a much more important role in preventing secondary expansion of tissue damage and hindering repair and regeneration than glial scarring after central nervous system (CNS) injury. However, relatively little is known about how fibrous scars form and how fibrous scar formation is regulated after CNS injury. Bromodomain-containing protein 4 (BRD4) is involved in fibrosis in many tissues, and transforming growth factor-β1 (TGF-β1)/Smad2/3 signaling is one of the critical pathways of fibrosis. However, it is unclear whether and how BRD4 affects fibrous scar formation after ischemicbraininjury. In the present study, whether BRD4 can regulate the formation of fibrous scars after ischemic stroke via TGF-β1/Smad2/3 signaling was assessed. MATERIALS AND METHODS Primary meningeal fibroblasts isolated from neonatal SD rats were treated with TGF-β1, SB431542 (a TGF-β1 receptor inhibitor) and JQ1 (a small-molecule BET inhibitor that can also inhibit BRD4). BRD4 was knocked down in adult Sprague-Dawley (SD) rats by using adenovirus before middle cerebral artery occlusion/reperfusion (MCAO/R) injury. The proliferation and migration of meningeal fibroblasts in vitro were evaluated with the Cell Counting Kit-8 (CCK-8) assay and scratch test, respectively. Neurological function was assessed with Longa scores, modified Bederson Scores and modified neurological severity scores (mNSSs). The infarct volume was assessed with TTC staining. The protein expression of synaptophysin (SY), BRD4, Smad2/3, p-Smad2/3, α-smooth muscle actin (α-SMA), collagen-1 (COL1) and fibronectin (FN) in vivo and in vitro was examined with immunocytochemistry, immunofluorescence, and Western blotting. KEY FINDINGS BRD4 expression was upregulated in a TGF-β1-induced meningeal fibroblast fibrosis model and was downregulated by the TGF-β1 receptor inhibitor SB431542 in vitro. JQ1, a small-molecule BET inhibitor, inhibited BRD4 and decreased TGF-β1-induced meningeal fibroblast proliferation, migration and activation. Furthermore, MCAO/R injury induced fibrosis and upregulated BRD4 expression in the cerebral infarct center. BRD4 knockdown by adenovirus inhibited fibrous scarring, promoted synaptic survival, decreased the infarct volume, and improved neurological function after MCAO/R injury. Moreover, inhibition of BRD4, either by JQ1 in vitro or adenovirus in vivo, decreased the phosphorylation of Smad2/3. CONCLUSIONS This study is the first to indicate that inhibition of BRD4 delays fibrous scarring after ischemic stroke through mechanisms involving the phosphorylation of Smad2/3.
Collapse
Affiliation(s)
- Xuemei Li
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huimin Zhu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Wen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiagui Huang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingfen Tian
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiangxia Ren
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhou
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Yang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
153
|
Hong X, Jian Y, Ding S, Zhou J, Zheng X, Zhang H, Zhou B, Zhuang C, Wan J, Tong X. Kir4.1 channel activation in NG2 glia contributes to remyelination in ischemic stroke. EBioMedicine 2022; 87:104406. [PMID: 36527899 PMCID: PMC9791134 DOI: 10.1016/j.ebiom.2022.104406] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Stroke is one of the most common neurological diseases in the world and is clinically manifested by transient or permanent brain dysfunction. It has a high mortality and disability rate, which severely affects people's health and diminishes the quality of life. However, there is no efficient treatment that can be considered curative and there are other less well-known theories of pathogenesis. Therefore, it is imperative to gain a full understanding of the pathophysiology of ischemia and to seek new therapeutic strategies. METHODS We first examined Kir4.1 channel and myelin based protein (MBP) expression in brain tissues from acute ischemic patients by Western blotting. We then established a transient ischemic mouse model (tMCAO) to conduct molecular, cell biological, transmission electron microscopy and pharmacokinetic studies, as well as in Kir4.1 cKO mice. Finally, neuroimaging and behavioral analyses were used to examine whether activation of Kir4.1 channel by luteolin could contribute to neuronal functional recovery in ischemic stroke. FINDINGS In acute ischemic stroke patients, we first demonstrated that Kir4.1 ion channels were greatly impaired and a severe demyelination of axons occurred in ischemic infarction area of cerebral cortex in these patients. Further evidence showed that the deficits of Kir4.1 channels in NG2 glia led to the myelin loss of axons in a transient ischemic mouse model (tMCAO). Treating ischemic mice with a natural botanical extract, luteolin augmented Kir4.1 channel currents in NG2 glia and consequently promoted remyelination of axons, alleviated the infarction area and ultimately improved motor function in a series of behavioral tests. INTERPRETATION Targeting Kir4.1 ion channels expressed in NG2 glial cells by luteolin treatment highlights an effective therapeutic strategy for a prompt brain functional recovery in ischemic stroke. FUNDING This work was supported by grants from the Ministry of Science and Technology China Brain Initiative (2022ZD0204702, to X.T.), the National Natural Science Foundation of China (82271466, 82171279, 31970904 and 31571063), the Program for Professor of Special Appointment (Eastern Scholar for Dr. X.T.) at Shanghai Institutions for Higher Learning (1510000084), Shanghai Pujiang Talent Award (15PJ1404600), Shanghai Municipal Science and Technology Major Project (2018SHZDZX05) and Shanghai Science and Technology Project (17411954000).
Collapse
Affiliation(s)
- Xiaoqi Hong
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujin Jian
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenghao Ding
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianpo Zhou
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoli Zheng
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huimin Zhang
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Butian Zhou
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Canbin Zhuang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqing Wan
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Corresponding author.
| | - Xiaoping Tong
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China,Corresponding author.
| |
Collapse
|
154
|
Application of Robotic Recovery Techniques to Stroke Survivors-Bibliometric Analysis. J Pers Med 2022; 12:jpm12122066. [PMID: 36556286 PMCID: PMC9788322 DOI: 10.3390/jpm12122066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Stroke is a significant disability and death cause worldwide and is conventionally defined as a neurological impairment relating to the intense focal harm of the central nervous system (CNS) by vascular causative components. Although the applicability of robotic rehabilitation is a topic with considerable practical significance because it has produced noticeably higher improvements in motor function than regular (physical and occupational) therapy and exempted the therapists, most of the existing bibliometric papers were not focused on stroke survivors. Additionally, a modular system is designed by joining several medical end-effector devices to a single limb segment, which addresses the issue of potentially dangerous pathological compensatory motions. Searching the Web of Science database, 31,930 papers were identified, and using the VOSviewer software and science mapping technology, data were extracted on the most prolific countries, the connections between them, the most valuable journals according to certain factors, their average year of publication, the most influential papers, and the most relevant topical issues (bubble map of term occurrence). The most prolific country in the analyzed field and over the entire period evaluated (1975-2022) is the United States, and the most prolific journal is Neurorehabilitation and Neural Repair, observing a marked increase in the three periods of scientific interest for this field. The present paper assesses numerous scientific publications to provide, through statistical interpretation of the data, a detailed description of the use of robotic rehabilitation in stroke survivors. The findings may aid scientists, academics, and clinicians in establishing precise goals in the optimization of the management of stroke survivors via robotic rehabilitation, but also through easier access to scientifically validated literature.
Collapse
|
155
|
Li M, Chen H, Zhu M. Mesenchymal stem cells for regenerative medicine in central nervous system. Front Neurosci 2022; 16:1068114. [PMID: 36583105 PMCID: PMC9793714 DOI: 10.3389/fnins.2022.1068114] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells, whose paracrine and immunomodulatory potential has made them a promising candidate for central nervous system (CNS) regeneration. Numerous studies have demonstrated that MSCs can promote immunomodulation, anti-apoptosis, and axon re-extension, which restore functional neural circuits. The therapeutic effects of MSCs have consequently been evaluated for application in various CNS diseases including spinal cord injury, cerebral ischemia, and neurodegenerative disease. In this review, we will focus on the research works published in the field of mechanisms and therapeutic effects of MSCs in CNS regeneration.
Collapse
Affiliation(s)
- Man Li
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxin Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Mingxin Zhu,
| |
Collapse
|
156
|
Preferred Migration of Mitochondria toward Cells and Tissues with Mitochondrial Damage. Int J Mol Sci 2022; 23:ijms232415734. [PMID: 36555376 PMCID: PMC9779580 DOI: 10.3390/ijms232415734] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are organelles that play a vital role in cellular survival by supplying ATP and metabolic substrates via oxidative phosphorylation and the Krebs cycle. Hence, mitochondrial dysfunction contributes to many human diseases, including metabolic syndromes, neurodegenerative diseases, cancer, and aging. Mitochondrial transfer between cells has been shown to occur naturally, and mitochondrial transplantation is beneficial for treating mitochondrial dysfunction. In this study, the migration of mitochondria was tracked in vitro and in vivo using mitochondria conjugated with green fluorescent protein (MTGFP). When MTGFP were used in a coculture model, they were selectively internalized into lung fibroblasts, and this selectivity depended on the mitochondrial functional states of the receiving fibroblasts. Compared with MTGFP injected intravenously into normal mice, MTGFP injected into bleomycin-induced idiopathic pulmonary fibrosis model mice localized more abundantly in the lung tissue, indicating that mitochondrial homing to injured tissue occurred. This study shows for the first time that exogenous mitochondria are preferentially trafficked to cells and tissues in which mitochondria are damaged, which has implications for the delivery of therapeutic agents to injured or diseased sites.
Collapse
|
157
|
Qin C, Wen M. miR-145 from Bone Marrow Mesenchymal Stem Cells (BMSC) Improves Cardiac Function After Myocardial Infarction in Rat with Diabetes. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study assesses the role of miR-145 from BMSC in the cardiac function after MI in rat with diabetes. Rat with T1DM model was established and then were treated with PBS, DM-BMSC with overexpression of miR-145, BMSC with the knockdown of miR-145 respectively after twenty-four hours
followed by analysis of the remodeling of vessels and protein, mRNA expressions. miR-145 in DM-BMSC was significantly reduced compared with control group and DM-BMSC prolonged the survival rate of rats. The formation of blood capillary and axon growth in DM-BMSC was increased and decreased
in BMSC with knockdown of miR-145. The therapeutic action of DM-BMSC could be improved notably and remodeling of vessels and protein was increased. Smad1 was a target gene of miR-145. In conclusion, cardiac function and neurological recovery in MI is improved by miR-145 through targeting Smad1
expression, indicating that miR-145 might be a novel target for the treatment of MI.
Collapse
Affiliation(s)
- Chuanyu Qin
- Department of Cardiology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161099, Heilongjiang Province, China
| | - Mingli Wen
- Department of Respiratory Medicine, The First Affiliated Hospital of Qiqihar Medical College, Qiqihar City, 161041, Heilongjiang Province, China
| |
Collapse
|
158
|
Yuan Y, Sun J, You T, Shen W, Xu W, Dong Q, Cui M. Extracellular Vesicle-Based Therapeutics in Neurological Disorders. Pharmaceutics 2022; 14:pharmaceutics14122652. [PMID: 36559145 PMCID: PMC9783774 DOI: 10.3390/pharmaceutics14122652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/26/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Neurological diseases remain some of the major causes of death and disability in the world. Few types of drugs and insufficient delivery across the blood-brain barrier limit the treatment of neurological disorders. The past two decades have seen the rapid development of extracellular vesicle-based therapeutics in many fields. As the physiological and pathophysiological roles of extracellular vesicles are recognized in neurological diseases, they have become promising therapeutics and targets for therapeutic interventions. Moreover, advanced nanomedicine technologies have explored the potential of extracellular vesicles as drug delivery systems in neurological diseases. In this review, we discussed the preclinical strategies for extracellular vesicle-based therapeutics in neurological disorders and the struggles involved in their clinical application.
Collapse
Affiliation(s)
- Yiwen Yuan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Jian Sun
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Tongyao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Weiwei Shen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Wenqing Xu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (Q.D.); (M.C.)
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200031, China
- Correspondence: (Q.D.); (M.C.)
| |
Collapse
|
159
|
Liu C, Yang ZX, Zhou SQ, Ding D, Hu YT, Yang HN, Han D, Hu SQ, Zong XM. Overexpression of vascular endothelial growth factor enhances the neuroprotective effects of bone marrow mesenchymal stem cell transplantation in ischemic stroke. Neural Regen Res 2022; 18:1286-1292. [PMID: 36453413 PMCID: PMC9838145 DOI: 10.4103/1673-5374.358609] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although bone marrow mesenchymal stem cells (BMSCs) might have therapeutic potency in ischemic stroke, the benefits are limited. The current study investigated the effects of BMSCs engineered to overexpress vascular endothelial growth factor (VEGF) on behavioral defects in a rat model of transient cerebral ischemia, which was induced by middle cerebral artery occlusion. VEGF-BMSCs or control grafts were injected into the left striatum of the infarcted hemisphere 24 hours after stroke. We found that compared with the stroke-only group and the vehicle- and BMSCs-control groups, the VEGF-BMSCs treated animals displayed the largest benefits, as evidenced by attenuated behavioral defects and smaller infarct volume 7 days after stroke. Additionally, VEGF-BMSCs greatly inhibited destruction of the blood-brain barrier, increased the regeneration of blood vessels in the region of ischemic penumbra, and reducedneuronal degeneration surrounding the infarct core. Further mechanistic studies showed that among all transplant groups, VEGF-BMSCs transplantation induced the highest level of brain-derived neurotrophic factor. These results suggest that BMSCs transplantation with vascular endothelial growth factor has the potential to treat ischemic stroke with better results than are currently available.
Collapse
Affiliation(s)
- Cui Liu
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Emergency Department of Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People’s Hospital of Lianyungang, Lianyungang, Lianyungang, Jiangsu Province, China
| | - Zhi-Xiang Yang
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Si-Qi Zhou
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ding Ding
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yu-Ting Hu
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hong-Ning Yang
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Dong Han
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Shu-Qun Hu
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Xue-Mei Zong, ; Shu-Qun Hu, .
| | - Xue-Mei Zong
- Institute of Emergency Rescue Medicine, Emergency Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Key Laboratory of Brain Diseases Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu Province, China,Correspondence to: Xue-Mei Zong, ; Shu-Qun Hu, .
| |
Collapse
|
160
|
Gualou Guizhi Decoction Improves Glucose Metabolism and Alleviates Microglia-Associated Inflammation after Cerebral Ischemia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9438250. [PMID: 36317102 PMCID: PMC9617704 DOI: 10.1155/2022/9438250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/24/2022] [Indexed: 11/06/2022]
Abstract
Background The classical prescription Gualou Guizhi decoction (GL), a mixture of Radix Trichosanthis, Ramulus Cinnamomi, Radix Paeoniae Alba, Radix Glycyrrhizae, Zingiberis Rhizoma Recens, and Fructus Ziziphus Jujuba, was clinically used in the treatment of limb spasms after stroke and has achieved remarkable therapeutic effects. However, the underlying mechanism still needs to be further explored. Methods Cerebral ischemia/reperfusion (CI/R) in Sprague-Dawley rats was induced by middle cerebral artery occlusion followed by filament removal. GL was intragastrically administered once daily for 7 or 14 consecutive days. The effect of GL on neurobehavioral impairment was evaluated. 18F-FDG micro-PET imaging was used to detect the effects of GL on glucose utilization in neural cells after CI/R. Immunohistochemical staining of glucose transporter 1 (Glut-1), glial fibrillary acidic protein (GFAP), and ionized calcium-binding adaptor molecule-1 (Iba-1) was further performed to show the effects of GL on cerebral glucose transport and the activation of inflammatory-related glial cells. Markers related to the microglial subtype were also assessed to investigate the effects of GL on microglia polarization. Results Neurological deficits induced by CI/R were significantly improved by GL administration. GL restored the glucose uptake in the ischemic hemisphere. Glut-1, the major glucose transporter in the brain, was significantly increased after GL treatment. Moreover, GL mitigated the activation of astrocytes and microglia after CI/R. Furthermore, GL significantly decreased proinflammatory M1-type microglial markers TNF-α and iNOS, while increasing anti-inflammatory M2 microglial markers CD206 and Arg-1. Conclusion GL enhanced the uptake and utilization of glucose in neural cells after CI/R. It exerted significant anti-inflammatory effects by regulating the polarization of microglia. These results provided further evidence supporting the clinical application of GL in the treatment of cerebral ischemic stroke.
Collapse
|
161
|
Treatment of rat brain ischemia model by NSCs-polymer scaffold transplantation. OPEN CHEM 2022. [DOI: 10.1515/chem-2022-0213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Abstract
Neural stem cells (NSCs) transplantation is a promising therapeutic strategy for ischemic stroke. However, significant cell death after transplantation greatly limits its effectiveness. Poly (trimethylene carbonate)15-F127-poly (trimethylene carbonate)15 (PTMC15-F127-PTMC15, PFP) is a biodegradable thermo-sensitive hydrogel biomaterial, which can control drug release and provide permissive substrates for donor NSCs. In our study, we seeded NSCs into PFP polymer scaffold loaded with three neurotrophic factors, including brain-derived neurotrophic factor, nerve growth factor, and Neurotrophin-3. And then we transplanted this NSCs-polymer scaffold in rat brains 14 days after middle cerebral artery occlusion. ELISA assay showed that PFP polymer scaffold sustained releasing three neurotrophic factors for at least 14 days. Western Blot and fluorescence immunostaining revealed that NSCs-polymer scaffold transplantation significantly reduced apoptosis of ischemic penumbra and promoted differentiation of the transplanted NSCs into mature neurons. Furthermore, infarct size was reduced, and neurological performance of the animals were improved by the transplanted NSCs-polymer scaffold. These results demonstrate that PFP polymer scaffold loaded with neurotrophic factors can enhance the effectiveness of stem cell transplantation therapy, which provides a new way for cell transplantation therapy in ischemic stroke.
Collapse
|
162
|
Lu H, Ashiqueali R, Lin CI, Walchale A, Clendaniel V, Matheson R, Fisher M, Lo EH, Selim M, Shehadah A. Histone Deacetylase 3 Inhibition Decreases Cerebral Edema and Protects the Blood–Brain Barrier After Stroke. Mol Neurobiol 2022; 60:235-246. [DOI: 10.1007/s12035-022-03083-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022]
Abstract
AbstractWe have previously shown that selective inhibition of histone deacetylase 3 (HDAC3) decreases infarct volume and improves long-term functional outcomes after stroke. In this study, we examined the effects of HDAC3 inhibition on cerebral edema and blood–brain barrier (BBB) leakage and explored its underlying mechanisms. Adult male Wistar rats were subjected to 2-h middle cerebral artery occlusion (MCAO) and randomly treated i.p. with either vehicle or a selective HDAC3 inhibitor (RGFP966) at 2 and 24 h after stroke. Modified neurological severity scores (mNSS) were calculated at 2 h, 1 day, and 3 days. H&E, Evans blue dye (EBD) assay, and fluorescein isothiocyanate (FITC)-dextran were employed to assess cerebral edema and BBB leakage. Western blot for matrix metalloproteinase-9 (MMP9), MMP-9 zymography, and immunostaining for HDAC3, GFAP, Iba-1, albumin, aquaporin-4, claudin-5, ZO-1, and NF-kB were performed. Early RGFP966 administration decreased cerebral edema (p = 0.002) and BBB leakage, as measured by EBD assay, FITC-dextran, and albumin extravasation (p < 0.01). RGFP966 significantly increased tight junction proteins (claudin-5 and ZO-1) in the peri-infarct area. RGFP966 also significantly decreased HDAC3 in GFAP + astrocytes, which correlated with better mNSS (r = 0.67, p = 0.03) and decreased cerebral edema (r = 0.64, p = 0.04). RGFP966 decreased aquaporin-4 in GFAP + astrocytes (p = 0.002), as well as, the inflammatory markers Iba-1, NF-kB, and MMP9 in the ischemic brain (p < 0.05). Early HDAC3 inhibition decreases cerebral edema and BBB leakage. BBB protection by RGFP966 is mediated in part by the upregulation of tight junction proteins, downregulation of aquaporin-4 and HDAC3 in astrocytes, and decreased neuroinflammation.
Collapse
|
163
|
De Becker A, Heestermans R, De Brouwer W, Bockstaele K, Maes K, Van Riet I. Genetic profiling of human bone marrow mesenchymal stromal cells after in vitro expansion in clinical grade human platelet lysate. Front Bioeng Biotechnol 2022; 10:1008271. [DOI: 10.3389/fbioe.2022.1008271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are non-hematopoietic cells that have a broad therapeutic potential. To obtain sufficient cells for clinical application, they must be expanded ex vivo. In the initial expansion protocols described, fetal calf serum (FCS) was used as the reference growth supplement, but more recently different groups started to replace FCS with platelet lysate (PL). We investigated in this study the impact of the culture supplement on gene expression of MSCs. Human bone marrow derived MSCs were expanded in vitro in FCS and PL supplemented medium. We found that MSCs expanded in PL-containing medium (PL-MSCs) express typical MSC immunomorphological features and can migrate, as their counterparts expanded in FCS-containing medium, through a layer of endothelial cells in vitro. Additionally, they show an increased proliferation rate compared to MSCs expanded in FCS medium (FCS-MSCs). RNA sequencing performed for MSCs cultured in both types of expansion medium revealed a large impact of the choice of growth supplement on gene expression: 1974 genes were at least twofold up- or downregulated. We focused on impact of genes involved in apoptosis and senescence. Our data showed that PL-MSCs express more anti-apoptotic genes and FCS-MSCs more pro-apoptotic genes. FCS-MSCs showed upregulation of senescence-related genes after four passages whereas this was rarer in PL-MSCs at the same timepoint. Since PL-MSCs show higher proliferation rates and anti-apoptotic gene expression, they might acquire features that predispose them to malignant transformation. We screened 10 MSC samples expanded in PL-based medium for the presence of tumor-associated genetic variants using a 165 gene panel and detected only 21 different genetic variants. According to our analysis, none of these were established pathogenic mutations. Our data show that differences in culture conditions such as growth supplement have a significant impact on the gene expression profile of MSCs and favor the use of PL over FCS for expansion of MSCs.
Collapse
|
164
|
Jiang XH, Li HF, Chen ML, Zhang YX, Chen HB, Chen RH, Xiao YC, Liu N. Treadmill exercise exerts a synergistic effect with bone marrow mesenchymal stem cell-derived exosomes on neuronal apoptosis and synaptic-axonal remodeling. Neural Regen Res 2022; 18:1293-1299. [PMID: 36453414 PMCID: PMC9838147 DOI: 10.4103/1673-5374.357900] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Treadmill exercise and mesenchymal stem cell transplantation are both practical and effective methods for the treatment of cerebral ischemia. However, whether there is a synergistic effect between the two remains unclear. In this study, we established rat models of ischemia/reperfusion injury by occlusion of the middle cerebral artery for 2 hours and reperfusion for 24 hours. Rat models were perfused with bone marrow mesenchymal stem cell-derived exosomes (MSC-exos) via the tail vein and underwent 14 successive days of treadmill exercise. Neurological assessment, histopathology, and immunohistochemistry results revealed decreased neuronal apoptosis and cerebral infarct volume, evident synaptic formation and axonal regeneration, and remarkably recovered neurological function in rats subjected to treadmill exercise and MSC-exos treatment. These effects were superior to those in rats subjected to treadmill exercise or MSC-exos treatment alone. Mechanistically, further investigation revealed that the activation of JNK1/c-Jun signaling pathways regulated neuronal apoptosis and synaptic-axonal remodeling. These findings suggest that treadmill exercise may exhibit a synergistic effect with MSC-exos treatment, which may be related to activation of the JNK1/c-Jun signaling pathway. This study provides novel theoretical evidence for the clinical application of treadmill exercise combined with MSC-exos treatment for ischemic cerebrovascular disease.
Collapse
Affiliation(s)
- Xin-Hong Jiang
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China,Fujian Institute of Cerebrovascular Disease, Fuzhou, Fujian Province, China,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Hang-Feng Li
- Department of Neurology, Longyan First Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Man-Li Chen
- Fujian Institute of Cerebrovascular Disease, Fuzhou, Fujian Province, China,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China,Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Yi-Xian Zhang
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China,Fujian Institute of Cerebrovascular Disease, Fuzhou, Fujian Province, China,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Hong-Bin Chen
- Fujian Institute of Cerebrovascular Disease, Fuzhou, Fujian Province, China,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China,Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Rong-Hua Chen
- Fujian Institute of Cerebrovascular Disease, Fuzhou, Fujian Province, China,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China,Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Ying-Chun Xiao
- Fujian Institute of Cerebrovascular Disease, Fuzhou, Fujian Province, China,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China,Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Nan Liu
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China,Fujian Institute of Cerebrovascular Disease, Fuzhou, Fujian Province, China,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China,Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China,Correspondence to: Nan Liu, .
| |
Collapse
|
165
|
Gao X, Li R, Luo L, Zhang D, Liu Q, Zhang J, Mao S. Alpha-asarone ameliorates neurological deterioration of intracerebral hemorrhagic rats by alleviating secondary brain injury via anti-excitotoxicity pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154363. [PMID: 35926378 DOI: 10.1016/j.phymed.2022.154363] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/14/2022] [Accepted: 07/26/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Secondary brain injury (SBI) has been confirmed as a leading cause for the poor prognosis of patients suffering from intracerebral hemorrhage (ICH). SBI co-exists in ischemia and hemorrhagic stroke. Neuro-excitotoxicity is considered the initiating factor of ICH-induced SBI. Our previous research has revealed alpha-asarone (ASA)'s efficacy against cerebral ischemia-reperfusion stroke by mitigating neuro-excitotoxicity. It is not yet known if ASA exhibit neuroprotection against ICH. PURPOSE This work aimed to investigate ASA's therapeutic effects and potential mechanisms of action against ICH in a classic rat model induced by collagenase Ⅶ injection. METHODS An in vivo ICH model of Sprague-Dawley rats was established by collagenase Ⅶ injection. We administrated different ASA doses (10, 20, or 40 mg/kg, i.p.) at 2 h post-ICH. Then, rats' short- and long-term neurobehavioral function, bodyweight change, and learning and memory ability were blindly evaluated. Histological, Nissl, and flow cytometry were applied to assess the neuronal damage post-ICH. The wet/dry method and Evans blue extravasation estimated brain edema and blood-brain barrier function. Pathway-related proteins were investigated by immunofluorescence staining, enzyme-linked immunosorbent assay, and Western-blot analysis. RESULTS The results demonstrated that ASA ameliorated neurological deterioration, bodyweight loss, and learning and memory ability of ICH rats. Histological, Nissl, and flow cytometry analyses showed that ASA reduced neuronal damage and apoptosis post-ICH. Besides, ASA probably mitigated brain edema and blood-brain barrier dysfunction via inhibiting astrocyte activation and consequent pro-inflammatory response. The mechanism investigation attributed ASA's efficacy to the following aspects: 1) promoting sodium ion excretion, thus blocking excitatory signal transduction along the axon; 2) preventing glutamate-involved pathways, i.e., decrease of N-methyl-d-aspartic acid receptor subunit 2B, increase of glutamate transporter-1, and alleviation of calcium-related cascades, mitochondrion-associated apoptosis, and neuronal autophagy; 3) enhancing the expression of GABAARs, thus abating neuronal excitotoxicity. CONCLUSION Our study first confirmed the effect of ASA on ameliorating the neurobehavioral deterioration of ICH rats, possibly via alleviation of glutamate-involved neuro-excitotoxicity, i.e., calcium cascades, mitochondrion-involved apoptosis, neuronal autophagy, and astrocyte-related inflammation. These findings not only provided a promising drug candidate for clinical treatment of ICH but also shed light on the future drug discovery against ICH.
Collapse
Affiliation(s)
- Xiaofeng Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rui Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Lijun Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Di Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jian Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Shengjun Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
166
|
Endothelial caveolin-1 regulates cerebral thrombo-inflammation in acute ischemia/reperfusion injury. EBioMedicine 2022; 84:104275. [PMID: 36152520 PMCID: PMC9508414 DOI: 10.1016/j.ebiom.2022.104275] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Thrombo-inflammation is an important checkpoint that orchestrates infarct development in ischemic stroke. However, the underlying mechanism remains largely unknown. Here, we explored the role of endothelial Caveolin-1 (Cav-1) in cerebral thrombo-inflammation. METHODS The correlation between serum Cav-1 level and clinical outcome was analyzed in acute ischemic stroke patients with successful recanalization. Genetic manipulations by endothelial-specific adeno-associated virus (AAV) and siRNA were applied to investigate the effects of Cav-1 in thrombo-inflammation in a transient middle cerebral artery occlusion (tMCAO) model. Thrombo-inflammation was analyzed by microthrombosis formation, myeloid cell infiltration, and endothelial expression of adhesion molecules as well as inflammatory factors. FINDINGS Reduced circulating Cav-1, with the potential to predict microembolic signals, was more frequently detected in recanalized stroke patients without early neurological improvement. At 24 h after tMCAO, serum Cav-1 was consistently reduced in mice. Endothelial Cav-1 was decreased in the peri-infarct region. Cav-1-/- endothelium, with prominent barrier disruption, displayed extensive microthrombosis, accompanied by increased myeloid cell inflammatory infiltration after tMCAO. Specific enhanced expression of endothelial Cav-1 by AAV-Tie1-Cav-1 remarkably reduced infarct volume, attenuated vascular hyper-permeability and alleviated thrombo-inflammation in both wild-type and Cav-1-/- tMCAO mice. Transcriptome analysis after tMCAO further designated Rxrg as the most significantly changed molecule resulting from the knockdown of Cav-1. Supplementation of RXR-γ siRNA reversed AAV-Tie1-Cav-1-induced amelioration of thrombo-inflammation without affecting endothelial tight junction. INTERPRETATION Endothelial Cav-1/RXR-γ may regulate infarct volume and neurological impairment, possibly through selectively controlling thrombo-inflammation coupling, in cerebral ischemia/reperfusion. FUNDING This work was supported by National Natural Science Foundation of China.
Collapse
|
167
|
Yao X, Wang W, Li Y, Cao Z, Wang Y, Yuan Y, Li X, Liang X, Yu Y, Liu L. Study of the mechanism by which MSCs combined with LITUS treatment improve cognitive dysfunction caused by traumatic brain injury. Neurosci Lett 2022; 787:136825. [PMID: 35933061 DOI: 10.1016/j.neulet.2022.136825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/28/2022] [Accepted: 07/31/2022] [Indexed: 11/26/2022]
Abstract
Traumatic brain injury (TBI) substantially affects the quality of life of patients, and an effective therapy is unavailable. Previous studies have shown that mesenchymal stem cells (MSCs) and low-intensity transcranial ultrasound (LITUS) are effective treatments for neurological damage, inflammation, edema and cognitive impairment caused by TBI. However, it is unclear whether the combination of the two treatments exerts an additive effect. In this study, a rat TBI model was established using the controlled cortical impact (CCI) method. Neurological function was assessed by determining the rat modified neurological score (mNSS), and cognitive function was assessed using the Y-maze. Pathological changes in the injured tissue were observed using hematoxylin-eosin (HE) staining and immunohistochemistry (IHC), and western blot was performed to detect the expression levels of Nestin, neuron-specific enolase (NSE), glial fibrillary acidic protein (GFAP), growth-associated protein-43 (GAP-43), postsynaptic density protein (PSD-95), brain-derived neurotrophic factor (BDNF), tumor necrosis factor-α (TNF-α), and aquaporin-4 (AQP-4). Real-time fluorescence quantitative polymerase chain reaction (RT-PCR) was performed to detect the expression levels of GAP-43, PSD-95, BDNF, TNF-α, and AQP-4 mRNA to investigate whether MSCs combined with LITUS exert an additive therapeutic effect of alleviating the cognitive dysfunction caused by TBI and the possible mechanisms involved. Rats exhibited cognitive dysfunction 28 days after TBI, and MSCs combined with LITUS treatment ameliorated the cognitive deficits caused by TBI via increasing Nestin, NSE, GAP-43, PSD-95, and BDNF expression and attenuating the inflammatory response and edema caused by TBI via reducing TNF-α and AQP-4 expression. According to these results, MSCs combined with LITUS is more effective than MSCs alone for the treatment of TBI, and the mechanism may be the promotion of neuronal proliferation and differentiation, and the attenuation of the inflammatory response and edema, which ameliorates the spatial learning memory impairment caused by TBI. MSCs combined with LITUS treatment represents a new approach for the clinical treatment of patients with TBI.
Collapse
Affiliation(s)
- Xinyu Yao
- Graduate School of Chengde Medical University, Shuangqiao District, Chengde, Hebei Province, China; First Hospital of Qinhuangdao, Culture Road, Seaport District, Qinhuangdao, Hebei Province, China.
| | - Wenzhu Wang
- China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, North Jiaomen Road, Fengtai District, Beijing, China.
| | - Yue Li
- First Hospital of Qinhuangdao, Culture Road, Seaport District, Qinhuangdao, Hebei Province, China.
| | - Zhendong Cao
- Graduate School of Chengde Medical University, Shuangqiao District, Chengde, Hebei Province, China
| | - Yongheng Wang
- Department of Neurosurgery, First Hospital of Qinhuangdao, Culture Road, Seaport District, Qinhuangdao, Hebei Province, China
| | - Yi Yuan
- School of Electrical Engineering, Yanshan University, Hebei Avenue, Seaport District, Qinhuangdao, Hebei Province, China.
| | - Xiaoling Li
- Applying Chemistry Key Lab, Yanshan University, Hebei Avenue, Seaport District, Qinhuangdao, Hebei Province, China
| | - Xin Liang
- Graduate School of Chengde Medical University, Shuangqiao District, Chengde, Hebei Province, China; First Hospital of Qinhuangdao, Culture Road, Seaport District, Qinhuangdao, Hebei Province, China.
| | - Yan Yu
- China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, North Jiaomen Road, Fengtai District, Beijing, China.
| | - Lanxiang Liu
- Graduate School of Chengde Medical University, Shuangqiao District, Chengde, Hebei Province, China; First Hospital of Qinhuangdao, Culture Road, Seaport District, Qinhuangdao, Hebei Province, China.
| |
Collapse
|
168
|
Edaravone Dexborneol Alleviates Cerebral Ischemic Injury via MKP-1-Mediated Inhibition of MAPKs and Activation of Nrf2. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4013707. [PMID: 36110124 PMCID: PMC9470337 DOI: 10.1155/2022/4013707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/27/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
The edaravone and dexborneol concentrated solution for injection (edaravone-dexborneol) is a medication used clinically to treat neurological impairment induced by ischemic stroke. This study was aimed at investigating the preventive effects and the underlying mechanisms of edaravone-dexborneol on cerebral ischemic injury. A rat four-vessel occlusion (4-VO) model was established, and the neuronal injury and consequent neurological impairment of rats was investigated. Brain tissue malondialdehyde (MDA), myeloperoxidase (MPO), and nitric oxide (NO) levels were determined. The levels of proteins in mitogen-activated protein kinases (MAPKs), nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor-κB (NF-κB) signaling pathways were determined by western immunoblotting. The function of mitogen-activated protein kinase phosphatase 1 (MKP-1) was investigated using both western blot and immunofluorescence methods, and the effect of the MKP-1 inhibitor, (2E)-2-benzylidene-3-(cyclohexylamino)-3H-inden-1-one (BCI), was investigated. The results indicated that edaravone-dexborneol alleviated neurological deficiency symptoms and decreased apoptosis and neuron damage in the hippocampal CA1 area of the ischemic rats. Edaravone-dexborneol increased the MKP-1 level; decreased the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK); inhibited NF-κB p65 activation; and boosted Nrf2 activation, all of which were partially reversed by the MKP-1 inhibitor, BCI. The above results indicated that the upregulation of MKP-1 contributed to the protective effects of edaravone-dexborneol against ischemic brain injury. Our findings support the hypothesis that edaravone-dexborneol can alleviate cerebral ischemic injury via the upregulation of MKP-1, which inhibits MAPKs and activates Nrf2.
Collapse
|
169
|
Effect of Electroacupuncture on Short-Chain Fatty Acids in Peripheral Blood after Middle Cerebral Artery Occlusion/Reperfusion in Rats Based on Gas Chromatography–Mass Spectrometry. Mediators Inflamm 2022; 2022:3997947. [PMID: 36052308 PMCID: PMC9427317 DOI: 10.1155/2022/3997947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022] Open
Abstract
Previous fundamental and clinical research has shown that electroacupuncture (EA) at the acupoints of Quchi (LI11) and Zusanli (ST36) can successfully alleviate motor dysfunction following stroke. Additionally, it has been discovered that gut microbiota and their metabolites play an essential role in stroke. However, the relationship between the metabolites of gut microbiota and the efficacy of EA is still unclear. Therefore, the aim of this study was to evaluate the mechanism of EA at LI11 and ST36 in the treatment of motor dysfunction after middle cerebral artery occlusion/reperfusion (MCAO/R) in model rats by comparing the differences and correlation between different short-chain fatty acids (SCFAs) and the recovery of motor function. The results indicated that EA at LI11 and ST36 acupoints enhanced the neurological function, motor function, and infarct volume of MCAO/R rats. The levels of acetic acid, propionic acid, and total SCFAs were considerably lower in the MCAO/R group than in the sham group (P < 0.05). Acetic acid, propionic acid, and total SCFA concentrations were substantially higher in the MCAO/R + EA group than in the MCAO/R group (P < 0.05). Finally, Pearson correlation analysis revealed that the propionic acid concentration was substantially favorably connected with the duration on the rotarod (r = 0.633 and P < 0.05) and highly negatively correlated with the modified neurological severity score (mNSS) (r = −0.698 and P < 0.05) and the percentage of cerebral infarct volume (r = −0.729 and P < 0.05). Taken together, these findings indicate that the increase in propionic acid may be one of the mechanisms and targets of EA at LI11 and ST36 acupoints to improve poststroke motor dysfunction in MCAO/R rats.
Collapse
|
170
|
Chen J, Zhuang Y, Zhang Y, Liao H, Liu R, Cheng J, Zhang Z, Sun J, Gao J, Wang X, Chen S, Zhang L, Che F, Wan Q. A synthetic BBB-permeable tripeptide GCF confers neuroprotection by increasing glycine in the ischemic brain. Front Pharmacol 2022; 13:950376. [PMID: 36046828 PMCID: PMC9420865 DOI: 10.3389/fphar.2022.950376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022] Open
Abstract
Background: We and others have previously demonstrated that glycine is neuroprotective in cerebral ischemia-reperfusion injury. But glycine has low permeability to the blood–brain barrier (BBB). To deliver glycine into the ischemic brain to confer neuroprotection, we designed a novel glycine-containing and BBB-permeable tripeptide, the H-glycine-cysteine-phenylalanine-OH (GCF). Methods: For the synthesis of GCF, phenylalanine was included to increase the BBB permeability of the tripeptide. Cysteine was conjugated with glycine to enable the release of glycine from GCF. With the use of immunofluorescence labeling and HPLC assays, we measured the distribution and level of GCF. We used TTC labeling, LDH release, and MTT assays to evaluate the neuroprotective effect of GCF. Results: Following intravenous injection in a rat model of cerebral ischemia-reperfusion injury, GCF was intensively distributed in the ischemic neurons. Intravenous injection of GCF, but not the non-cleavable acetyl-GCF, resulted in the elevation of glycine in the ischemic brain. GCF but not acetyl-GC conferred neuroprotection in ischemic stroke animals. Conclusion: GCF protects against cerebral ischemia-reperfusion injury in the rat. In contrast to peptide drugs that exert therapeutic effect by interfering with signaling interaction, GCF acts as a BBB shuttle and prodrug to deliver glycine to confer neuroprotection, representing a novel therapeutic strategy for acute ischemic stroke.
Collapse
Affiliation(s)
- Juan Chen
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Yang Zhuang
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Ya Zhang
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Huabao Liao
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Rui Liu
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Jing Cheng
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Zhifeng Zhang
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Jiangdong Sun
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| | - Jingchen Gao
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| | - Xiyuran Wang
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| | - Shujun Chen
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| | - Liang Zhang
- Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Fengyuan Che
- Central Laboratory, Department of Neurology, Linyi People’s Hospital, Qingdao University, Linyi, China
- *Correspondence: Qi Wan, ; Fengyuan Che,
| | - Qi Wan
- Department of Pathophysiology, School of Basic Medicine, Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
- Qingdao Gui-Hong Intelligent Medical Technology Co., Ltd., Qingdao, China
- *Correspondence: Qi Wan, ; Fengyuan Che,
| |
Collapse
|
171
|
Hong Y, Lyu J, Zhu L, Wang X, Peng M, Chen X, Deng Q, Gao J, Yuan Z, Wang D, Xu G, Xu M. High-frequency repetitive transcranial magnetic stimulation (rTMS) protects against ischemic stroke by inhibiting M1 microglia polarization through let-7b-5p/HMGA2/NF-κB signaling pathway. BMC Neurosci 2022; 23:49. [PMID: 35927640 PMCID: PMC9351069 DOI: 10.1186/s12868-022-00735-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022] Open
Abstract
Background Microglia assume opposite phenotypes in response to ischemic brain injury, exerting neurotoxic and neuroprotective effects under different ischemic stages. Modulating M1/M2 polarization is a potential therapy for treating ischemic stroke. Repetitive transcranial magnetic stimulation (rTMS) held the capacity to regulate neuroinflammation and astrocytic polarization, but little is known about rTMS effects on microglia. Therefore, the present study aimed to examine the rTMS influence on microglia polarization and the underlying possible molecular mechanisms in ischemic stroke models. Methods Previously reported 10 Hz rTMS protocol that regulated astrocytic polarization was used to stimulate transient middle cerebral artery occlusion (MCAO) rats and oxygen and glucose deprivation/reoxygenation (OGD/R) injured BV2 cells. Specific expression levels of M1 marker iNOS and M2 marker CD206 were measured by western blotting and immunofluorescence. MicroRNA expression changes detected by high-throughput second-generation sequencing were validated by RT-PCR and fluorescence in situ hybridization (FISH) analysis. Dual-luciferase report assay and miRNA knock-down were applied to verify the possible mechanisms regulated by rTMS. Microglia culture medium (MCM) from different groups were collected to measure the TNF-α and IL-10 concentrations, and detect the influence on neuronal survival. Finally, TTC staining and modified Neurological Severity Score (mNSS) were used to determine the effects of MCM on ischemic stroke volume and neurological functions. Results The 10 Hz rTMS inhibited ischemia/reperfusion induced M1 microglia and significantly increased let-7b-5p level in microglia. HMGA2 was predicted and proved to be the target protein of let-7b-5p. HMGA2 and its downstream NF-κB signaling pathway were inhibited by rTMS. Microglia culture medium (MCM) collected from rTMS treated microglia contained lower TNF-α concentration but higher IL-10 concentration than no rTMS treated MCM, reducing ischemic volumes and neurological deficits of MCAO mice. However, knockdown of let-7b-5p by antagomir reversed rTMS effects on microglia phenotype and associated HMGA/NF-κB activation and neurological recovery. Conclusion High-frequency rTMS could alleviate ischemic stroke injury through inhibiting M1 microglia polarization via regulating let-7b-5p/HMGA2/NF-κB signaling pathway in MCAO models. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00735-7.
Collapse
Affiliation(s)
- Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Jinfeng Lyu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Xixi Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Mengna Peng
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Qiwen Deng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Jie Gao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Zhenhua Yuan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China
| | - Di Wang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Mengyi Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, 68# Changle Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
172
|
Investigation of the effects of high cervical spinal cord electrical stimulation on improving neurological dysfunction and its potential mechanism in rats with traumatic brain injury. Neuroreport 2022; 33:509-517. [PMID: 35767234 PMCID: PMC9287104 DOI: 10.1097/wnr.0000000000001811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To explore the effects of high cervical spinal cord electrical stimulation (cSCS) on the recovery of neurological function and its possible mechanism in rats with traumatic brain injury (TBI). 72 rats were randomly divided into: (1) a sham group; (2) a traumatic brain injury (TBI) group; (3) a TBI+cSCS group; (4) a LY294002+TBI+cSCS group. The degree of neurological dysfunction was evaluated by modified Neurological severity score (mNSS). The pathological changes of the brain tissue in the injured area were observed by HE staining, and the apoptosis of neuron cells were observed by TUNEL staining. The expressions of BDNF and VEGFmRNA were detected by polymerase chain reaction (PCR), and the expressions of p-AKT, AKT, Bcl-2, Bax and caspase-3 proteins were detected by western blot. Compared with that of the TBI and LY294002+TBI+cSCS groups, the mNSS of the TBI+cSCS group were significantly lower on day 3 and 7 ( P <0.05). Compared with that in the TBI and LY294002+TBI+cSCS groups, the apoptosis of neuron cells in the TBI+cSCS group decreased significantly ( P < 0.05). Compared with the TBI and LY294002+TBI+cSCS group, the expression of Bcl-2 protein increased and the expressions of Bax and Caspase-3 proteins decreased in the TBI+cSCS group ( P < 0.05). Compared with that in the TBI and LY294002+TBI+cSCS groups, the intensity of p-Akt/Akt in the TBI+cSCS group increased ( P < 0.05). We found that cSCS had a protective effect on neuron cells after craniocerebral injury and could improve neurological dysfunction in rats, the mechanism of which might be that cSCS made the PI3K/Akt pathway more active after TBI.
Collapse
|
173
|
Su Y, Ke C, Li C, Huang C, Wan C. Intermittent hypoxia promotes the recovery of motor function in rats with cerebral ischemia by regulating mitochondrial function. Exp Biol Med (Maywood) 2022; 247:1364-1378. [PMID: 35665627 PMCID: PMC9442452 DOI: 10.1177/15353702221098962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Hypoxia preconditioning is neuroprotective, but the therapeutic effects of intermittent hypoxia were not fully considered. The present study investigated the neuroprotective effect and mechanism of intermittent hypoxia on motor function after cerebral ischemia and explored alternative clinical treatment options. In total, 36 8-week-old male Sprague-Dawley rats were subjected to 60 min of transient middle cerebral artery occlusion (tMCAO) and then randomly divided into a sham-operated group (SHAM), tMCAO-sedentary group (SED), and tMCAO-intermittent hypoxia group (IH). The intervention was performed 1 week after tMCAO and lasted 4 weeks. Rats in the IH group were placed in an animal hypoxic chamber (altitude 5000 m and oxygen concentration of 13%) for 4 h/day and 7 days/week, and rats in the SED group were placed in a normoxic environment for 4 weeks. Body weights, neurological deficit scores, cerebral infarction volume ratios, gait analyses, mitochondrial structure, adenosine triphosphate (ATP) content and AMO-activated protein kinase (AMPK), peroxisome proliferator-activated receptor γ co-activator-1α (PGC-1α), and silencing regulatory protein 3 (Sirt3) expression in the peri-ischemic region brain tissues were detected during the intervention. Compared with the SED group, the body weight of the IH group gradually recovered, and the neurological deficit scores were significantly reduced (P < 0.05). The gait analysis results showed that the pressure of the affected paw and the maximum content area, swing speed, stride length, and other parameters were significantly restored (P < 0.05). The cerebral infarction volume ratio was significantly reduced (P < 0.01). Mitochondrial morphological structure damage in the peri-ischemic region brain tissues recovered, the number was significantly increased (P < 0.05), and the expression of AMPK, PGC-1α, and Sirt3 proteins (P < 0.05), and ATP content were significantly increased (P < 0.05). Intermittent hypoxia may activate the AMPK-PGC-1α-Sirt3 signaling pathway, promote mitochondrial biogenesis, repair mitochondrial ultrastructural damage, and improve mitochondrial function to reduce brain damage and promote motor function recovery in rats with cerebral ischemia.
Collapse
|
174
|
Intracerebral Transplantation of Mesenchymal Stromal Cell Compounded with Recombinant Peptide Scaffold against Chronic Intracerebral Hemorrhage Model. Stem Cells Int 2022; 2022:8521922. [PMID: 35966129 PMCID: PMC9372516 DOI: 10.1155/2022/8521922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background Due to the lack of effective therapies, stem cell transplantation is an anticipated treatment for chronic intracerebral hemorrhage (ICH), and higher cell survival and engraftment are considered to be the key for recovery. Mesenchymal stromal cells (MSCs) compounded with recombinant human collagen type I scaffolds (CellSaics) have a higher potential for cell survival and engraftment compared with solo-MSCs, and we investigated the validity of intracerebral transplantation of CellSaic in a chronic ICH model. Methods Rat CellSaics (rCellSaics) were produced by rat bone marrow-derived MSC (rBMSCs). The secretion potential of neurotrophic factors and the cell proliferation rate were compared under oxygen-glucose deprivation (OGD) conditions. rCellSaics, rBMSCs, or saline were transplanted into the hollow cavity of a rat chronic ICH model. Functional and histological analyses were evaluated, and single-photon emission computed tomography for benzodiazepine receptors was performed to monitor sequential changes in neuronal integrity. Furthermore, human CellSaics (hCellSaics) were transplanted into a chronic ICH model in immunodeficient rats. Antibodies neutralizing brain-derived neurotrophic factor (BDNF) were used to elucidate its mode of action. Results rCellSaics demonstrated a higher secretion potential of trophic factors and showed better cell proliferation in the OGD condition. Animals receiving rCellSaics displayed better neurological recovery, higher intracerebral BDNF, and better cell engraftment; they also showed a tendency for less brain atrophy and higher benzodiazepine receptor preservation. hCellSaics also promoted significant functional recovery, which was reversed by BDNF neutralization. Conclusion Intracerebral transplantation of CellSaics enabled neurological recovery in a chronic ICH model and may be a good option for clinical application.
Collapse
|
175
|
Han Z, Li L, Zhao H, Wang R, Yan F, Tao Z, Fan J, Zheng Y, Zhao F, Huang Y, Tian Y, Li G, Luo Y. MicroRNA-193a-5p Rescues Ischemic Cerebral Injury by Restoring N2-Like Neutrophil Subsets. Transl Stroke Res 2022:10.1007/s12975-022-01071-y. [PMID: 35906328 DOI: 10.1007/s12975-022-01071-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/21/2022] [Accepted: 07/24/2022] [Indexed: 10/16/2022]
Abstract
Circulating neutrophils are activated shortly after stroke and in turn affect the fate of ischemic brain tissue, and microRNAs (miRNA) participate in regulating neuroinflammation. We probed the role of neutrophilic miRNA in ischemic stroke. miR-193a-5p was decreased in circulating neutrophils of acute ischemic stroke (AIS) patients and healthy controls. In another set of AIS patients treated with recombinant tissue plasminogen activator, higher neutrophilic miR-193a-5p levels were associated with favorable outcomes at 3 months and non-symptomatic intracerebral hemorrhage. An experimental stroke model and human neutrophil-like HL-60 cells were further transfected with agomiR-193a-5p/antagomiR-193a-5p or ubiquitin-conjugating enzyme V2 (UBE2V2)-siRNA prior to model induction for in vivo and in vitro studies. Results of 2,3,5-triphenyl tetrazolium chloride staining and neurological function evaluations at post-experimental stroke showed that intravenous agomiR-193a-5p transfusion protected against ischemic cerebral injury in the acute stage and promoted neurological recovery in the subacute stage. This protective role was suggested to correlate with neutrophil N2 transformation based on the N2-like neutrophil proportions in the bone marrow, peripheral blood, and spleen of the experimental stroke model and the measurement of neutrophil phenotype-associated molecule levels. Mechanistically, analyses indicated that UBE2V2 might be a target of miR-193a-5p. Cerebral injury and neuroinflammation aggravated by miR-193a-5p inhibition were reversed by UBE2V2 silencing. In conclusion, miR-193a-5p protects against cerebral ischemic injury by restoring neutrophil N2 phenotype-associated neuroinflammation suppression, likely, in part, via UBE2V2 induction.
Collapse
Affiliation(s)
- Ziping Han
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Lingzhi Li
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Haiping Zhao
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Rongliang Wang
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Feng Yan
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Zhen Tao
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Junfen Fan
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yangmin Zheng
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Fangfang Zhao
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yuyou Huang
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Yue Tian
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Guangwen Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, China
| | - Yumin Luo
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing, 100053, China. .,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China. .,Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
176
|
Liu K, Cai Z, Zhang Q, He J, Cheng Y, Wei S, Yin M. Determination of significant parameters in remote ischemic postconditioning for ischemic stroke in experimental models: A systematic review and meta-analysis study. CNS Neurosci Ther 2022; 28:1492-1508. [PMID: 35896511 PMCID: PMC9437239 DOI: 10.1111/cns.13925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/04/2022] Open
Abstract
Objectives To systematically review studies using remote ischemia postconditioning (RIPostC) for ischemic stroke in experimental models and obtain factors that significantly influence treatment outcomes. Materials and Methods Peer‐reviewed studies were identified and selected based on the eligibility criteria, followed by extraction of data on potentially influential factors related to model preparation, postconditioning, and measure time based on outcome measures including infarct size, neurological scales, and cell tests with autophagy, apoptosis, normal‐neuron, and damaged‐neuron counting. Then, all data were preprocessed, grouped, and meta‐analyzed with the indicator of the standardized mean difference. Results Fifty‐seven studies with 224 experiments (91 for infarct size, 92 for neurological scales, and 41 for cell‐level tests) were included. There was little statistical difference between different model preparations, treated body parts, number of treatments, and sides. And treatment effect was generally a positive correlation with the duration of conditioning time to stroke onset with exceptions at some time points. Based on infarct size, the number of cycles per treatment, duration of occlusion, and release per cycle showed significant differences. Combined with the effect sizes by other measures, the occlusion/release duration of 8–10 min per cycle is better than 5 min, and three cycles per treatment were most frequently used with good effects. Effect also varied when measuring at different times, showing statistical differences in infarct size and most neurological scales. RIPostC is confirmed as an effective therapeutic intervention for ischemic stroke, while the RIPostC‐mediated autophagy level being activated or inhibited remained conflicting. Conclusions Conditioning time, number of cycles per treatment, duration of occlusion, and release per cycle were found to influence the treatment effects of RIPostC significantly. More studies on the relevant influential factors and autophagy mechanisms are warranted.
Collapse
Affiliation(s)
- Kezhou Liu
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Zhengting Cai
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Quanwei Zhang
- School of Management, Hangzhou Dianzi University, Hangzhou, China
| | - Jiatong He
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Yinuo Cheng
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| | - Shaonong Wei
- HDU-ITMO Joint Institute, Hangzhou Dianzi University, Hangzhou, China
| | - Mengjie Yin
- Department of Biomedical Engineering, School of Automation (Artificial Intelligence), Hangzhou Dianzi University, Hangzhou, China
| |
Collapse
|
177
|
Wang Y, Liu X, Zhang W, He S, Zhang Y, Orgah J, Wang Y, Zhu Y. Synergy of "Yiqi" and "Huoxue" components of QishenYiqi formula in ischemic stroke protection via lysosomal/inflammatory mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115301. [PMID: 35436536 DOI: 10.1016/j.jep.2022.115301] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/27/2022] [Accepted: 04/12/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ischemic stroke is one of the leading causes of mortality and long-term disability worldwide. Currently, approved therapies of intravenous thrombolysis and mechanical thrombectomy are limited only to selected patients with rescuable brain tissue. Chinese medicine that benefits Qi (Yiqi, YQ) and activates blood (Huoxue, HX) is widely used in the clinic for treating stroke, but their mechanisms are not well understood yet. We have previously reported that QishenYiqi (QSYQ) formula exerts cerebral protective effect and promotes post-stroke recovery. AIM OF THE STUDY This study aimed to explore the chemical basis and molecular mechanism of anti-stroke therapy of QSYQ and its YQ and HX components further. MATERIALS AND METHODS Serum pharmacochemistry was performed to identify the bioactive constituents in QSYQ for cerebral protection. The survival rate, mNSS test, open field test, gait analysis, cerebral infarction volume, and blood-brain barrier (BBB) integrity were determined to uncover the synergistic and differential contributions of YQ and HX components in a cerebral ischemia/reperfusion injury (CI/RI) model. Bioinformatic mining of QSYQ proteomics data and experimental validation were executed to access the functional mechanism of YQ and HX components. RESULTS Eleven prototype ingredients and six metabolites were successfully identified or tentatively characterized in rat plasma. Therapeutically, YQ and HX components of QSYQ synergistically boosted the survival rate, improved neurological and motor functions, alleviated cerebral infarction as well as protected BBB integrity in CI/RI model in rats. Individually, YQ component contributed more to ameliorating locomotive ability than that of HX component. Mechanistically, HX component played a more prominent role in the modulation of galectin-3 mediated inflammation whereas YQ component regulated lysosomal-autophagy signaling. CONCLUSIONS This study identifies major prototype ingredients and metabolites of QSYQ in plasma which may contribute to its cerebral protection. YQ and HX components of QSYQ differentially and synergistically protect the brain from CI/RI by regulating galectin-3-mediated inflammation and lysosomal-autophagy signaling. These findings demonstrate that a maximal stroke protection by a component-based Chinese medicine could be attributed to the combination of its individual components via different mechanisms. It may shed new light on our understanding of the TCM principle of tonifying Qi and activating blood, particularly in a setting of ischemic stroke.
Collapse
Affiliation(s)
- Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, XiHu District, Hangzhou, 310058, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Wen Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Taiping Qiao Street No.27, Xicheng District, Beijing, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yiqian Zhang
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin Tasly Holding Group Co, Ltd, Tianjin, China
| | - John Orgah
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, XiHu District, Hangzhou, 310058, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin, 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biomedicine, 220 Dongting Road, TEDA, Tianjin, 300457, China.
| |
Collapse
|
178
|
Zheng S, Mu S, Li J, Zhang S, Wei L, Wang M, Xu Y, Wang S. Cerebral venous hemodynamic responses in a mouse model of traumatic brain injury. Brain Res 2022; 1792:148014. [PMID: 35839929 DOI: 10.1016/j.brainres.2022.148014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/28/2022] [Accepted: 07/10/2022] [Indexed: 11/24/2022]
Abstract
Traumatic brain injury (TBI) is a serious public health problem that endangers human health and is divided into primary and secondary injuries. Previous work has confirmed that changes in cerebral blood flow (CBF) are related to the progression of secondary injury, although clinical studies have shown that CBF monitoring cannot fully and accurately evaluate disease progression. These studies have almost ignored the monitoring of venous blood flow; however, as an outflow channel of the cerebral circulation, it warrants discussion. To explore the regulation of venous blood flow after TBI, the present study established TBI mouse models of different severities, observed changes in cerebral venous blood flow by laser speckle flow imaging, and recorded intracranial pressure (ICP) after brain injury to evaluate the correlation between venous blood flow and ICP. Behavioral and histopathological assessments were performed after the intervention. The results showed that there was a significant negative correlation between ICP and venous blood flow (r = -0.795, P < 0.01), and both recovered to varying degrees in the later stages of observation. The blood flow changes in regional microvessels were similar to those in venous, and the expression of angiogenesis proteins around the impact area was significantly increased. In conclusion, this study based on the TBI mouse model, recorded the changes in venous blood flow and ICP and revealed that venous blood flow can be used as an indicator of the progression of secondary brain injury.
Collapse
Affiliation(s)
- Shaorui Zheng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Department of Neurosurgery, Affiliated Hospital of Putian University, Putian 351100, China
| | - Shuwen Mu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China
| | - Jun Li
- Department of Neurosurgery, The 900th Hospital of Joint Logistic Support Force, Fujian Medical University Fuzong Clinical College, Fuzhou 350025, China
| | - Shangming Zhang
- Department of Neurosurgery, The 900th Hospital of Joint Logistic Support Force, Fujian Medical University Fuzong Clinical College, Fuzhou 350025, China
| | - Liangfeng Wei
- Department of Neurosurgery, The 900th Hospital of Joint Logistic Support Force, Fujian Medical University Fuzong Clinical College, Fuzhou 350025, China
| | - Mingyue Wang
- Department of Pathology, The 900th Hospital of Joint Logistic Support Force, Fujian Medical University Fuzong Clinical College, Fuzhou 350025, China
| | - Yongjun Xu
- Laboratory of Basic Medicine, The 900th Hospital of Joint Logistic Support Force, Fujian Medical University Fuzong Clinical College, Fuzhou 350025, China.
| | - Shousen Wang
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou 350025, China; Department of Neurosurgery, The 900th Hospital of Joint Logistic Support Force, Fujian Medical University Fuzong Clinical College, Fuzhou 350025, China.
| |
Collapse
|
179
|
Zi Y, Yang K, He J, Wu Z, Liu J, Zhang W. Strategies to enhance drug delivery to solid tumors by harnessing the EPR effects and alternative targeting mechanisms. Adv Drug Deliv Rev 2022; 188:114449. [PMID: 35835353 DOI: 10.1016/j.addr.2022.114449] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/03/2022] [Accepted: 07/06/2022] [Indexed: 12/20/2022]
Abstract
The Enhanced Permeability and Retention (EPR) effect has been recognized as the central paradigm in tumor-targeted delivery in the last decades. In the wake of this concept, nanotechnologies have reached phenomenal levels in research. However, clinical tumors display a poor manifestation of EPR effect. Factors including tumor heterogeneity, complicating tumor microenvironment, and discrepancies between laboratory models and human tumors largely contribute to poor efficiency in tumor-targeted delivery and therapeutic failure in clinical translation. In this article, approaches for evaluation of EPR effect in human tumor were overviewed as guidance to employ EPR effect for cancer treatment. Strategies to augment EPR-mediated tumoral delivery are discussed in different dimensions including enhancement of vascular permeability, depletion of tumor extracellular matrix and optimization of nanoparticle design. Besides, the recent development in alternative tumor-targeted delivery mechanisms are highlighted including transendothelial pathway, endogenous cell carriers and non-immunogenic bacteria-mediated delivery. In addition, the emerging preclinical models better reflect human tumors are introduced. Finally, more rational applications of EPR effect in other disease and field are proposed. This article elaborates on fundamental reasons for the gaps between theoretical expectation and clinical outcomes, attempting to provide some perspective directions for future development of cancer nanomedicines in this still evolving landscape.
Collapse
Affiliation(s)
- Yixuan Zi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Kaiyun Yang
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zimei Wu
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
180
|
A previous hemorrhagic stroke protects against a subsequent stroke via microglia alternative polarization. Commun Biol 2022; 5:654. [PMID: 35780249 PMCID: PMC9250506 DOI: 10.1038/s42003-022-03621-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022] Open
Abstract
Microglia in hemorrhagic stroke contribute to both acute-phase exacerbation and late-phase attenuation of injury. Here, by using the mouse model, we reported that the shift in polarization of microglia from M1 to M2 phenotype could be altered by a past ‘mini’ stroke, resulting in better neurological function recovery, faster attenuation of lesion volume, and better survival. In mice with a previous stroke, M2 predominance appeared markedly in advance compared to mice without a previous stroke. Mechanistically, the RBC-mediated M2 polarization of microglia was synergistically enhanced by T cells: microglia cocultured with RBCs alone resulted in mild alterations to M2 markers, whereas in the presence of T cells, they expressed an early and sustained M2 response. These results suggest that by harnessing the microglia-mediated M2 polarization response, we could help mitigate devastating sequelae before a prospective hemorrhagic stroke even happens. Microglia shift in polarization from an M1 to a M2 phenotype following a past “mini” stroke, resulting in better neurological function recovery, faster attenuation of lesion volume, and better survival.
Collapse
|
181
|
Karizmeh MS, Shabani M, Shabani M, Sardari M, Babaei JF, Nabavizadeh F, Sadr SS, Adeli S. Preconditioning exercise reduces hippocampal neuronal damage via increasing Klotho expression in ischemic rats. Brain Res Bull 2022; 188:133-142. [DOI: 10.1016/j.brainresbull.2022.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 01/24/2023]
|
182
|
Hou K, Xiao ZC, Dai HL. p38 MAPK Endogenous Inhibition Improves Neurological Deficits in Global Cerebral Ischemia/Reperfusion Mice. Neural Plast 2022; 2022:3300327. [PMID: 35811833 PMCID: PMC9259354 DOI: 10.1155/2022/3300327] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/02/2022] [Accepted: 05/31/2022] [Indexed: 12/02/2022] Open
Abstract
Cerebral ischemia/reperfusion (I/R) injury is a complex pathophysiological process that can lead to neurological function damage and the formation of cerebral infarction. The p38 MAPK pathway has attracted considerable attention in cerebral I/R injury (IRI), but little research has been carried out on its direct role in vivo. In this study, to observe the effects of p38 MAPK endogenous inhibition on cerebral IRI, p38 heterozygous knockdown (p38KI/+) mice were used. We hypothesized that p38 signaling might be involved in I/R injury and neurological damage reduction and that neurological behavioral deficits improve when p38 MAPK is inhibited. First, we examined the neurological damage and neurological behavioral deficit effects of I/R injury in WT mice. Cerebral I/R injury was induced by the bilateral common carotid artery occlusion (BCCAO) method. The cerebral infarction area and volume were assessed and analyzed by 2,3,5-triphenyltetrazolium chloride (TTC) staining. p38 MAPK and caspase-3 were detected by western blotting. Neuronal apoptosis was measured using TUNEL staining. Neurological deficits were detected by behavioral testing. Furthermore, to assess whether these neuroprotective effects occurred when p38 MAPK was inhibited, p38 heterozygous knockdown (p38KI/+) mice were used. We found that p38 MAPK endogenous inhibition rescued hippocampal cell apoptosis, reduced ischemic penumbra, and improved neurological behavioral deficits. These findings showed that p38 MAPK endogenous inhibition had a neuroprotective effect on IRI and that p38 MAPK may be a potential therapeutic target for cerebral IRI.
Collapse
Affiliation(s)
- Kun Hou
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650500, China
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Zhi-cheng Xiao
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
- Department of Anatomy and Developmental Biology, Monash University, Clayton 3800, Australia
| | - Hai-Long Dai
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| |
Collapse
|
183
|
Wang X, Zhang C, Li Y, Xu T, Xiang J, Bai Y, Zhang Y, Wang Q, Zhang T, Liao L. High-Throughput mRNA Sequencing Reveals Potential Therapeutic Targets of Febuxostat in Secondary Injury After Intracerebral Hemorrhage. Front Pharmacol 2022; 13:833805. [PMID: 35814252 PMCID: PMC9260037 DOI: 10.3389/fphar.2022.833805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/12/2022] [Indexed: 11/13/2022] Open
Abstract
Febuxostat is a urate-lowering medication for the treatment of patients with gout. This study was performed to elucidate the effects and underlying mechanisms of febuxostat on neuronal injury induced by intracerebral hemorrhage (ICH) in mice. The results showed that the administration of febuxostat improved neurological severity scores and blood–brain barrier (BBB) permeability. Moreover, febuxostat attenuated neuronal cell death and cytokine levels compared with the ICH group. Next, we conducted a transcriptome analysis of the neuroprotective effects of febuxostat. The overlapping significant differentially expressed genes (DEGs) were identified. Gene ontology (GO) analysis revealed that the overlapping significant DEGs were most enriched in five items. The intersecting DEGs of the aforementioned five pathways were Wisp1, Wnt7b, Frzb, and Pitx2. In addition, GO terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways revealed that DEGs were mainly involved in the wnt signaling pathway. Furthermore, the expression of Wisp1 and Wnt7b in the perihematomal region at 72 h post-ICH was observed. The results showed that both Wisp1 and Wnt7b were increased in the ICH group and were decreased by the administration of febuxostat. Taken together, the study showed that febuxostat protected against secondary brain injury after ICH and the Wnt7b-Wisp1 pathway was closely related to neuroprotective effects.
Collapse
Affiliation(s)
- Xueyan Wang
- Department of Pharmacy, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Chenyu Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Ting Xu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jin Xiang
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Bai
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Qi Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Tiejun Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Tiejun Zhang, ; Linchuan Liao,
| | - Linchuan Liao
- Department of Forensic Analytical Toxicology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Tiejun Zhang, ; Linchuan Liao,
| |
Collapse
|
184
|
Yang G, Fan X, Mazhar M, Yang S, Xu H, Dechsupa N, Wang L. Mesenchymal Stem Cell Application and Its Therapeutic Mechanisms in Intracerebral Hemorrhage. Front Cell Neurosci 2022; 16:898497. [PMID: 35769327 PMCID: PMC9234141 DOI: 10.3389/fncel.2022.898497] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/18/2022] [Indexed: 12/15/2022] Open
Abstract
Intracerebral hemorrhage (ICH), a common lethal subtype of stroke accounting for nearly 10–15% of the total stroke disease and affecting two million people worldwide, has a high mortality and disability rate and, thus, a major socioeconomic burden. However, there is no effective treatment available currently. The role of mesenchymal stem cells (MSCs) in regenerative medicine is well known owing to the simplicity of acquisition from various sources, low immunogenicity, adaptation to the autogenic and allogeneic systems, immunomodulation, self-recovery by secreting extracellular vesicles (EVs), regenerative repair, and antioxidative stress. MSC therapy provides an increasingly attractive therapeutic approach for ICH. Recently, the functions of MSCs such as neuroprotection, anti-inflammation, and improvement in synaptic plasticity have been widely researched in human and rodent models of ICH. MSC transplantation has been proven to improve ICH-induced injury, including the damage of nerve cells and oligodendrocytes, the activation of microglia and astrocytes, and the destruction of blood vessels. The improvement and recovery of neurological functions in rodent ICH models were demonstrated via the mechanisms such as neurogenesis, angiogenesis, anti-inflammation, anti-apoptosis, and synaptic plasticity. Here, we discuss the pathological mechanisms following ICH and the therapeutic mechanisms of MSC-based therapy to unravel new cues for future therapeutic strategies. Furthermore, some potential strategies for enhancing the therapeutic function of MSC transplantation have also been suggested.
Collapse
Affiliation(s)
- Guoqiang Yang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Acupuncture and Rehabilitation, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Maryam Mazhar
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Houping Xu
- Preventive Treatment Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- *Correspondence: Nathupakorn Dechsupa,
| | - Li Wang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
- Li Wang,
| |
Collapse
|
185
|
Wang M, Zhou C, Yu L, Kong D, Ma W, Lv B, Wang Y, Wu W, Zhou M, Cui G. Upregulation of MDH1 acetylation by HDAC6 inhibition protects against oxidative stress-derived neuronal apoptosis following intracerebral hemorrhage. Cell Mol Life Sci 2022; 79:356. [PMID: 35678904 PMCID: PMC11073123 DOI: 10.1007/s00018-022-04341-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/18/2022] [Accepted: 04/30/2022] [Indexed: 11/25/2022]
Abstract
Oxidative stress impairs functional recovery after intracerebral hemorrhage (ICH). Histone deacetylase 6 (HDAC6) plays an important role in the initiation of oxidative stress. However, the function of HDAC6 in ICH and the underlying mechanism of action remain elusive. We demonstrated here that HDAC6 knockout mice were resistant to oxidative stress following ICH, as assessed by the MDA and NADPH/NADP+ assays and ROS detection. HDAC6 deficiency also resulted in reduced neuronal apoptosis and lower expression levels of apoptosis-related proteins. Further mechanistic studies showed that HDAC6 bound to malate dehydrogenase 1 (MDH1) and mediated-MDH1 deacetylation on the lysine residues at position 121 and 298. MDH1 acetylation was inhibited in HT22 cells that were challenged with ICH-related damaging agents (Hemin, Hemoglobin, and Thrombin), but increased when HDAC6 was inhibited, suggesting an interplay between HDAC6 and MDH1. The acetylation-mimetic mutant, but not the acetylation-resistant mutant, of MDH1 protected neurons from oxidative injury. Furthermore, HDAC6 inhibition failed to alleviate brain damage after ICH when MDH1 was knockdown. Taken together, our study showed that HDAC6 inhibition protects against brain damage during ICH through MDH1 acetylation.
Collapse
Affiliation(s)
- Miao Wang
- Department of Geriatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China.
- Department of Neurology, Xuzhou First People's Hospital, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chao Zhou
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Lu Yu
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Delian Kong
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurology, Xuzhou First People's Hospital, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Weijing Ma
- Department of Neurology, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bingchen Lv
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Yan Wang
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Weifeng Wu
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Mingyue Zhou
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China
| | - Guiyun Cui
- Institute of Nervous System Diseases and Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, No. 99 West Huaihai Road, Xuzhou, 221006, Jiangsu Province, People's Republic of China.
| |
Collapse
|
186
|
Li J, Ren H, Wang Y, Hoang DM, Li Y, Yao X. Mechanism of Stat1 in the neuronal Ca 2+ overload after intracerebral hemorrhage via the H3K27ac/Trpm7 axis. J Neurophysiol 2022; 128:253-262. [PMID: 35642851 DOI: 10.1152/jn.00083.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is classified as a subtype of stroke and Calcium (Ca2+) overload is a catalyst for ICH. This study explored the mechanisms of Stat1 (signal transducer and activator of transcription 1) in the neuronal Ca2+ overload after ICH. ICH mouse models and in vitro cell models were established. Stat1 and transient receptor potential melastatin 7 (Trpm7) were detected up-regulated in ICH models. Afterwards, the mice were infected with the lentivirus containing sh-Stat1, and HT22 cells were treated with si-Stat1 and the lentivirus containing pcDNA3.1-Trpm7. The neurologic functional impairment, histopathological damage, and Nissl body in mice were all measured. HT22 cell viability and apoptosis were identified. The levels of Ca2+, Trpm7 mRNA, H3K27 acetylation (H3K27ac), CaMKII-α, and p-Stat1 protein in the tissues and cells were determined. We found that silencing Stat1 alleviated ICH damage and repressed the neuronal Ca2+ overload after ICH. H3K27ac enrichment in the Trpm7 promoter region was examined and we found that p-Stat1 accelerated Trpm7 transcription via promoting H3K27ac in the Trpm7 promoter region. Besides, Trpm7 overexpression increased Ca2+ overload and aggravated ICH. Overall, p-Stat1 promoted Trpm7 transcription and further aggravated the Ca2+ overload after ICH.
Collapse
Affiliation(s)
- Jialin Li
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China.,Tianjin Key Labaratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Hecheng Ren
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Yanbing Wang
- Tianjin Key Labaratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China.,Department of Nursing, Tianjin Huanhu Hospital, Tianjin, China
| | - Dung Minh Hoang
- Department of Radiology, New York University School of Medicine, New York, NY, United States
| | - Yongsheng Li
- Department of Neurology, New York University School of Medicine, New York, NY, United States
| | - Xiuhua Yao
- Tianjin Key Labaratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
187
|
Lingling D, Miaomiao Q, Yili L, Hongyun H, Yihao D. Attenuation of histone H4 lysine 16 acetylation (H4K16ac) elicits a neuroprotection against ischemic stroke by alleviating the autophagic/lysosomal dysfunction in neurons at the penumbra. Brain Res Bull 2022; 184:24-33. [DOI: 10.1016/j.brainresbull.2022.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/06/2022] [Accepted: 03/24/2022] [Indexed: 11/02/2022]
|
188
|
Zheng J, Mao X, Wang D, Xia S. Preconditioned MSCs Alleviate Cerebral Ischemia-Reperfusion Injury in Rats by Improving the Neurological Function and the Inhibition of Apoptosis. Brain Sci 2022; 12:631. [PMID: 35625017 PMCID: PMC9140028 DOI: 10.3390/brainsci12050631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 02/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have great application prospects in the treatment of ischemic injury. However, their long-time cultivation before transplantation and poor survival after transplantation greatly limit the therapeutic effect and applications. This study aimed to investigate whether MSCs under the ischemic microenvironment could improve their survival and better alleviate cerebral ischemic injury. Firstly, we used ischemic brain tissue to culture MSCs and evaluated the functional changes of MSCs. Then a middle cerebral artery occlusion (MCAO) model was induced in rats, and the pretreated MSCs were injected via the tail vein. The adhesive removal test, rotarod test, modified neurological severity score, and pathological analyses were applied to assess the rats' neurological function. Then the expression of neuron and apoptosis related markers was detected. The results indicated that ischemic brain tissue pretreated MSCs promoted the proliferation and the release of the growth factors of MSCs. Meanwhile, in MCAO model rats, transplantation of pretreated MSCs enhanced the neurogenesis, attenuated behavioral changes, reduced infarct size, and inhibited apoptosis. The expression of B-cell lymphoma-2 (Bcl-2), brain-derived neurotrophic factor (BDNF), glial fibrillary acidic protein (GFAP), NF-L, and NeuN were increased, while BCL2-Associated X (Bax) and Caspase-3 decreased. Our results suggest that MSCs pretreatment with stroke brain tissue could be an effective strategy in treating cerebral ischemic injury.
Collapse
Affiliation(s)
- Jin Zheng
- Department of Neurology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Xueyu Mao
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai 201199, China; (X.M.); (D.W.); (S.X.)
| | - Delong Wang
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai 201199, China; (X.M.); (D.W.); (S.X.)
| | - Shiliang Xia
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai 201199, China; (X.M.); (D.W.); (S.X.)
| |
Collapse
|
189
|
Xiao L, Zheng H, Li J, Zeng M, He D, Liang J, Sun K, Luo Y, Li F, Ping B, Yuan W, Zhou H, Wang Q, Sun H. Targeting NLRP3 inflammasome modulates gut microbiota, attenuates corticospinal tract injury and ameliorates neurobehavioral deficits after intracerebral hemorrhage in mice. Biomed Pharmacother 2022; 149:112797. [PMID: 35279596 DOI: 10.1016/j.biopha.2022.112797] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 11/27/2022] Open
Abstract
Intracerebral hemorrhage (ICH) has a high mortality and disability rate. Fewer studies focus on white matter injury (WMI) after ICH, especially the corticospinal tract (CST) injury located in the spinal cord, which correlates with motor impairments. Recent studies have shown that gut microbiota dysbiosis occurs after ICH. Furthermore, NLRP3 inflammasome can be activated after ICH, resulting in inflammatory cascade reactions and aggravating brain injury. However, no direct and causal correlation among NLRP3 inflammasome inhibition, altered gut microbiota, and CST injury following ICH has been reported. This study aimed to investigate the effect of MCC950, a selective NLRP3 inflammasome inhibitor, on the gut microbiota and CST injury after ICH. We observed that compared with the sham group, the members of Firmicutes, such as Faecalibaculum and Dubosiella, were depleted in the ICH + Vehicle group, whereas the members of Proteobacteria and Campilobacterota were enriched, such as Enterobacter and Helicobacter. After treatment with MCC950, the Bacteroides, Bifidobacterium and Paenibacillus were relatively abundant in the gut flora of mice. Moreover, we observed CST injury located in cervical enlargement of the spinal cord, and MCC950 alleviated it. Furthermore, treatment with MCC950 decreased the mNSS score and brain water content in ICH. Taken together, the present study showed that MCC950 modulated gut microbiota, effectively attenuated CST injury located in cervical enlargement of the spinal cord, and ameliorated neurological deficits after ICH. This study provided a novel report that links NLRP3 inflammasome inhibition, gut microbiota alteration and CST injury following ICH and profound implications for ICH treatment.
Collapse
Affiliation(s)
- Linglong Xiao
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Huaping Zheng
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jing Li
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Meiqin Zeng
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Dian He
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jianhao Liang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Kaijian Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yunhao Luo
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Feng Li
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Baohong Ping
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Wen Yuan
- Laboratory Animal Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hongwei Zhou
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qinghua Wang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Haitao Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre for Brain Science and BrainInspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
190
|
Wang Y, Wu H, Han Z, Sheng H, Wu Y, Wang Y, Guo X, Zhu Y, Li X, Wang Y. Guhong injection promotes post-stroke functional recovery via attenuating cortical inflammation and apoptosis in subacute stage of ischemic stroke. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154034. [PMID: 35276592 DOI: 10.1016/j.phymed.2022.154034] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND As a leading cause of death and disability, alternative therapies for stroke are still limited by its complicated pathophysiological manifestations. Guhong injection (GHI), consisting of safflower aqueous extract and aceglutamide, has been widely applied for the clinical treatment of cerebrovascular diseases, especially ischemic stroke and post-stroke recovery, in China. Recently, a series of studies have reported the positive effect of GHI against cerebral ischemia/reperfusion injury via targeting various molecular mechanisms. However, questions remain on whether treatment with GHI contributes to better functional recovery after stroke and if so, the potential mechanisms and active substances. PURPOSE The aim of this work was to explore the potential therapeutic possibilities of GHI for the neurological and behavioral recovery after stroke and to investigate the underlying molecular mechanisms as well as active substances. METHODS The neural and motor deficits as well as cortical lesions after GHI treatment were investigated in a mouse model of transient ischemic stroke. Based on the substance identification of GHI, network pharmacology combined with an experimental verification method was used to systematically decipher the biological processes and signaling pathways closely related to GHI intervention in response to post-stroke functional outcomes. Subsequently, ingenuity pathway analysis (IPA) analysis was performed to determine the anti-stroke active substances targeting to the hub targets involved in the significant molecular pathways regulated by GHI treatment. RESULTS Therapeutically, administration of GHI observably ameliorated the post-stroke recovery of neural and locomotor function as well as reduced infarct volume and histopathological damage to the cerebral cortex in subacute stroke mice. According to 26 identified or tentatively characterized substances in GHI, the compound-target-pathway network was built. Bioinformatics analysis suggested that inflammatory and apoptotic pathways were tightly associated with the anti-stroke effect of GHI. Based on protein-protein interaction network analysis, the hub targets (such as NF-κB p65, TNF-α, IL-6, IL-1β, Bax, Bcl-2, and Caspase-3) involved in inflammation and apoptosis were selected. On the one hand, immunofluorescence and ELISA results showed that GHI (10 ml/kg) treatment obviously reduced NF-κB p65 nuclear translocation as well as decreased the abnormally elevated concentrations of proinflammatory cytokines (TNF-α, IL-6, and IL-1β) in damaged cortex tissues. On the other hand, GHI (10 ml/kg) treatment significantly downregulated the number of TUNEL-positive apoptotic cells in ischemic cortex and effectively restored the abnormal expression of Bax, Bcl-2, and Caspase-3. Based on the results of IPA, hydroxysafflor yellow A, baicalin, scutellarin, gallic acid, syringin, chlorogenic acid, kaempferol, kaempferol-3-O-β-rutinoside, and rutin acted synergistically on core targets, which could be considered as the active substances of GHI. CONCLUSION Overall, the current findings showed that the beneficial action of GHI on improving post-stroke functional recovery of subacute stroke mice partly via the modulation of cortical inflammation and apoptosis. These findings not only provide a reliable reference for the clinical application of GHI, but also shed light on a promising alternative therapeutic strategy for ischemic stroke patients.
Collapse
Affiliation(s)
- Yule Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Huimin Wu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Zhu Han
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Hongda Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Yuhan Wu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China
| | - Yingchao Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, 291 Fucheng Road, Qiantang District, Hangzhou 310020, China
| | - Xinran Guo
- School of Humanities and Management, Wannan Medical College, 34 Yinhunan Road, Jinghu District, Wuhu 241001, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, Jinghai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xuecai Li
- Tonghua Guhong Pharmaceutical Co., Ltd., 5099 Jianguo Road, Meihekou 135099, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310012, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, Jinghai District, Tianjin 301617, China.
| |
Collapse
|
191
|
Kong L, Li W, Chang E, Wang W, Shen N, Xu X, Wang X, Zhang Y, Sun W, Hu W, Xu P, Liu X. mtDNA-STING Axis Mediates Microglial Polarization via IRF3/NF-κB Signaling After Ischemic Stroke. Front Immunol 2022; 13:860977. [PMID: 35450066 PMCID: PMC9017276 DOI: 10.3389/fimmu.2022.860977] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/16/2022] [Indexed: 12/31/2022] Open
Abstract
Neuroinflammation is initiated in response to ischemic stroke, and is usually characterized by microglial activation and polarization. Stimulator of interferon genes (STING) has been shown to play a critical role in anti-tumor immunity and inflammatory diseases. Nevertheless, the effect and underlying mechanisms of STING on microglial polarization after ischemic stroke remain unclarified. In this study, acute ischemic stroke was simulated using a model of middle cerebral artery occlusion (MCAO) at adult male C57BL/6 mice in vivo and the BV2 microglia oxygen-glucose deprivation/reperfusion (OGD/R) model in vitro. The specific STING inhibitor C-176 was administered intraperitoneally at 30min after MCAO. We found that the expression of microglial STING was increased following MCAO and OGD/R. Pharmacologic inhibition of STING with C-176 reduced the ischemia/reperfusion (I/R)-induced brain infarction, edema and neuronal injury. Moreover, blockade of STING improved neurological performance and cognitive function and attenuated neuronal degeneration in the hippocampus after MCAO. Mechanistically, both in vivo and in vitro, we delineated that STING could promote the polarization of microglia towards the M1 phenotype and restrain M2 microglia polarization via downstream pathways, including interferon regulatory factor 3 (IRF3) and nuclear factor-κB (NF-κB). In addition, mitochondrial DNA (mtDNA), which is released to microglial cytoplasm induced by I/R injury, could facilitate microglia towards M1 modality through STING signaling pathway. Treatment with C-176 abolished the detrimental effects of mtDNA on stroke outcomes. Taken together, these findings suggest that STING, activated by mtDNA, could polarize microglia to the M1 phenotype following MCAO. Inhibition of STING may serve as a potential therapeutic strategy to mitigate neuroinflammation after ischemic stroke.
Collapse
Affiliation(s)
- Lingqi Kong
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenyu Li
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - E Chang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wuxuan Wang
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Nan Shen
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiang Xu
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinyue Wang
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Zhang
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wen Sun
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Hu
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Pengfei Xu
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinfeng Liu
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
192
|
Chen Y, Yang B, Xu L, Shi Z, Han R, Yuan F, Ouyang J, Yan X, Ostrikov KK. Inhalation of Atmospheric-Pressure Gas Plasma Attenuates Brain Infarction in Rats With Experimental Ischemic Stroke. Front Neurosci 2022; 16:875053. [PMID: 35516812 PMCID: PMC9063166 DOI: 10.3389/fnins.2022.875053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies suggest the potential efficacy of neuroprotective effects of gaseous atmospheric-pressure plasma (APP) treatment on neuronal cells. However, it remains unclear if the neuroprotective properties of the gas plasmas benefit the ischemic stroke treatment, and how to use the plasmas in the in vivo ischemic stroke models. Rats were subjected to 90 min middle cerebral artery occlusion (MCAO) to establish the ischemic stroke model and then intermittently inhaled the plasma for 2 min at 60 min MCAO. The regional cerebral blood flow (CBF) was monitored. Animal behavior scoring, magnetic resonance imaging (MRI), 2,3,5-triphenyltetrazolium chloride (TTC) staining, and hematoxylin and eosin (HE) staining were performed to evaluate the therapeutic efficacy of the gas plasma inhalation on MCAO rats. Intermittent gas plasma inhalation by rats with experimental ischemic stroke could improve neurological function, increase regional CBF, and decrease brain infarction. Further MRI tests showed that the gas plasma inhalation could limit the ischemic lesion progression, which was beneficial to improve the outcomes of the MCAO rats. Post-stroke treatment with intermittent gas plasma inhalation could reduce the ischemic lesion progression and decrease cerebral infarction volume, which might provide a new promising strategy for ischemic stroke treatment.
Collapse
Affiliation(s)
- Ye Chen
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bingyan Yang
- School of Physics, Beijing Institute of Technology, Beijing, China
| | - Lixin Xu
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhongfang Shi
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruoyu Han
- School of Physics, Beijing Institute of Technology, Beijing, China
| | - Fang Yuan
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiting Ouyang
- School of Physics, Beijing Institute of Technology, Beijing, China
- *Correspondence: Jiting Ouyang,
| | - Xu Yan
- Department of Pathophysiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Xu Yan,
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics and Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
193
|
Zhang Q, Kong WL, Yuan JJ, Chen Q, Gong CX, Liu L, Wang FX, Huang JC, Yang GQ, Zhou K, Xu R, Xiong XY, Yang QW. Redistribution of Histone Marks on Inflammatory Genes Associated With Intracerebral Hemorrhage-Induced Acute Brain Injury in Aging Rats. Front Neurosci 2022; 16:639656. [PMID: 35495024 PMCID: PMC9051396 DOI: 10.3389/fnins.2022.639656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
The contribution of histone mark redistribution to the age-induced decline of endogenous neuroprotection remains unclear. In this study, we used an intracerebral hemorrhage (ICH)-induced acute brain injury rat model to study the transcriptional and chromatin responses in 13- and 22-month-old rats. Transcriptome analysis (RNA-seq) revealed that the expression of neuroinflammation-associated genes was systematically upregulated in ICH rat brains, irrespective of age. Further, we found that interferon-γ (IFN-γ) response genes were activated in both 13- and 22-month-old rats. Anti-IFN-γ treatment markedly reduced ICH-induced acute brain injury in 22-month-old rats. At the chromatin level, ICH induced the redistribution of histone modifications in the promoter regions, especially H3K4me3 and H3K27me3, in neuroinflammation-associated genes in 13- and 22-month-old rats, respectively. Moreover, ICH-induced histone mark redistribution and gene expression were found to be correlated. Collectively, these findings demonstrate that histone modifications related to gene expression are extensively regulated in 13- and 22-month-old rats and that anti-IFN-γ is effective for ICH treatment, highlighting the potential of developing therapies targeting histone modifications to cure age-related diseases, including brain injury and neuroinflammation.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Wei-lin Kong
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Jun-Jie Yuan
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Qiong Chen
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Chang-Xiong Gong
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Fa-Xiang Wang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Jia-Cheng Huang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Guo-Qiang Yang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Kai Zhou
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Rui Xu
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
- Acupuncture and Tuina School, Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan, Chengdu, China
- Xiao-Yi Xiong ;
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Qing-Wu Yang
| |
Collapse
|
194
|
Wang A, Zhao W, Yan K, Huang P, Zhang H, Ma X. Preclinical Evidence of Paeoniflorin Effectiveness for the Management of Cerebral Ischemia/Reperfusion Injury: A Systematic Review and Meta-Analysis. Front Pharmacol 2022; 13:827770. [PMID: 35462929 PMCID: PMC9032804 DOI: 10.3389/fphar.2022.827770] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/24/2022] [Indexed: 01/01/2023] Open
Abstract
Background: Vessel recanalization is the main treatment for ischemic stroke; however, not all patients benefit from it. This lack of treatment benefit is related to the accompanying ischemia-reperfusion (I/R) injury. Therefore, neuroprotective therapy for I/R Injury needs to be further studied. Paeonia lactiflora Pall. is a commonly used for ischemic stroke management in traditional Chinese medicine; its main active ingredient is paeoniflorin (PF). We aimed to determine the PF’s effects and the underlying mechanisms in instances of cerebral I/R injury.Methods: We searched seven databases from their inception to July 2021.SYRCLE’s risk of bias tool was used to assess methodological quality. Review Manager 5.3 and STATA 12.0 software were used for meta-analysis.Results: Thirteen studies, including 282 animals overall, were selected. The meta-analyses showed compared to control treatment, PF significantly reduced neurological severity scores, cerebral infarction size, and brain water content (p = 0.000). In the PF treatment groups, the apoptosis cells and levels of inflammatory factors (IL-1β) decreased compared to those in the control groups (p = 0.000).Conclusion: Our results suggest that PF is a promising therapeutic for cerebral I/R injury management. However, to evaluate the effects and safety of PF in a more accurate manner, additional preclinical studies are necessary.
Collapse
Affiliation(s)
- Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Zhao
- Yidu Central Hospital of Weifang, Weifang, China
| | - Kaituo Yan
- Yidu Central Hospital of Weifang, Weifang, China
| | - Pingping Huang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongwei Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- *Correspondence: Xiaochang Ma,
| |
Collapse
|
195
|
Positive Effect of α-Asaronol on the Incidence of Post-Stroke Epilepsy for Rat with Cerebral Ischemia-Reperfusion Injury. Molecules 2022; 27:molecules27061984. [PMID: 35335346 PMCID: PMC8952411 DOI: 10.3390/molecules27061984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 11/30/2022] Open
Abstract
In the present study, we confirmed that α-asaronol, which is a product of the active metabolites of alpha Asarone, did not affect n-butylphthalide efficacy when n-butylphthalide and α-asaronol were co-administered to rats with cerebral ischemia-reperfusion injury. Our research revealed that the co-administration of α-asaronol and n-butylphthalide could further improve neurological function, reduce brain infarct volume, increase the number of Nissl bodies, and decrease the ratios of apoptotic cells and the expression of the caspase-3 protein for cerebral ischemia-reperfusion injury model compared to n-butylphthalide alone. Additionally, α-asaronol could significantly decrease the incidence of post-stroke epilepsy versus n-butylphthalide. This study provides valuable data for the follow-up prodrug research of α-asaronol and n-butylphthalide.
Collapse
|
196
|
Neural stem cell therapy in conjunction with curcumin loaded in niosomal nanoparticles enhanced recovery from traumatic brain injury. Sci Rep 2022; 12:3572. [PMID: 35246564 PMCID: PMC8897489 DOI: 10.1038/s41598-022-07367-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/16/2022] [Indexed: 12/28/2022] Open
Abstract
Despite a great amount of effort, there is still a need for reliable treatments of traumatic brain injury (TBI). Recently, stem cell therapy has emerged as a new avenue to address neuronal regeneration after TBI. However, the environment of TBI lesions exerts negative effects on the stem cells efficacy. Therefore, to maximize the beneficial effects of stem cells in the course of TBI, we evaluated the effect of human neural stem/progenitor cells (hNS/PCs) and curcumin-loaded niosome nanoparticles (CM-NPs) on behavioral changes, brain edema, gliosis, and inflammatory responses in a rat model of TBI. After TBI, hNS/PCs were transplanted within the injury site and CM-NPs were orally administered for 10 days. Finally, the effect of combination therapy was compared to several control groups. Our results indicated a significant improvement of general locomotor activity in the hNS/PCs + CM-NPs treatment group compared to the control groups. We also observed a significant improvement in brain edema in the hNS/PCs + CM-NPs treatment group compared to the other groups. Furthermore, a significant decrease in astrogliosis was seen in the combined treatment group. Moreover, TLR4-, NF-κB-, and TNF-α- positive cells were significantly decreased in hNS/PCs + CM-NPs group compared to the control groups. Taken together, this study indicated that combination therapy of stem cells with CM-NPs can be an effective therapy for TBI.
Collapse
|
197
|
Wan T, Zhu W, Zhao Y, Zhang X, Ye R, Zuo M, Xu P, Huang Z, Zhang C, Xie Y, Liu X. Astrocytic phagocytosis contributes to demyelination after focal cortical ischemia in mice. Nat Commun 2022; 13:1134. [PMID: 35241660 PMCID: PMC8894352 DOI: 10.1038/s41467-022-28777-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/11/2022] [Indexed: 01/24/2023] Open
Abstract
Ischemic stroke can cause secondary myelin damage in the white matter distal to the primary injury site. The contribution of astrocytes during secondary demyelination and the underlying mechanisms are unclear. Here, using a mouse of distal middle cerebral artery occlusion, we show that lipocalin-2 (LCN2), enriched in reactive astrocytes, expression increases in nonischemic areas of the corpus callosum upon injury. LCN2-expressing astrocytes acquire a phagocytic phenotype and are able to uptake myelin. Myelin removal is impaired in Lcn2−/− astrocytes. Inducing re-expression of truncated LCN2(Δ2–20) in astrocytes restores phagocytosis and leads to progressive demyelination in Lcn2−/− mice. Co-immunoprecipitation experiments show that LCN2 binds to low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. Knockdown of Lrp1 reduces LCN2-induced myelin engulfment by astrocytes and reduces demyelination. Altogether, our findings suggest that LCN2/LRP1 regulates astrocyte-mediated myelin phagocytosis in a mouse model of ischemic stroke. Ischemic stroke can cause secondary demyelination. Whether phagocytic astrocytes can contribute to such demyelination is unclear. Here, the authors show that lipocalin-2 (LCN-2) expression increased in astrocytes upon injury. LCN-2 expressing astrocytes acquire a phagocytic phenotype and contribute to secondary demyelination in a mouse model of ischemic stroke.
Collapse
Affiliation(s)
- Ting Wan
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Wusheng Zhu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Ying Zhao
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Xiaohao Zhang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Ruidong Ye
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Meng Zuo
- Department of Neurology, Southwest Hospital and the First Affiliated Hospital, Army Medical University, Chongqing, 400000, China
| | - Pengfei Xu
- Stroke Center & Department of Neurology, The Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Zhenqian Huang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Chunni Zhang
- Department of Clinical Laboratory, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China.
| | - Yi Xie
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China.
| | - Xinfeng Liu
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210000, China. .,Stroke Center & Department of Neurology, The Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.
| |
Collapse
|
198
|
Guo X, Jin X, Han K, Kang S, Tian S, Lv X, Feng M, Zheng H, Zuo Y, Xu G, Hu M, Xu J, Lv P, Chang YZ. Iron promotes neurological function recovery in mice with ischemic stroke through endogenous repair mechanisms. Free Radic Biol Med 2022; 182:59-72. [PMID: 35202785 DOI: 10.1016/j.freeradbiomed.2022.02.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/17/2022]
Abstract
The endogenous repair mechanisms play an important role in the recovery of nerve function after stroke, such as gliosis, synaptic plasticity, remyelination and nerve regeneration. Iron is the most abundant trace metal element in the brain and plays a crucial role in the maintenance of normal cerebral function. It is an important coenzyme factor in the process of cell metabolism, DNA synthesis, purine catabolism and neurotransmitter synthesis and decomposition. However, it is unclear what role iron plays in the long-term recovery of neurological function after stroke. In this study, we first observed that changes in iron metabolism occurred during neurological function recovery in the mice with distal middle cerebral artery occlusion (dMCAO). Our data showed that plasticity changes due to endogenous repair mechanisms resulted in improvements in cerebral cortex function. These changes involved gliosis, synaptic function reconstruction, remyelination, and activation of neural stem cells. In order to examine the potential role of iron, we synthesized liposomal-encapsulated deferoxamine (DFO) nanoparticles to further explore the effect and the mechanism of iron on the recovery of neurological function in dMCAO mice. Our results showed that liposome-DFO decreased iron deposition and reversed plasticity changes in cerebral cortex function after stroke, which delayed neurological function recovery. This experiment shows that the increasing iron level promotes endogenous repair in ischemic stroke. Our finding reveals the change regularity of iron and emphasizes the beneficial role of iron in the recovery process of neurological function, which provides an important basis for the prevention and/or treatment of ischemia-reperfusion and recovery after stroke.
Collapse
Affiliation(s)
- Xin Guo
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Xiaofang Jin
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Kang Han
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Shaomeng Kang
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Siyu Tian
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Xin Lv
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Mudi Feng
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Huiwen Zheng
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Yong Zuo
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Guodong Xu
- Department of Neurology, Hebei General Hospital; Shijiazhuang 050051, Hebei, China
| | - Ming Hu
- Department of Neurology, Hebei General Hospital; Shijiazhuang 050051, Hebei, China
| | - Jing Xu
- Department of Neurology, Hebei General Hospital; Shijiazhuang 050051, Hebei, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China; Department of Neurology, Hebei General Hospital; Shijiazhuang 050051, Hebei, China.
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China.
| |
Collapse
|
199
|
Yang R, Shen YJ, Chen M, Zhao JY, Chen SH, Zhang W, Song JK, Li L, Du GH. Quercetin attenuates ischemia reperfusion injury by protecting the blood-brain barrier through Sirt1 in MCAO rats. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2022; 24:278-289. [PMID: 34292112 DOI: 10.1080/10286020.2021.1949302] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/25/2021] [Indexed: 05/25/2023]
Abstract
The purpose of the present study was to examine the protective action and mechanisms of quercetin on the blood-brain barrier (BBB) in rats subjected to transient middle cerebral artery occlusion (tMCAO) and reperfusion. Quercetin (10, 30, 50 mg/kg) was intraperitoneally administered at the onset of reperfusion. The results showed that quercetin significantly reduced cerebral infarct volume, neurological deficit, BBB permeability and ROS generation via Sirt1/Nrf2/HO-1 signaling pathway. Moreover, EX527, a selective inhibitor of Sirt1, reversed these neuroprotective effects. Our findings indicate that quercetin has neuroprotective effects against cerebral ischemia-reperfusion injury by protecting BBB through Sirt1 signaling pathway in MCAO rats.
Collapse
Affiliation(s)
- Ran Yang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yan-Jia Shen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Miao Chen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jia-Ying Zhao
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shu-Han Chen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wen Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jun-Ke Song
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Li Li
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Guan-Hua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
200
|
ECM1 modified HF-MSCs targeting HSC attenuate liver cirrhosis by inhibiting the TGF-β/Smad signaling pathway. Cell Death Dis 2022; 8:51. [PMID: 35136027 PMCID: PMC8827057 DOI: 10.1038/s41420-022-00846-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022]
Abstract
Hair follicle-derived mesenchymal stem cells (HF-MSCs) show considerable therapeutic potential for liver cirrhosis (LC). To improve the effectiveness of naïve HF-MSC treatments on LC, we used bioinformatic tools to identify an exogenous gene targeting HSCs among the differentially expressed genes (DEGs) in LC to modify HF-MSCs. Extracellular matrix protein 1 (ECM1) was identified as a DEG that was significantly downregulated in the cirrhotic liver. Then, ECM1-overexpressing HF-MSCs (ECM1-HF-MSCs) were transplanted into mice with LC to explore the effectiveness and correlated mechanism of gene-overexpressing HF-MSCs on LC. The results showed that ECM1-HF-MSCs significantly improved liver function and liver pathological injury in LC after cell therapy relative to the other treatment groups. Moreover, we found that ECM1-HF-MSCs homed to the injured liver and expressed the hepatocyte-specific surface markers ALB, CK18, and AFP. In addition, hepatic stellate cell (HSC) activation was significantly inhibited in the cell treatment groups in vivo and in vitro, especially in the ECM1-HF-MSC group. Additionally, TGF-β/Smad signal inhibition was the most significant in the ECM1-HF-MSC group in vivo and in vitro. The findings indicate that the genetic modification of HF-MSCs with bioinformatic tools may provide a broad perspective for precision treatment of LC.
Collapse
|