1
|
Wong Zhang DE, Gibson Hughes TA, Figueiredo Galvao HB, Lo C, Dinh QN, Zhang SR, Kim HA, Selvaraji S, Clarkson AN, Arumugam TV, Drummond G, Sobey CG, De Silva TM. Post-stroke cognitive impairment and brain hemorrhage are augmented in hypertensive mice. J Cereb Blood Flow Metab 2024:271678X241262127. [PMID: 38886874 DOI: 10.1177/0271678x241262127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Hypertension is a major risk factor for both stroke and cognitive impairment, but it is unclear whether it may specifically affect post-stroke cognitive impairment. We assessed the effect of hypertension and/or stroke on brain injury, cognitive outcome, and the brain transcriptomic profile. C57BL/6J mice (n = 117; 3-5 mo.) received s.c. infusion of either saline or angiotensin II followed by sham surgery or photothrombotic stroke targeting the prefrontal cortex seven days later. Cognitive function was assessed with the Barnes maze and RNA sequencing was used to quantify transcriptomic changes in the brain. Angiotensin II treatment produced spontaneous hemorrhaging after stroke. In the Barnes maze, hypertensive mice that received stroke surgery had an increased escape latency compared to other groups (day 3: hypertensive + stroke = 166.6 ± 6.0 s vs. hypertensive + sham = 122.8 ± 13.8 s vs. normotensive + stroke = 139.9 ± 10.1 s vs. normotensive + sham = 101.9 ± 16.7 s), consistent with impaired cognition. RNA sequencing revealed >1500 differentially expressed genes related to neuroinflammation in hypertensive + stroke vs. normotensive + stroke, which included genes associated with apoptosis, microRNAs, autophagy, anti-cognitive biomarkers and Wnt signaling. Overall, we show that the combination of hypertension and stroke resulted in greater learning impairment and brain injury.
Collapse
Affiliation(s)
- David E Wong Zhang
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Tayla A Gibson Hughes
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Hericka B Figueiredo Galvao
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Cecilia Lo
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Shenpeng R Zhang
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Hyun Ah Kim
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Sharmalee Selvaraji
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Andrew N Clarkson
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Thiruma V Arumugam
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Grant Drummond
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - T Michael De Silva
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| |
Collapse
|
2
|
Chung K, Ullah I, Yi Y, Kang E, Yun G, Heo S, Kim M, Chung SE, Park S, Lim J, Lee M, Rhim T, Lee SK. Intranasal Delivery of Anti-Apoptotic siRNA Complexed with Fas-Signaling Blocking Peptides Attenuates Cellular Apoptosis in Brain Ischemia. Pharmaceutics 2024; 16:290. [PMID: 38399343 PMCID: PMC10892455 DOI: 10.3390/pharmaceutics16020290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Ischemic stroke-induced neuronal cell death leads to the permanent impairment of brain function. The Fas-mediating extrinsic apoptosis pathway and the cytochrome c-mediating intrinsic apoptosis pathway are two major molecular mechanisms contributing to neuronal injury in ischemic stroke. In this study, we employed a Fas-blocking peptide (FBP) coupled with a positively charged nona-arginine peptide (9R) to form a complex with negatively charged siRNA targeting Bax (FBP9R/siBax). This complex is specifically designed to deliver siRNA to Fas-expressing ischemic brain cells. This complex enables the targeted inhibition of Fas-mediating extrinsic apoptosis pathways and cytochrome c-mediating intrinsic apoptosis pathways. Specifically, the FBP targets the Fas/Fas ligand signaling, while siBax targets Bax involved in mitochondria disruption in the intrinsic pathway. The FBP9R carrier system enables the delivery of functional siRNA to hypoxic cells expressing the Fas receptor on their surface-a finding validated through qPCR and confocal microscopy analyses. Through intranasal (IN) administration of FBP9R/siCy5 to middle cerebral artery occlusion (MCAO) ischemic rat models, brain imaging revealed the complex specifically localized to the Fas-expressing infarcted region but did not localize in the non-infarcted region of the brain. A single IN administration of FBP9R/siBax demonstrated a significant reduction in neuronal cell death by effectively inhibiting Fas signaling and preventing the release of cytochrome c. The targeted delivery of FBP9R/siBax represents a promising alternative strategy for the treatment of brain ischemia.
Collapse
Affiliation(s)
- Kunho Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Irfan Ullah
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
- Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Yujong Yi
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Eunhwa Kang
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Gyeongju Yun
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Seoyoun Heo
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Minkyung Kim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Seong-Eun Chung
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Seongjun Park
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Jaeyeoung Lim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Minhyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Taiyoun Rhim
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| | - Sang-Kyung Lee
- Department of Bioengineering and Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea; (K.C.); (Y.Y.); (S.H.)
| |
Collapse
|
3
|
Lotfi A, Abbasi M, Karami N, Arghavanfar H, Kazeminasab F, Rosenkranz SK. Effects of treadmill training on myelin proteomic markers and cerebellum morphology in a rat model of cuprizone-induced toxic demyelination. J Neuroimmunol 2024; 387:578286. [PMID: 38215583 DOI: 10.1016/j.jneuroim.2024.578286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 01/14/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system (CNS). If demyelination is persistent, it will result in irreversible axonal injury and loss. The purpose of the current study was to investigate the effects of treadmill training on myelin proteomic markers and cerebellum morphology in a rat model of cuprizone-induced toxic demyelination. METHODS Thirty male rats were randomly assigned to five groups (n = 6 per group), consisting of a healthy control group (Control), a cuprizone (CPZ) group, and three exercise training groups: exercise training before and during the CPZ administration (EX-CPZ-EX), exercise training before the CPZ administration (EX-CPZ), and exercise training during the CPZ administration (CPZ-EX). A rat model of CPZ-induced toxic demyelination consisted of feeding the rats cuprizone pellets (0.2%) for 6 weeks. All exercise groups performed a treadmill training protocol 5 days/week for 6 weeks. Levels of Myelin proteolipid protein (PLP), Myelin oligodendrocyte glycoprotein (MOG), axonal injury in the cerebellar tissue, and volume, weight, and length of the cerebellum were determined. RESULTS Results indicated a significant decrease in PLP and MOG in the CPZ groups compared to the Control group (****p < 0.0001). There was a significant increase in PLP and MOG and a significant decrease in axonal injury in the EX-CPZ-EX group as compared to other CPZ groups (****p < 0.0001), and CPZ-MS and CPZ-EX were not significantly different from one another. However, there were no significant differences between the groups for the volume, weight, or length of the cerebellum. CONCLUSION Treadmill training improved myelin sheath structural proteins and axonal injury in cerebellar tissue in a rat model of CPZ-induced toxic demyelination.
Collapse
Affiliation(s)
- Alireza Lotfi
- Department of Exercise Physiology, Ilam Branch, Islamic Azad University, Ilam, Iran
| | - Maryam Abbasi
- Department of Exercise Physiology, Ilam Branch, Islamic Azad University, Ilam, Iran.
| | - Nasrin Karami
- Department of Exercise Physiology, Ilam Branch, Islamic Azad University, Ilam, Iran
| | - Hadis Arghavanfar
- Department of Exercise Physiology, Ilam Branch, Islamic Azad University, Ilam, Iran
| | - Fatemeh Kazeminasab
- Department of Physical Education and Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran
| | - Sara K Rosenkranz
- Department of Kinesiology and Nutrition Sciences, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
4
|
Li L, Wang L, Zhang L. Therapeutic Potential of Natural Compounds from Herbs and Nutraceuticals in Alleviating Neurological Disorders: Targeting the Wnt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2411-2433. [PMID: 38284360 DOI: 10.1021/acs.jafc.3c07536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
As an important signaling pathway in multicellular eukaryotes, the Wnt signaling pathway participates in a variety of physiological processes. Recent studies have confirmed that the Wnt signaling pathway plays an important role in neurological disorders such as stroke, Alzheimer's disease, and Parkinson's disease. The regulation of Wnt signaling by natural compounds in herbal medicines and nutraceuticals has emerged as a potential strategy for the development of new drugs for neurological disorders. Purpose: The aim of this review is to evaluate the latest research results on the efficacy of natural compounds derived from herbs and nutraceuticals in the prevention and treatment of neurological disorders by regulating the Wnt pathway in vivo and in vitro. A manual and electronic search was performed for English articles available from PubMed, Web of Science, and ScienceDirect from the January 2010 to February 2023. Keywords used for the search engines were "natural products,″ "plant derived products,″ "Wnt+ clinical trials,″ and "Wnt+,″ and/or paired with "natural products″/″plant derived products", and "neurological disorders." A total of 22 articles were enrolled in this review, and a variety of natural compounds from herbal medicine and nutritional foods have been shown to exert therapeutic effects on neurological disorders through the Wnt pathway, including curcumin, resveratrol, and querctrin, etc. These natural products possess antioxidant, anti-inflammatory, and angiogenic properties, confer neurovascular unit and blood-brain barrier integrity protection, and affect neural stem cell differentiation, synaptic formation, and neurogenesis, to play a therapeutic role in neurological disorders. In various in vivo and in vitro studies and clinical trials, these natural compounds have been shown to be safe and tolerable with few adverse effects. Natural compounds may serve a therapeutic role in neurological disorders by regulating the Wnt pathway. This summary of the research progress of natural compounds targeting the Wnt pathway may provide new insights for the treatment of neurological disorders and potential targets for the development of new drugs.
Collapse
Affiliation(s)
- Lei Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning PR China
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning PR China
| | - Lijuan Zhang
- Departments of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang 110000, Liaoning PR China
| |
Collapse
|
5
|
Zhang M, Liu Q, Meng H, Duan H, Liu X, Wu J, Gao F, Wang S, Tan R, Yuan J. Ischemia-reperfusion injury: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:12. [PMID: 38185705 PMCID: PMC10772178 DOI: 10.1038/s41392-023-01688-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemia-reperfusion (I/R) injury paradoxically occurs during reperfusion following ischemia, exacerbating the initial tissue damage. The limited understanding of the intricate mechanisms underlying I/R injury hinders the development of effective therapeutic interventions. The Wnt signaling pathway exhibits extensive crosstalk with various other pathways, forming a network system of signaling pathways involved in I/R injury. This review article elucidates the underlying mechanisms involved in Wnt signaling, as well as the complex interplay between Wnt and other pathways, including Notch, phosphatidylinositol 3-kinase/protein kinase B, transforming growth factor-β, nuclear factor kappa, bone morphogenetic protein, N-methyl-D-aspartic acid receptor-Ca2+-Activin A, Hippo-Yes-associated protein, toll-like receptor 4/toll-interleukine-1 receptor domain-containing adapter-inducing interferon-β, and hepatocyte growth factor/mesenchymal-epithelial transition factor. In particular, we delve into their respective contributions to key pathological processes, including apoptosis, the inflammatory response, oxidative stress, extracellular matrix remodeling, angiogenesis, cell hypertrophy, fibrosis, ferroptosis, neurogenesis, and blood-brain barrier damage during I/R injury. Our comprehensive analysis of the mechanisms involved in Wnt signaling during I/R reveals that activation of the canonical Wnt pathway promotes organ recovery, while activation of the non-canonical Wnt pathways exacerbates injury. Moreover, we explore novel therapeutic approaches based on these mechanistic findings, incorporating evidence from animal experiments, current standards, and clinical trials. The objective of this review is to provide deeper insights into the roles of Wnt and its crosstalk signaling pathways in I/R-mediated processes and organ dysfunction, to facilitate the development of innovative therapeutic agents for I/R injury.
Collapse
Affiliation(s)
- Meng Zhang
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
| | - Qian Liu
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hui Meng
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hongxia Duan
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Xin Liu
- Second Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fei Gao
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Rubin Tan
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China.
| |
Collapse
|
6
|
Chen Y, Ren P, He X, Yan F, Gu R, Bai J, Zhang X. Olfactory bulb neurogenesis depending on signaling in the subventricular zone. Cereb Cortex 2023; 33:11102-11111. [PMID: 37746807 DOI: 10.1093/cercor/bhad349] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
Olfaction is a crucial sense that is essential for the well-being and survival of individuals. Olfactory bulb (OB) is the first olfactory relay station, and its function depends on newly generated neurons from the subventricular zone (SVZ). These newly born neurons constantly migrate through the rostral migratory stream to integrate into existing neural networks within the OB, thereby contributing to olfactory information processing. However, the mechanisms underlying the contribution of SVZ adult neurogenesis to OB neurogenesis remain largely elusive. Adult neurogenesis is a finely regulated multistep process involving the proliferation of adult neural stem cells (aNSCs) and neural precursor cells, as well as the migration and differentiation of neuroblasts, and integration of newly generated neurons into preexisting neuronal circuitries. Recently, extensive studies have explored the mechanism of SVZ and OB neurogenesis. This review focused on elucidating various molecules and signaling pathways associated with OB neurogenesis dependent on the SVZ function. A better understanding of the mechanisms underlying the OB neurogenesis on the adult brain is an attractive prospect to induce aNSCs in SVZ to generate new neurons to ameliorate olfactory dysfunction that is involved in various diseases. It will also contribute to developing new strategies for the human aNSCs-based therapies.
Collapse
Affiliation(s)
- Yali Chen
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Peng Ren
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Xiongjie He
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Fang Yan
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Rou Gu
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Jie Bai
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Xianwen Zhang
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
7
|
Chen J, Chen Z, Yu D, Yan Y, Hao X, Zhang M, Zhu T. Neuroprotective Effect of Hydrogen Sulfide Subchronic Treatment Against TBI-Induced Ferroptosis and Cognitive Deficits Mediated Through Wnt Signaling Pathway. Cell Mol Neurobiol 2023; 43:4117-4140. [PMID: 37624470 PMCID: PMC10661805 DOI: 10.1007/s10571-023-01399-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023]
Abstract
Emerging evidence shows that targeting ferroptosis may be a potential therapeutic strategy for treating traumatic brain injury (TBI). Hydrogen sulfide (H2S) has been proven to play a neuroprotective role in TBI, but little is known about the effects of H2S on TBI-induced ferroptosis. In addition, it is reported that the Wnt signaling pathway can also actively regulate ferroptosis. However, whether H2S inhibits ferroptosis via the Wnt signaling pathway after TBI remains unclear. In this study, we first found that in addition to alleviating neuronal damage and cognitive impairments, H2S remarkably attenuated abnormal iron accumulation, decreased lipid peroxidation, and improved the expression of glutathione peroxidase 4, demonstrating the potent anti-ferroptosis action of H2S after TBI. Moreover, Wnt3a or liproxstatin-1 treatment obtained similar results, suggesting that activation of the Wnt signaling pathway can render the cells less susceptible to ferroptosis post-TBI. More importantly, XAV939, an inhibitor of the Wnt signaling pathway, almost inversed ferroptosis inactivation and reduction of neuronal loss caused by H2S treatment, substantiating the involvement of the Wnt signaling pathway in anti-ferroptosis effects of H2S. In conclusion, the Wnt signaling pathway might be the critical mechanism in realizing the anti-ferroptosis effects of H2S against TBI. TBI induces ferroptosis-related changes characterized by iron overload, impaired antioxidant system, and lipid peroxidation at the chronic phase after TBI. However, NaHS subchronic treatment reduces the susceptibility to TBI-induced ferroptosis, at least partly by activating the Wnt signaling pathway.
Collapse
Affiliation(s)
- Jie Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Pep-Tides, The Affiliated Xi'an International Medical Center Hospital, Northwest University, No.777 Xitai Road, Xi'an, 710100, Shaanxi, China
| | - Zhennan Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Dongyu Yu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yufei Yan
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xiuli Hao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Mingxia Zhang
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Pep-Tides, The Affiliated Xi'an International Medical Center Hospital, Northwest University, No.777 Xitai Road, Xi'an, 710100, Shaanxi, China
| | - Tong Zhu
- Clinical Experimental Center, Xi'an Engineering Technology Research Center for Cardiovascular Active Pep-Tides, The Affiliated Xi'an International Medical Center Hospital, Northwest University, No.777 Xitai Road, Xi'an, 710100, Shaanxi, China.
| |
Collapse
|
8
|
Kang X, Wang D, Zhang L, Huang T, Liu S, Feng X, Guo Y, Zhang Z, Wang Z, Ren H, Yuan G. Exendin-4 ameliorates tau hyperphosphorylation and cognitive impairment in type 2 diabetes through acting on Wnt/β-catenin/NeuroD1 pathway. Mol Med 2023; 29:118. [PMID: 37667187 PMCID: PMC10478475 DOI: 10.1186/s10020-023-00718-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is an independent risk factor for Alzheimer's disease (AD). Exendin-4 (Ex-4), a widely used glucagon-like peptide-1 receptor agonist drug in the treatment of T2D, has been demonstrated the therapeutic effects on diabetic encephalopathy (DE). Especially, the Ex-4 ameliorates the tau hyperphosphorylation and cognitive impairment in DE. And these crucial alterations are also important bridge between T2D and AD. However, its unique mechanism is unclear. METHODS The db/db mice, high-fat-diet (HFD) / streptozotocin (STZ)-induced diabetic (HF-diabetic) mice, and high-glucose-damaged (HGD) HT-22 hippocampal cells were enrolled to examine the effects of Ex-4 on AD-like changes in T2D. The Novel object recognition test (NORT) and Morris water maze test (MWMT) were conducted to evaluate the cognitive impairment. The Dickkopf-1 (DKK1) was employed to weaken the activation of the Wnt/β-catenin pathway to explore the mechanism of Ex-4 in protecting the brain functions. The JASPAR was based to predict the interaction between NeuroD1 and the promoter region of Ins2. Moreover, the chromatin immunoprecipitation coupled with quantitative polymerase chain reaction (ChIP-qPCR) and luciferase reporter assays were performed. RESULTS Ex-4 alleviated the tau hyperphosphorylation, increased the brain-derived insulin, and improved the PI3K/AKT/GSK3-β signalling in db/db mice, HF-diabetic mice, and HGD HT-22 hippocampal neuronal cells. The NORT and MWMT indicated that Ex-4 alleviated the learning and memory deficits in HF-diabetic mice. The inhibitor Dickkopf-1 (DKK1) of the Wnt/β-catenin pathway significantly blocked the protective effects of Ex-4. Regarding further molecular mechanisms, NeuroD1 was affected by Ex-4 in vivo and in vitro, and the knockdown or overexpression of NeuroD1 suggested its crucial role in promoting the brain insulin by Ex-4. Meanwhile, the ChIP‒qPCR and luciferase reporter assays confirmed the combination between NeuroD1 and the promoter region of the insulin-encoding gene Ins2. And this interaction could be promoted by Ex-4. CONCLUSIONS Our study proposes that Ex-4 alleviates tau hyperphosphorylation and cognitive dysfunction by increasing Ins2-derived brain insulin through the Wnt/β-catenin/NeuroD1 signaling in T2D. And its also show new lights on part of the progress and mechanism on treatment targets for the DE in T2D.
Collapse
Affiliation(s)
- Xiaonan Kang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Wang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lu Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Teng Huang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Siyue Liu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaohui Feng
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaoyao Guo
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ziyin Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhongjing Wang
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Huihui Ren
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Branch of National Clinical Research Center for Metabolic Disease, Hubei, People's Republic of China.
| | - Gang Yuan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Branch of National Clinical Research Center for Metabolic Disease, Hubei, People's Republic of China.
| |
Collapse
|
9
|
Geng C, Liu S, Wang J, Wang S, Zhang W, Rong H, Cao Y, Wang S, Li Z, Zhang Y. Targeting the cochlin/SFRP1/CaMKII axis in the ocular posterior pole prevents the progression of nonpathologic myopia. Commun Biol 2023; 6:884. [PMID: 37644183 PMCID: PMC10465513 DOI: 10.1038/s42003-023-05267-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Myopia is a major public health issue. However, interventional modalities for nonpathologic myopia are limited due to its complicated pathogenesis and the lack of precise targets. Here, we show that in guinea pig form-deprived myopia (FDM) and lens-induced myopia (LIM) models, the early initiation, phenotypic correlation, and stable maintenance of cochlin protein upregulation at the interface between retinal photoreceptors and retinal pigment epithelium (RPE) is identified by a proteomic analysis of ocular posterior pole tissues. Then, a microarray analysis reveals that cochlin upregulates the expression of the secreted frizzled-related protein 1 (SFRP1) gene in human RPE cells. Moreover, SFRP-1 elevates the intracellular Ca2+ concentration and activates Ca2+/calmodulin-dependent protein kinase II (CaMKII) signaling in a simian choroidal vascular endothelial cell line, and elicits vascular endothelial cell dysfunction. Furthermore, genetic knockdown of the cochlin gene and pharmacological blockade of SFRP1 abrogates the reduced choroidal blood perfusion and prevents myopia progression in the FDM model. Collectively, this study identifies a novel signaling axis that may involve cochlin in the retina, SFRP1 in the RPE, and CaMKII in choroidal vascular endothelial cells and contribute to the pathogenesis of nonpathologic myopia, implicating the potential of cochlin and SFRP1 as myopia interventional targets.
Collapse
Affiliation(s)
- Chao Geng
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China
| | - Siyi Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China
| | - Jindan Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China
| | - Sennan Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China
| | - Weiran Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China
| | - Hua Rong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China
| | - Yunshan Cao
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou University, 730000, Lanzhou, Gansu Province, China
| | - Shuqing Wang
- School of Pharmacy, Tianjin Medical University, 300070, Tianjin, China
| | - Zhiqing Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China
| | - Yan Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 300384, Tianjin, China.
| |
Collapse
|
10
|
Lei R, Wang S, Liu A, Cheng J, Zhang Z, Ren J, Yao X, Kong X, Ma W, Che F, Chen J, Wan Q. Bilateral transcranial direct-current stimulation promotes migration of subventricular zone-derived neuroblasts toward ischemic brain. FASEB Bioadv 2023; 5:277-286. [PMID: 37415929 PMCID: PMC10320846 DOI: 10.1096/fba.2023-00017] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 07/08/2023] Open
Abstract
Ischemic insult stimulates proliferation of neural stem cells (NSCs) in the subventricular zone (SVZ) after stroke. However, only a fraction of NSC-derived neuroblasts from SVZ migrate toward poststroke brain region. We have previously reported that direct-current stimulation guides NSC migration toward the cathode in vitro. Accordingly, we set up a new method of transcranial direct-current stimulation (tDCS), in which the cathodal electrode is placed on the ischemic hemisphere and anodal electrode on the contralateral hemisphere of rats subjected to ischemia-reperfusion injury. We show that the application of this bilateral tDCS (BtDCS) promotes the migration of NSC-derived neuroblasts from SVZ toward the cathode direction into poststroke striatum. Reversing the position of the electrodes blocks the effect of BtDCS on the migration of neuroblasts from SVZ. BtDCS protects against neuronal death and improves the functional recovery of stroke animals. Thus, the migration of NSC-derived neuroblasts from SVZ toward poststroke brain region contributes to the effect of BtDCS against ischemia-induced neuronal death, supporting a potential development of noninvasive BtDCS as an endogenous neurogenesis-based stroke therapy.
Collapse
Affiliation(s)
- Ruixue Lei
- Department of Pathology, Anyang Tumour HospitalThe Affiliated Anyang Tumor Hospital of Henan University of Science and TechnologyAnyangHenanChina
- Department of Physiology, School of MedicineWuhan UniversityWuhanChina
| | - Shu Wang
- Department of Physiology, School of MedicineWuhan UniversityWuhanChina
| | - Anchun Liu
- Department of Physiology, School of MedicineWuhan UniversityWuhanChina
| | - Jing Cheng
- Department of Physiology, School of MedicineWuhan UniversityWuhanChina
| | - Zhifeng Zhang
- Department of Physiology, School of MedicineWuhan UniversityWuhanChina
| | - Jinyang Ren
- Institute of Neuroregeneration & Neurorehabilitation, Department of NeurosurgeryQingdao UniversityQingdaoChina
| | - Xujin Yao
- Institute of Neuroregeneration & Neurorehabilitation, Department of NeurosurgeryQingdao UniversityQingdaoChina
| | - Xiangyi Kong
- Institute of Neuroregeneration & Neurorehabilitation, Department of NeurosurgeryQingdao UniversityQingdaoChina
| | - Wenlong Ma
- Institute of Neuroregeneration & Neurorehabilitation, Department of NeurosurgeryQingdao UniversityQingdaoChina
| | - Fengyuan Che
- Central Laboratory, Department of NeurologyLinyi People's Hospital, Qingdao UniversityLinyiShandongChina
| | - Juan Chen
- Department of Neurology, the Central Hospital of Wuhantongji medical collof Huazhong University of Science & TechnologyWuhanChina
| | - Qi Wan
- Department of Physiology, School of MedicineWuhan UniversityWuhanChina
- Institute of Neuroregeneration & Neurorehabilitation, Department of NeurosurgeryQingdao UniversityQingdaoChina
- Qingdao Gui‐Hong Intelligent Medical Technology Co. LtdQingdaoChina
| |
Collapse
|
11
|
Li L, Li X, Han R, Wu M, Ma Y, Chen Y, Zhang H, Li Y. Therapeutic Potential of Chinese Medicine for Endogenous Neurogenesis: A Promising Candidate for Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:ph16050706. [PMID: 37242489 DOI: 10.3390/ph16050706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Strokes are a leading cause of morbidity and mortality in adults worldwide. Extensive preclinical studies have shown that neural-stem-cell-based treatments have great therapeutic potential for stroke. Several studies have confirmed that the effective components of traditional Chinese medicine can protect and maintain the survival, proliferation, and differentiation of endogenous neural stem cells through different targets and mechanisms. Therefore, the use of Chinese medicines to activate and promote endogenous nerve regeneration and repair is a potential treatment option for stroke patients. Here, we summarize the current knowledge regarding neural stem cell strategies for ischemic strokes and the potential effects of these Chinese medicines on neuronal regeneration.
Collapse
Affiliation(s)
- Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Meirong Wu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yaolei Ma
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuzhao Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
12
|
Guo T, Chen M, Liu J, Wei Z, Yuan J, Wu W, Wu Z, Lai Y, Zhao Z, Chen H, Liu N. Neuropilin-1 promotes mitochondrial structural repair and functional recovery in rats with cerebral ischemia. J Transl Med 2023; 21:297. [PMID: 37138283 PMCID: PMC10155168 DOI: 10.1186/s12967-023-04125-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/10/2023] [Indexed: 05/05/2023] Open
Abstract
OBJECTIVES Available literature documents that ischemic stroke can disrupt the morphology and function of mitochondria and that the latter in other disease models can be preserved by neuropilin-1 (NRP-1) via oxidative stress suppression. However, whether NRP-1 can repair mitochondrial structure and promote functional recovery after cerebral ischemia is still unknown. This study tackled this very issue and explored the underlying mechanism. METHODS Adeno-associated viral (AAV)-NRP-1 was stereotaxically inoculated into the cortex and ipsilateral striatum posterior of adult male Sprague-Dawley (SD) rats before a 90-min transient middle cerebral artery occlusion (tMCAO) and subsequent reperfusion. Lentivirus (LV)-NRP-1 was transfected into rat primary cortical neuronal cultures before a 2-h oxygen-glucose deprivation and reoxygenation (OGD/R) injury to neurons. The expression and function of NRP-1 and its specific protective mechanism were investigated by Western Blot, immunofluorescence staining, flow cytometry, magnetic resonance imaging, transmission electron microscopy, etc. The binding was detected by molecular docking and molecular dynamics simulation. RESULTS Both in vitro and in vivo models of cerebral ischemia/reperfusion (I/R) injury presented a sharp increase in NRP-1 expression. The expression of AAV-NRP-1 markedly ameliorated the cerebral I/R-induced damage to the motor function and restored the mitochondrial morphology. The expression of LV-NRP-1 alleviated mitochondrial oxidative stress and bioenergetic deficits. AAV-NRP-1 and LV-NRP-1 treatments increased the wingless integration (Wnt)-associated signals and β-catenin nuclear localization. The protective effects of NRP-1 were reversed by the administration of XAV-939. CONCLUSIONS NRP-1 can produce neuroprotective effects against I/R injury to the brain by activating the Wnt/β-catenin signaling pathway and promoting mitochondrial structural repair and functional recovery, which may serve as a promising candidate target in treating ischemic stroke.
Collapse
Affiliation(s)
- Ting Guo
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Ji Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zengyu Wei
- Emergency Department, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinjin Yuan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Wenwen Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zhiyun Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Yongxing Lai
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zijun Zhao
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
13
|
Huang L, Liu M, Jiang W, Ding H, Han Y, Wen M, Li Y, Liu X, Zeng H. Bradykinin/bradykinin 1 receptor promotes brain microvascular endothelial cell permeability and proinflammatory cytokine release by downregulating Wnt3a. J Biochem Mol Toxicol 2022; 36:e23213. [PMID: 36111657 PMCID: PMC10078380 DOI: 10.1002/jbt.23213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/29/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022]
Abstract
Stroke is a life-threatening disease with limited therapeutic options. Damage to the blood-brain barrier (BBB) is the key pathological feature of ischemic stroke. This study explored the role of the bradykinin (BK)/bradykinin 1 receptor (B1R) and its mechanism of action in the BBB. Human brain microvascular endothelial cells (BMECs) were used to test for cellular responses to BK by using the Cell Counting Kit-8 assay, 5-ethynyl-2'-deoxyuridine staining, enzyme-linked immunosorbent assay, flow cytometry, immunofluorescence, cellular permeability assays, and western blotting to evaluate cell viability, cytokine production, and reactive oxygen species (ROS) levels in vitro. A BBB induced by middle cerebral artery occlusion was used to evaluate BBB injuries, and the role played by BK/B1R in ischemic/reperfusion (I/R) was explored in a rat model. Results showed that BK reduced the viability of BMECs and increased the levels of proinflammatory cytokines (interleukin 6 [IL-6], IL-18, and monocyte chemoattractant protein-1) and ROS. Additionally, cellular permeability was increased by BK treatment, and the expression of tight junction proteins (claudin-5 and occludin) was decreased. Interestingly, Wnt3a expression was inhibited by BK and exogenous Wnt3a restored the effects of BK on BMECs. In an in vivo I/R rat model, knockdown of B1R significantly decreased infarct volume and inflammation in I/R rats. Our results suggest that BK might be a key inducer of BBB injury and B1R knockdown might provide a beneficial effect by upregulating Wnt3a.
Collapse
Affiliation(s)
- Linqiang Huang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Mengting Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, The Second School of Clinical Medicine Southern Medical University Guangzhou China
| | - Wenqiang Jiang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Hongguang Ding
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Yongli Han
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Miaoyun Wen
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Ya Li
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, School of Medicine South China University of Technology Guangzhou China
| | - Xiaoyu Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, The Second School of Clinical Medicine Southern Medical University Guangzhou China
| | - Hongke Zeng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| |
Collapse
|
14
|
Overexpression of Brain- and Glial Cell Line-Derived Neurotrophic Factors Is Neuroprotective in an Animal Model of Acute Hypobaric Hypoxia. Int J Mol Sci 2022; 23:ijms23179733. [PMID: 36077134 PMCID: PMC9456324 DOI: 10.3390/ijms23179733] [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: 07/10/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Currently, the role of the neurotrophic factors BDNF and GDNF in maintaining the brain’s resistance to the damaging effects of hypoxia and functional recovery of neural networks after exposure to damaging factors are actively studied. The assessment of the effect of an increase in the level of these neurotrophic factors in brain tissues using genetic engineering methods on the resistance of laboratory animals to hypoxia may pave the way for the future clinical use of neurotrophic factors BDNF and GDNF in the treatment of hypoxic damage. This study aimed to evaluate the antihypoxic and neuroprotective properties of BDNF and GDNF expression level increase using adeno-associated viral vectors in modeling hypoxia in vivo. To achieve overexpression of neurotrophic factors in the central nervous system’s cells, viral constructs were injected into the brain ventricles of newborn male C57Bl6 (P0) mice. Acute hypobaric hypoxia was modeled on the 30th day after the injection of viral vectors. Survival, cognitive, and mnestic functions in the late post-hypoxic period were tested. Evaluation of growth and weight characteristics and the neurological status of animals showed that the overexpression of neurotrophic factors does not affect the development of mice. It was found that the use of adeno-associated viral vectors increased the survival rate of male mice under hypoxic conditions. The present study indicates that the neurotrophic factors’ overexpression, induced by the specially developed viral constructs carrying the BDNF and GDNF genes, is a prospective neuroprotection method, increasing the survival rate of animals after hypoxic injury.
Collapse
|
15
|
Zhao X, Qiao D, Guan D, Wang K, Cui Y. Chrysophanol Ameliorates Hemin-Induced Oxidative Stress and Endoplasmic Reticulum Stress by Regulating MicroRNA-320-5p/Wnt3a Pathway in HT22 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9399658. [PMID: 35936221 PMCID: PMC9355772 DOI: 10.1155/2022/9399658] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022]
Abstract
Oxidative stress, endoplasmic reticulum (ER) stress, and neuronal cell apoptosis have been considered as the main pathogenesis factors of brain injury after intracerebral hemorrhage (ICH). Chrysophanol (CHR) has been proved to have neuroprotective effects, but the role and underlying mechanisms of CHR in ICH remain unclear. HT22 cells were dealt with hemin to mimic an in vitro ICH model and then subjected to treatment with or without CHR. The cell viability, apoptosis, ER stress, and oxidative stress were evaluated by conducting the cell counting kit-8 (CCK-8), TdT-mediated dUTP nick end labeling (TUNEL) staining assays, western blot, and corresponding kit, respectively. Further, microRNA-sequencing, bioinformatic analysis, dual-luciferase reporter method, and rescue experiments were conducted to explore the molecular mechanisms of CHR alleviating hemin-induced ER in HT22 cell. Our data revealed that CHR increased cells viability, antiapoptosis, anti-ER stress, and antioxidative stress under conditions of hemin-induced HT22 cell injury. Mechanically, it was observed that Wnt3a was competitively sponged by miR-320-5p, and CHR activated β-catenin pathway by regulating miR-320-5p/Wnt3a molecular axis. Finally, results from the rescue experiment suggested that CHR inhibited hemin-induced cells apoptosis, ER stress, and oxidative stress through regulating the miR-320-5p/Wnt3a axis in HT22 cells. In conclusion, CHR prevented hemin-induced apoptosis, ER stress, and oxidative stress via inhibiting the miR-320-5p/Wnt3a/β-catenin pathway in HT22 cells. Our results certified that CHR could be served as a promising treatment for brain damage following ICH.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Pharmacy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Dongge Qiao
- Nursing Department, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Dongsheng Guan
- Department of Encephalopathy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Kun Wang
- Department of Pharmacy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Yinglin Cui
- Department of Encephalopathy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| |
Collapse
|
16
|
Han R, Zhang P, Li H, Chen Y, Hao Y, Guo Q, Zhang A, Li D. Differential Expression and Correlation Analysis of Global Transcriptome for Hemorrhagic Transformation After Acute Ischemic Stroke. Front Neurosci 2022; 16:889689. [PMID: 35757529 PMCID: PMC9214200 DOI: 10.3389/fnins.2022.889689] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
In order to explore the epigenetic characteristics of hemorrhagic transformation (HT) after acute ischemic stroke, we used transcriptome sequencing technology to analyze the global transcriptome expression profile of patients with and without HT after acute ischemic stroke and to study the differential expression of messenger RNA (mRNA), long noncoding RNA (lncRNA), circular RNA (circRNA) and mircoRNA (miRNA) between the two groups. To further explore the role of differentially expressed genes in HT, we annotated the function of differentially expressed genes by using gene ontology (GO) and pathway analysis on the results and showed that there were 1,051 differential expressions of lncRNAs, 2,575 differential expressions of mRNAs, 447 differential expressions of circRNAs and 47 miRNAs in patients with HT compared with non-HT patients. Pathway analysis showed that ubiquitin-mediated proteolysis, MAPK signal pathway, axon guidance, HIF-1 signal pathway, NOD-like receptor signal pathway, beta-alanine metabolism, Wnt signal pathway, sphingolipid signal pathway, neuroactive ligand-receptor interaction, and intestinal immune network used in IgA production play an important role in HT. Terms such as iron homeostasis, defense response, immune system process, DNA conformational change, production of transforming growth factor beta-2, and oxidoreductase activity were enriched in the gene list, suggesting a potential correlation with HT. A total of 261 lncRNA-miRNA relationship pairs and 21 circRNA-miRNA relationship pairs were obtained; additionally, 5 circRNAs and 13 lncRNAs were screened, which can be used as competing endogenous RNA (ceRNA) to compete with miRNA in the co-expression network. Co-expression network analysis shows that these differentially expressed circRNA and lncRNA may play a vital role in HT and provide valuable information for new biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Rongrong Han
- Department of Clinical Medicine, Jining Medical University, Jining, China
| | - Peng Zhang
- Department of Clinical Medicine, Jining Medical University, Jining, China
| | - Hongfang Li
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yun Chen
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yongnan Hao
- Department of Emergency Stroke, Affiliated Hospital of Jining Medical University, Jining, China
| | - Qiang Guo
- Department of Emergency Stroke, Affiliated Hospital of Jining Medical University, Jining, China
| | - Aimei Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Daojing Li
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|
17
|
Mo Z, Zeng Z, Liu Y, Zeng L, Fang J, Ma Y. Activation of Wnt/Beta-Catenin Signaling Pathway as a Promising Therapeutic Candidate for Cerebral Ischemia/Reperfusion Injury. Front Pharmacol 2022; 13:914537. [PMID: 35668927 PMCID: PMC9163667 DOI: 10.3389/fphar.2022.914537] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/22/2022] [Indexed: 12/11/2022] Open
Abstract
Stroke is one of the leading causes of mortality, and survivors experience serious neurological and motor behavioral deficiencies. Following a cerebral ischemic event, substantial alterations in both cellular and molecular activities occur because of ischemia/reperfusion injury. Wnt signaling is an evolutionarily conserved signaling pathway that has been manifested to play a key role in embryo development and function maintenance in adults. Overactivation of Wnt signaling has previously been investigated in cancer-based research studies. Recently, abnormal Wnt signaling activity has been observed in ischemic stroke, which is accompanied by massive blood–brain barrier (BBB) disruption, neuronal apoptosis, and neuroinflammation within the central nervous system (CNS). Significant therapeutic effects were observed after reactivating the adynamic signaling activity of canonical Wnt signaling in different cell types. To better understand the therapeutic potential of Wnt as a novel target for stroke, we reviewed the role of Wnt signaling in the pathogenesis of stroke in different cell types, including endothelial cells, neurons, oligodendrocytes, and microglia. A comprehensive understanding of Wnt signaling among different cells may help to evaluate its potential value for the development of novel therapeutic strategies based on Wnt activation that can ameliorate complications and improve functional rehabilitation after ischemic stroke.
Collapse
Affiliation(s)
- Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhongyi Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Yuxiang Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Linsheng Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Jiansong Fang, ; Yinzhong Ma,
| | - Yinzhong Ma
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Jiansong Fang, ; Yinzhong Ma,
| |
Collapse
|
18
|
Zhou Z, Ren X, Zheng L, Li A, Zhou W. LncRNA NEAT1 stabilized Wnt3a via U2AF2 and activated Wnt/β-catenin pathway to alleviate ischemia stroke induced injury. Brain Res 2022; 1788:147921. [PMID: 35452660 DOI: 10.1016/j.brainres.2022.147921] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ischaemic stroke is the leading cause of mortality and disability in the world. LncRNA NEAT1 has been shown to play an important role in ischaemic injury, but the molecular mechanism remains unclear. METHODS qRT-PCR was used to determine the expression of lncRNA NEAT1 in OGD/R-induced BV-2 cells. Cell viability was assessed by an MTT assay, and cell apoptosis was assessed by flow cytometry. The expression of related proteins was evaluated by Western blotting and ELISA. The interactions among lncRNA NEAT1, U2AF2 and Wnt3a mRNA was demonstrated by RIP and RNA pulldown assays. XAV-939 was used as an inhibitor of the Wnt/β-catenin pathway. RESULTS LncRNA NEAT1 was found to be downregulated in OGD/R-induced BV-2 cells. Overexpression of lncRNA NEAT1 protected BV-2 cells against OGD/R-induced injury. LncRNA NEAT1 enhanced the stability of Wnt3a mRNA via U2AF2. Knockdown of Wnt3a or blockade of the Wnt/β-catenin pathway rescued the effect of lncRNA NEAT1. CONCLUSIONS LncRNA NEAT1 protected cells against OGD/R-induced apoptosis and the inflammatory response by activating the Wnt/β-catenin pathway through upregulation of Wnt3a in a U2AF2-dependent manner. LncRNA NEAT1 could be a promising therapeutic candidate for ischaemic stroke treatment in the future.
Collapse
Affiliation(s)
- Zhiwen Zhou
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Xiang Ren
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Lijun Zheng
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Aiping Li
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Wensheng Zhou
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China.
| |
Collapse
|
19
|
Ma R, Kutchy NA, Chen L, Meigs DD, Hu G. Primary cilia and ciliary signaling pathways in aging and age-related brain disorders. Neurobiol Dis 2022; 163:105607. [PMID: 34979259 PMCID: PMC9280856 DOI: 10.1016/j.nbd.2021.105607] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Brain disorders are characterized by the progressive loss of structure and function of the brain as a consequence of progressive degeneration and/or death of nerve cells. Aging is a major risk factor for brain disorders such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and stroke. Various cellular and molecular events have been shown to play a role in the progress of neurodegenerative diseases. Emerging studies suggest that primary cilia could be a key regulator in brain diseases. The primary cilium is a singular cellular organelle expressed on the surface of many cell types, such as astrocytes and neurons in the mature brain. Primary cilia detect extracellular cues, such as Sonic Hedgehog (SHH) protein, and transduce these signals into cells to regulate various signaling pathways. Abnormalities in ciliary length and frequency (ratio of ciliated cells) have been implicated in various human diseases, including brain disorders. This review summarizes current findings and thoughts on the role of primary cilia and ciliary signaling pathways in aging and age-related brain disorders.
Collapse
Affiliation(s)
- Rong Ma
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Naseer A Kutchy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Anatomy, Physiology and Pharmacology, School of Veterinary Medicine, St. George's University, Grenada
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, Guangdong 515063, China; Key Laboratory of Intelligent Manufacturing Technology, Ministry of Education, Shantou University, Shantou, Guangdong 515063, China
| | - Douglas D Meigs
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
20
|
Guo R, Wang X, Fang Y, Chen X, Chen K, Huang W, Chen J, Hu J, Liang F, Du J, Dordoe C, Tian X, Lin L. rhFGF20 promotes angiogenesis and vascular repair following traumatic brain injury by regulating Wnt/β-catenin pathway. Biomed Pharmacother 2021; 143:112200. [PMID: 34649342 DOI: 10.1016/j.biopha.2021.112200] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
The pathology of cerebrovascular disorders takes an important role in traumatic brain injury (TBI) by increasing intracranial pressure. Fibroblast growth factor 20 (FGF20) is a brain-derived neurotrophic factor, that has been shown to play an important role in the survival of dopaminergic neurons and the treatment of Parkinson's disease (PD). However, little is known about the role of FGF20 in the treatment of TBI and its underlying mechanism. The purpose of this study was to evaluate the protective effect of recombinant human FGF20 (rhFGF20) on protecting cerebral blood vessels after TBI. In this study, we indicated that rhFGF20 could reduce brain edema, Evans blue penetration and upregulated the expression of blood-brain barrier (BBB)-related tight junction (TJ) proteins, exerting a protective effect on the BBB in vivo after TBI. In the TBI repair phase, rhFGF20 promoted angiogenesis, neurological and cognitive function recovery. In tumor necrosis factor-α (TNF-α)-induced human brain microvascular endothelial cells (hCMEC/D3), an in vitro BBB disruption model, rhFGF20 reversed the impairment in cell migration and tube formation induced by TNF-α. Moreover, in both the TBI mouse model and the in vitro model, rhFGF20 increased the expression of β-catenin and GSK3β, which are the two key regulators in the Wnt/β-catenin signaling pathway. In addition, the Wnt/β-catenin inhibitor IWR-1-endo significantly reversed the effects of rhFGF20. These results indicate that rhFGF20 may prevent vascular repair and angiogenesis through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yani Fang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiongjian Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Kun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenting Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China
| | - Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fei Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xianxi Tian
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China.
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Beijing 100730, China.
| |
Collapse
|
21
|
Kwon HS, Kim YE, Park HH, Son JW, Choi H, Lee YJ, Kim HY, Lee KY, Koh SH. Neuroprotective Effects of GV1001 in Animal Stroke Model and Neural Cells Subject to Oxygen-Glucose Deprivation/Reperfusion Injury. J Stroke 2021; 23:420-436. [PMID: 34649386 PMCID: PMC8521247 DOI: 10.5853/jos.2021.00626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/16/2021] [Indexed: 12/04/2022] Open
Abstract
Background and Purpose Previous studies have revealed the diverse neuroprotective effects of GV1001. In this study, we investigated the effects of GV1001 on focal cerebral ischemia-reperfusion injury (IRI) in rats and oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury in neural stem cells (NSCs) and cortical neurons.
Methods Focal cerebral IRI was induced by transient middle cerebral artery occlusion (MCAO). Brain diffusion-weighted imaging (DWI) was performed 2 hours after occlusion, and a total of 37 rats were treated by reperfusion with GV1001 or saline 2 hours after occlusion. Fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging, immunohistochemistry, and neurobehavioral function analyses were performed. Additionally, OGD/R-injured NSCs and cortical neurons were treated with different GV1001 concentrations. Cell viability, proliferation, migration, and oxidative stress were determined by diverse molecular analyses.
Results In the stroke model, GV1001 protected neural cells against IRI. The most effective dose of GV1001 was 60 μM/kg. The infarct volume on FLAIR 48 hours after MCAO compared to lesion volume on DWI showed a significantly smaller ratio in the GV1001-treated group. GV1001-treated rats exhibited better behavioral functions than the saline-treated rats. Treatment with GV1001 increased the viability, proliferation, and migration of the OGD/R-injured NSCs. Free radicals were significantly restored by treatment with GV1001. These neuroprotective effects of GV1001 have also been demonstrated in OGD/R-injured cortical neurons. Conclusions The results suggest that GV1001 has neuroprotective effects against IRI in NSCs, cortical neurons, and the rat brain. These effects are mediated through the induction of cellular proliferation, mitochondrial stabilization, and anti-apoptotic, anti-aging, and antioxidant effects.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea
| | - Ye Eun Kim
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea
| | - Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea
| | - Jeong-Woo Son
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea
| | - Hojin Choi
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea
| | - Young Joo Lee
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea
| | - Hyun Young Kim
- Department of Neurology, Hanyang University Seoul Hospital, Hanyang University College of Medicine, Seoul, Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Hangyang University College of Medicine, Guri, Korea.,Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| |
Collapse
|
22
|
Liu C, Chen S, Zhang H, Chen Y, Gao Q, Chen Z, Liu Z, Wang J. Bioinformatic analysis for potential biological processes and key targets of heart failure-related stroke. J Zhejiang Univ Sci B 2021; 22:718-732. [PMID: 34514752 DOI: 10.1631/jzus.b2000544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This study aimed to uncover underlying mechanisms and promising intervention targets of heart failure (HF)-related stroke. HF-related dataset GSE42955 and stroke-related dataset GSE58294 were obtained from the Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was conducted to identify key modules and hub genes. Gene Ontology (GO) and pathway enrichment analyses were performed on genes in the key modules. Genes in HF- and stroke-related key modules were intersected to obtain common genes for HF-related stroke, which were further intersected with hub genes of stroke-related key modules to obtain key genes in HF-related stroke. Key genes were functionally annotated through GO in the Reactome and Cytoscape databases. Finally, key genes were validated in these two datasets and other datasets. HF- and stroke-related datasets each identified two key modules. Functional enrichment analysis indicated that protein ubiquitination, Wnt signaling, and exosomes were involved in both HF- and stroke-related key modules. Additionally, ten hub genes were identified in stroke-related key modules and 155 genes were identified as common genes in HF-related stroke. OTU deubiquitinase with linear linkage specificity(OTULIN) and nuclear factor interleukin 3-regulated(NFIL3) were determined to be the key genes in HF-related stroke. Through functional annotation, OTULIN was involved in protein ubiquitination and Wnt signaling, and NFIL3 was involved in DNA binding and transcription. Importantly, OTULIN and NFIL3 were also validated to be differentially expressed in all HF and stroke groups. Protein ubiquitination, Wnt signaling, and exosomes were involved in HF-related stroke. OTULIN and NFIL3 may play a key role in HF-related stroke through regulating these processes, and thus serve as promising intervention targets.
Collapse
Affiliation(s)
- Chiyu Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Sixu Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Haifeng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Yangxin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Qingyuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Zhiteng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China
| | - Zhaoyu Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China. .,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou 510120, China.
| |
Collapse
|
23
|
Owino S, Giddens MM, Jiang JG, Nguyen TT, Shiu FH, Lala T, Gearing M, McCrary MR, Gu X, Wei L, Yu SP, Hall RA. GPR37 modulates progenitor cell dynamics in a mouse model of ischemic stroke. Exp Neurol 2021; 342:113719. [PMID: 33839144 PMCID: PMC9826632 DOI: 10.1016/j.expneurol.2021.113719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/27/2021] [Accepted: 04/06/2021] [Indexed: 01/11/2023]
Abstract
The generation of neural stem and progenitor cells following injury is critical for the function of the central nervous system, but the molecular mechanisms modulating this response remain largely unknown. We have previously identified the G protein-coupled receptor 37 (GPR37) as a modulator of ischemic damage in a mouse model of stroke. Here we demonstrate that GPR37 functions as a critical negative regulator of progenitor cell dynamics and gliosis following ischemic injury. In the central nervous system, GPR37 is enriched in mature oligodendrocytes, but following injury we have found that its expression is dramatically increased within a population of Sox2-positive progenitor cells. Moreover, the genetic deletion of GPR37 did not alter the number of mature oligodendrocytes following injury but did markedly increase the number of both progenitor cells and injury-induced Olig2-expressing glia. Alterations in the glial environment were further evidenced by the decreased activation of oligodendrocyte precursor cells. These data reveal that GPR37 regulates the response of progenitor cells to ischemic injury and provides new perspectives into the potential for manipulating endogenous progenitor cells following stroke.
Collapse
Affiliation(s)
- Sharon Owino
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michelle M. Giddens
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jessie G. Jiang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - TrangKimberly T. Nguyen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fu Hung Shiu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Marla Gearing
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;,Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Myles R. McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan P. Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA;,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA 30033, USA
| | - Randy A. Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
24
|
Chiareli RA, Carvalho GA, Marques BL, Mota LS, Oliveira-Lima OC, Gomes RM, Birbrair A, Gomez RS, Simão F, Klempin F, Leist M, Pinto MCX. The Role of Astrocytes in the Neurorepair Process. Front Cell Dev Biol 2021; 9:665795. [PMID: 34113618 PMCID: PMC8186445 DOI: 10.3389/fcell.2021.665795] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are highly specialized glial cells responsible for trophic and metabolic support of neurons. They are associated to ionic homeostasis, the regulation of cerebral blood flow and metabolism, the modulation of synaptic activity by capturing and recycle of neurotransmitters and maintenance of the blood-brain barrier. During injuries and infections, astrocytes act in cerebral defense through heterogeneous and progressive changes in their gene expression, morphology, proliferative capacity, and function, which is known as reactive astrocytes. Thus, reactive astrocytes release several signaling molecules that modulates and contributes to the defense against injuries and infection in the central nervous system. Therefore, deciphering the complex signaling pathways of reactive astrocytes after brain damage can contribute to the neuroinflammation control and reveal new molecular targets to stimulate neurorepair process. In this review, we present the current knowledge about the role of astrocytes in brain damage and repair, highlighting the cellular and molecular bases involved in synaptogenesis and neurogenesis. In addition, we present new approaches to modulate the astrocytic activity and potentiates the neurorepair process after brain damage.
Collapse
Affiliation(s)
| | | | | | - Lennia Soares Mota
- Department of Pharmacology, Federal University of Goias, Goiânia, Brazil
| | | | | | - Alexander Birbrair
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Renato Santiago Gomez
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Simão
- Research Division, Vascular Cell Biology, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | | | - Marcel Leist
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
25
|
Xiao Z, Cao Z, Yang J, Jia Z, Du Y, Sun G, Lu Y, Pei L. Baicalin promotes hippocampal neurogenesis via the Wnt/β-catenin pathway in a chronic unpredictable mild stress-induced mouse model of depression. Biochem Pharmacol 2021; 190:114594. [PMID: 33964281 DOI: 10.1016/j.bcp.2021.114594] [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: 03/15/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 11/29/2022]
Abstract
Hippocampal neurogenesis is known to be related to depressive symptoms. Increasing evidence indicates that Wnt/β-catenin signaling regulates multiple aspects of adult hippocampal neurogenesis. Baicalin is a major flavonoid compound with multiple pharmacological effects such as anti-inflammatory, anti-apoptotic, and neuroprotective effects. The current study aimed to explore the antidepressant effects of baicalin and its possible molecular mechanisms affecting hippocampal neurogenesis via the regulation of the Wnt/β-catenin signaling pathway. A chronic mild unpredictable stress (CUMS) model of depression was used in the study. The CUMS-induced mice were treated with baicalin (50 and 100 mg/kg) for 21 days, orally, and the fluoxetine was used as positive control drug. The results indicated that baicalin alleviated CUMS-induced depression-like behaviour, and improved the nerve cells' survival of the hippocampal dentate gyrus (DG) in CUMS-induced depression of model mice and increased Ki-67- and doublecortin (DCX)-positive cells to restore CUMS-induced suppression of hippocampal neurogenesis. The related proteins in the Wnt/β-catenin signaling pathway, which declined in the CUMS-induced depression model of mice, were upregulated after baicalin treatment, including Wingless3a (Wnt3a), dishevelled2 (DVL2), and β-catenin. Further study found that the phosphorylation rate of glycogen synthase kinase-3β (GSK3β) and β-catenin nuclear translocation increased, as the levels of the β-catenin target genes cyclinD1, c-myc, NeuroD1, and Ngn2 upregulated after baicalin treatment. In conclusion, these findings suggest that baicalin may promote hippocampal neurogenesis, thereby exerting the antidepressant effect via regulation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Zhigang Xiao
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Province Academy of Chinese Medicine Sciences, Shijiazhuang 050031, China
| | - Zhuoqing Cao
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Province Academy of Chinese Medicine Sciences, Shijiazhuang 050031, China
| | - Jiali Yang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Province Academy of Chinese Medicine Sciences, Shijiazhuang 050031, China
| | - Zhixia Jia
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Province Academy of Chinese Medicine Sciences, Shijiazhuang 050031, China
| | - Yuru Du
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China
| | - Guoqiang Sun
- Hebei Province Academy of Chinese Medicine Sciences, Shijiazhuang 050031, China
| | - Ye Lu
- Hebei Province Academy of Chinese Medicine Sciences, Shijiazhuang 050031, China.
| | - Lin Pei
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Province Academy of Chinese Medicine Sciences, Shijiazhuang 050031, China.
| |
Collapse
|
26
|
Chen H, Zhao Y, Zhang J, Xie Y, Jin M. Promoting effects of MiR-135b on human multiple myeloma cells via regulation of the Wnt/β-catenin/Versican signaling pathway. Cytokine 2021; 142:155495. [PMID: 33765653 DOI: 10.1016/j.cyto.2021.155495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 03/04/2021] [Indexed: 01/02/2023]
Abstract
MicroRNA (MiR)-135b and its mediated Wnt/β-catenin signaling pathway are involved in human malignancies. However, their roles in multiple myeloma (MM) remained poorly understood. Our study aimed to uncover their roles in MM. MiR-135b and Versican expressions were measured using quantitative real-time polymerase chain reaction (qRT-PCR). MM cell proliferation, apoptosis, migration and invasion were detected by cell counting kit-8 (CCK-8) assay, flow cytometry, wound healing assay and transwell assay, respectively. Relative expression of Wnt/β-catenin signaling pathway-related protein was quantified by Western blot. MiR-135b was upregulated in the serum of MM patients, and miR-135b upregulation promoted MM cell proliferation, migration and invasion but suppressed apoptosis. Also, miR-135b upregulation promoted activation of Wnt/β-catenin signaling pathway. However, downregulation of miR-135b caused an opposite effect. After incubating cells with miR-135b inhibitor and Wnt/β-catenin signaling pathway agonist Lithium chloride (LiCl), which reversed the effects of downregulating miR-135b. Versican is the downstream effector of the Wnt/β-catenin signaling pathway, and its silencing reversed the effects of LiCl on MM cells. In conclusion, miR-135b and its mediated Wnt/β-catenin signaling pathway promoted proliferation, migration and invasion but suppressed apoptosis of MM cells through regulating Versican, providing a possible treatment for MM.
Collapse
Affiliation(s)
- Hong Chen
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhao
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jiajia Zhang
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yan Xie
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Mulan Jin
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
27
|
Phosphofructokinase-1 Inhibition Promotes Neuronal Differentiation of Neural Stem Cells and Functional Recovery After Stroke. Neuroscience 2021; 459:27-38. [PMID: 33556456 DOI: 10.1016/j.neuroscience.2021.01.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
Abstract
Ischemic stroke is a major cause of long-term disability. Neuronal differentiation of neural stem cells (NSCs) is crucial for brain repair after stroke. However, the underlying mechanisms remain unclear. Here, the role and potential mechanisms of phosphofructokinase-1 (PFK-1), the rate-limiting enzyme of glycolysis, was investigated in stroke using middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation models. We found that stroke increased the PFK-1 expression of NSCs. However, PFK-1 inhibition promoted neuronal differentiation of NSCs and facilitated the dendritic maturation of newborn neurons in vitro and in vivo. Moreover, PFK-1 inhibition also improved the spatial memory performance of MCAO rats. Additionally, we proved that the effect of PFK-1 inhibition above might be achieved by promoting β-catenin nuclear translocation and activating its downstream signaling, independent of Wnt signaling. Thus, these observations reveal a critical role of PFK-1 in stroke, which may provide a novel target for regenerative repair after stroke.
Collapse
|
28
|
Ji YB, Gao Q, Tan XX, Huang XW, Ma YZ, Fang C, Wang SN, Qiu LH, Cheng YX, Guo FY, Chang J. Lithium alleviates blood-brain barrier breakdown after cerebral ischemia and reperfusion by upregulating endothelial Wnt/β-catenin signaling in mice. Neuropharmacology 2021; 186:108474. [PMID: 33524408 DOI: 10.1016/j.neuropharm.2021.108474] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
Although upregulation of endothelial Wnt/β-catenin signaling may be used to treat blood-brain barrier (BBB) breakdown caused by cerebral ischemia/reperfusion injury, no agents based on this mechanism are available clinically. Lithium, a medication used for treating bipolar mood disorders, upregulates Wnt/β-catenin signaling, but whether lithium alleviates BBB breakdown after ischemic stroke by upregulating endothelial Wnt/β-catenin signaling is unclear. Here, we evaluated the BBB-protective effect of lithium in adult mice with 1-h middle cerebral artery occlusion and 48-h reperfusion (MCAO/R) by determining neurological outcomes, BBB function and related molecular components. Furthermore, we assessed the effect and dependence of lithium on Wnt/β-catenin signaling in brain microvascular endothelial cells in cell culture and in mice with conditional endothelial knockout of Wnt7 co-receptor Gpr124. Our data show that lithium treatment (3 mmol/kg) significantly decreased infarct volume (34.1 ± 1.8% versus 58.3 ± 2.8% in vehicle controls, P < 0.0001) and improved neurological outcomes of mice following MCAO/R. Importantly, lithium significantly increased BBB integrity shown by reduction of Evans blue leakage (by 45.7%, P = 0.0064) and blood IgG extravasation (by 65.8%, P < 0.0001) into infarcted brain tissue. Mechanistically, lithium upregulated the activity of endothelial Wnt/β-catenin signaling in vivo and in vitro, increased the protein levels of tight junctions (Claudin-5 and ZO-1), and reduced MMP-9 expression. Furthermore, the protective effect of lithium on cerebral damage and BBB integrity was abolished in endothelial Gpr124 knockout mice, indicating the protection of lithium on BBB was mainly dependent on the Gpr124-mediated endothelial Wnt/β-catenin signaling. Taken together, our findings indicate that lithium may serve as a therapeutic candidate for treating the BBB breakdown in the early stage of ischemic stroke following reperfusion therapy.
Collapse
Affiliation(s)
- Ya-Bin Ji
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiang Gao
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Xi-Xi Tan
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Neurology, Yangjiang People's Hospital, Yangjiang, 529500, China
| | - Xiao-Wen Huang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yin-Zhong Ma
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Cheng Fang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Sheng-Nan Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lin-Hui Qiu
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yong-Xian Cheng
- School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, 518061, China
| | - Fu-You Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450001, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
29
|
Yang X, Song D, Chen L, Xiao H, Ma X, Jiang Q, Cheng O. Curcumin promotes neurogenesis of hippocampal dentate gyrus via Wnt/β-catenin signal pathway following cerebral ischemia in mice. Brain Res 2020; 1751:147197. [PMID: 33160958 DOI: 10.1016/j.brainres.2020.147197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/31/2020] [Accepted: 10/31/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVES To investigate whether curcumin promotes hippocampal neurogenesis in the cerebral ischemia (CI) mice via Wnt/β-catenin signaling pathway. METHODS Male C57BL/6 mice were randomly divided into groups: sham operation group (Sham), cerebral ischemic group (CI), curcumin treatment group (50, 100 mg/kg/d, i.p.) and curcumin (100 mg/kg/d) + DKK1 (a blocker of Wnt receptor, 200 ng/d, icv) group. CI was induced by bilateral common carotid arteries occlusion (BCCAO) for 20 min. The Morris water maze test was conducted to detect spatial learning and memory. Immunofluorescence staining was used to examine the proliferation and differentiation of immature neurons in the hippocampal dentate gyrus. The proteins involved in neurogenesis and Wnt signaling pathway were examined using Western blot assay. RESULTS Curcumin significantly alleviated cognitive deficits induced by CI. Curcumin dose-dependently increased the proliferation of neural stem cells and promoted the differentiation and maturation of newly generated neural cells into neurons. Curcumin also increased the expression of proteins involved in neurogenesis (including Ngn2, Pax6 and NeuroD 1) and the Wnt/β-catenin signaling pathway. Moreover, the forenamed effects of curcumin were abolished by pretreatment with DKK1, a blocker of Wnt receptor. CONCLUSION Curcumin promotes hippocampal neurogenesis by activating Wnt/β-catenin signaling pathway to ameliorate cognitive deficits after acute CI.
Collapse
Affiliation(s)
- Xuemei Yang
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Dan Song
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China; Laboratory Research Center, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lili Chen
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China; Laboratory Research Center, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Huan Xiao
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xiaojiao Ma
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Qingsong Jiang
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Lipid Emulsion Improves Functional Recovery in an Animal Model of Stroke. Int J Mol Sci 2020; 21:ijms21197373. [PMID: 33036206 PMCID: PMC7582956 DOI: 10.3390/ijms21197373] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 12/16/2022] Open
Abstract
Stroke is a life-threatening condition that leads to the death of many people around the world. Reperfusion injury after ischemic stroke is a recurrent problem associated with various surgical procedures that involve the removal of blockages in the brain arteries. Lipid emulsion was recently shown to attenuate ischemic reperfusion injury in the heart and to protect the brain from excitotoxicity. However, investigations on the protective mechanisms of lipid emulsion against ischemia in the brain are still lacking. This study aimed to determine the neuroprotective effects of lipid emulsion in an in vivo rat model of ischemic reperfusion injury through middle cerebral artery occlusion (MCAO). Under sodium pentobarbital anesthesia, rats were subjected to MCAO surgery and were administered with lipid emulsion through intra-arterial injection during reperfusion. The experimental animals were assessed for neurological deficit wherein the brains were extracted at 24 h after reperfusion for triphenyltetrazolium chloride staining, immunoblotting and qPCR. Neuroprotection was found to be dosage-dependent and the rats treated with 20% lipid emulsion had significantly decreased infarction volumes and lower Bederson scores. Phosphorylation of Akt and glycogen synthase kinase 3-β (GSK3-β) were increased in the 20% lipid-emulsion treated group. The Wnt-associated signals showed a marked increase with a concomitant decrease in signals of inflammatory markers in the group treated with 20% lipid emulsion. The protective effects of lipid emulsion and survival-related expression of genes such as Akt, GSK-3β, Wnt1 and β-catenin were reversed by the intra-peritoneal administration of XAV939 through the inhibition of the Wnt/β-catenin signaling pathway. These results suggest that lipid emulsion has neuroprotective effects against ischemic reperfusion injury in the brain through the modulation of the Wnt signaling pathway and may provide potential insights for the development of therapeutic targets.
Collapse
|
31
|
Qu M, Zhao J, Zhao Y, Sun J, Liu L, Wei L, Zhang Y. Vascular protection and regenerative effects of intranasal DL-3-N-butylphthalide treatment after ischaemic stroke in mice. Stroke Vasc Neurol 2020; 6:74-79. [PMID: 32958696 PMCID: PMC8005898 DOI: 10.1136/svn-2020-000364] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 12/23/2022] Open
Abstract
Objective To investigate the effects of DL-3-N-butylphthalide (NBP) via intranasal delivery after ischaemic stroke in mice. Methods C57BL/6 mice were divided into three groups: sham, stroke with vehicle and stroke with NBP treatment. Ischaemic stroke was induced by permanent ligation of right middle cerebral artery with 7 min common carotid artery occlusion. NBP (100 mg/kg) or vehicle was intranasally administered at 1 hour after stroke and repeated once a day until sacrifice. Bromodeoxyuridine (BrdU) (50 mg/kg/day) was given from the third day until sacrifice. Sensorimotor function was tested during 1–21 days after stroke. Local cerebral blood flow in the ischaemic and peri-infarct regions was measured using laser Doppler flowmetry before, during and 3 days after ischaemia. Expressions of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase as well as regenerative marker BrdU in the peri-infarct region were analysed by western blotting and immunohistochemical methods. Results Compared with the vehicle group, NBP treatment significantly increased the VEGF expression in the poststroke brain. Stroke mice that received NBP showed significantly less vascular damage after stroke and more new neurons and blood vessels in the peri-infarct region at 21 days after stroke. In the adhesive removal test, the sensorimotor function of stroke mice treated with NBP performed significantly better at 1, 3 and 7 days after stroke compared with vehicle controls. Conclusion Daily intranasal NBP treatment provides protective and neurogenic/angiogenic effects in the poststroke brain, accompanied with functional improvements after a focal ischaemic stroke in mice.
Collapse
Affiliation(s)
- Mengyao Qu
- Neurology, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China.,Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jingjie Zhao
- Chinese Traditional Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Yingying Zhao
- Neurology, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Jinmei Sun
- Neurology, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Liping Liu
- China National Clinical Research Center for Neurological Diseases, Beijing, China.,Neurology, Tiantan Clinical Trial and Research Center for Stroke, Beijing, China
| | - Ling Wei
- Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yongbo Zhang
- Neurology, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| |
Collapse
|
32
|
Menet R, Lecordier S, ElAli A. Wnt Pathway: An Emerging Player in Vascular and Traumatic Mediated Brain Injuries. Front Physiol 2020; 11:565667. [PMID: 33071819 PMCID: PMC7530281 DOI: 10.3389/fphys.2020.565667] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
The Wnt pathway, which comprises the canonical and non-canonical pathways, is an evolutionarily conserved mechanism that regulates crucial biological aspects throughout the development and adulthood. Emergence and patterning of the nervous and vascular systems are intimately coordinated, a process in which Wnt pathway plays particularly important roles. In the brain, Wnt ligands activate a cell-specific surface receptor complex to induce intracellular signaling cascades regulating neurogenesis, synaptogenesis, neuronal plasticity, synaptic plasticity, angiogenesis, vascular stabilization, and inflammation. The Wnt pathway is tightly regulated in the adult brain to maintain neurovascular functions. Historically, research in neuroscience has emphasized essentially on investigating the pathway in neurodegenerative disorders. Nonetheless, emerging findings have demonstrated that the pathway is deregulated in vascular- and traumatic-mediated brain injuries. These findings are suggesting that the pathway constitutes a promising target for the development of novel therapeutic protective and restorative interventions. Yet, targeting a complex multifunctional signal transduction pathway remains a major challenge. The review aims to summarize the current knowledge regarding the implication of Wnt pathway in the pathobiology of ischemic and hemorrhagic stroke, as well as traumatic brain injury (TBI). Furthermore, the review will present the strategies used so far to manipulate the pathway for therapeutic purposes as to highlight potential future directions.
Collapse
Affiliation(s)
- Romain Menet
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
33
|
Fei YX, Zhu JP, Zhao B, Yin QY, Fang WR, Li YM. XQ-1H regulates Wnt/GSK3β/β-catenin pathway and ameliorates the integrity of blood brain barrier in mice with acute ischemic stroke. Brain Res Bull 2020; 164:269-288. [PMID: 32916221 DOI: 10.1016/j.brainresbull.2020.08.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 07/30/2020] [Accepted: 08/30/2020] [Indexed: 02/08/2023]
Abstract
10-O-(N, N-dimethylaminoethyl) ginkgolide B methanesulfonate (XQ-1H), a novel analog of ginkgolide B, has been preliminarily recognized to show bioactivities against ischemia-induced injury. However, the underlying mechanism still remains to be fully elucidated. The aim of this study was to investigate the effect of XQ-1H against cerebral ischemia/reperfusion injury (CIRI) from the perspective of blood brain barrier (BBB) protection, and explore whether the underlying mechanism is associated with Wnt/GSK3β/β-catenin signaling pathway activation. The therapeutic effects of XQ-1H were evaluated in mice subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) and in immortalized mouse cerebral endothelial cells (bEnd.3) challenged by oxygen and glucose deprivation/reoxygenation (OGD/R). Results showed that treatment with XQ-1H improved neurological behavior, reduced brain infarction volume, diminished edema, and attenuated the disruption of BBB in vivo. In vitro, XQ-1H increased cell viability and maintained the barrier function of bEnd.3 monolayer after OGD/R. Moreover, the protection of XQ-1H was accompanied with activation of Wnt/GSK3β/β-catenin pathway and upregulation of tight junction proteins. Notably, the protection of XQ-1H was abolished by Wnt/GSK3β/β-catenin inhibitor XAV939 or β-catenin siRNA, indicating XQ-1H exerted protection in a Wnt/GSK3β/β-catenin dependent profile. In summary, XQ-1H attenuated brain injury and maintained BBB integrity after CIRI, and the possible underlying mechanism may be related to the activation of Wnt/GSK3β/β-catenin pathway and upregulation of tight junction proteins.
Collapse
Affiliation(s)
- Yu-Xiang Fei
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jian-Ping Zhu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bo Zhao
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Qi-Yang Yin
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wei-Rong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yun-Man Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
34
|
Marchetti B. Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson's disease. Redox Biol 2020; 36:101664. [PMID: 32863224 PMCID: PMC7395594 DOI: 10.1016/j.redox.2020.101664] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress and inflammation have long been recognized to contribute to Parkinson's disease (PD), a common movement disorder characterized by the selective loss of midbrain dopaminergic neurons (mDAn) of the substantia nigra pars compacta (SNpc). The causes and mechanisms still remain elusive, but a complex interplay between several genes and a number of interconnected environmental factors, are chiefly involved in mDAn demise, as they intersect the key cellular functions affected in PD, such as the inflammatory response, mitochondrial, lysosomal, proteosomal and autophagic functions. Nuclear factor erythroid 2 -like 2 (NFE2L2/Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling cascade, a vital pathway for mDAn neurogenesis and neuroprotection, emerge as critical intertwinned actors in mDAn physiopathology, as a decline of an Nrf2/Wnt/β-catenin prosurvival axis with age underlying PD mutations and a variety of noxious environmental exposures drive PD neurodegeneration. Unexpectedly, astrocytes, the so-called "star-shaped" cells, harbouring an arsenal of "beneficial" and "harmful" molecules represent the turning point in the physiopathological and therapeutical scenario of PD. Fascinatingly, "astrocyte's fil rouge" brings back to Nrf2/Wnt resilience, as boosting the Nrf2/Wnt resilience program rejuvenates astrocytes, in turn (i) mitigating nigrostriatal degeneration of aged mice, (ii) reactivating neural stem progenitor cell proliferation and neuron differentiation in the brain and (iii) promoting a beneficial immunomodulation via bidirectional communication with mDAns. Then, through resilience of Nrf2/Wnt/β-catenin anti-ageing, prosurvival and proregenerative molecular programs, it seems possible to boost the inherent endogenous self-repair mechanisms. Here, the cellular and molecular aspects as well as the therapeutical options for rejuvenating glia-neuron dialogue will be discussed together with major glial-derived mechanisms and therapies that will be fundamental to the identification of novel diagnostic tools and treatments for neurodegenerative diseases (NDs), to fight ageing and nigrostriatal DAergic degeneration and promote functional recovery.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology Section, Medical School, University of Catania, Via S. Sofia 65, 95125, Catania, Italy; Oasi Research Institute-IRCCS, Neuropharmacology Section, Via Conte Ruggero 73, 94018, Troina, EN, Italy.
| |
Collapse
|
35
|
Xu D, Li F, Hou K, Gou X, Fang W, Li Y. XQ-1H attenuates ischemic injury in PC12 cells via Wnt/β-catenin signaling though inhibition of apoptosis and promotion of proliferation. Cell Biol Int 2020; 44:2363-2369. [PMID: 32761926 DOI: 10.1002/cbin.11438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/15/2020] [Accepted: 08/01/2020] [Indexed: 01/10/2023]
Abstract
10-O-(N,N-dimethylaminoethyl)-ginkgolide B methanesulfonate (XQ-1H) is a new derivative of ginkgolide B and has previously been proven to exert neuroprotective effects on ischemic injury. However, it is not clear whether XQ-1H affects the cell survival and proliferation in oxygen-glucose deprivation/reoxygenation (OGD/R) damaged PC12 cells. Our results showed that OGD/R improved cell viability after 24 hr of posttreatment with XQ-1H (10 or 5 μM), inhibiting cell injury and apoptosis by upregulating the expression of brain-derived neurotrophic factor, nerve growth factor, and antiapoptotic B-cell lymphoma-extra large, while reducing proapoptotic cleaved caspase-3 protein. By introducing the Wnt/β-catenin signaling inhibitor XAV-939 and 5-bromo-2'-deoxyuridine staining, it was proved that XQ-1H promoted the proliferation of PC12 cells in a Wnt-signal-dependent manner via inhibiting the activation of glycogen synthase kinase-3β after phosphatidylinositol 3-kinase/protein kinase B signal activation, thereby activating Wnt1, β-catenin, and the expression of downstream neurogenic differentiation 1 and cyclin D1, which was comparable to Wnt/β-catenin signaling agonist 4,6-disubstituted pyrrolopyrimidine. We conclude that XQ-1H, after OGD/R damage to PC12 cells, may limit cell apoptosis in a Wnt/β-catenin signal-dependent manner, promoting cell proliferation and survival.
Collapse
Affiliation(s)
- Dan Xu
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Fengyang Li
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Kai Hou
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xue Gou
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Weirong Fang
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yunman Li
- Department of Physiology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
36
|
Xu D, Li F, Xue G, Hou K, Fang W, Li Y. Effect of Wnt signaling pathway on neurogenesis after cerebral ischemia and its therapeutic potential. Brain Res Bull 2020; 164:1-13. [PMID: 32763283 DOI: 10.1016/j.brainresbull.2020.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/08/2019] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Neurogenesis process in the chronic phase of ischemic stroke has become the focus of research on stroke treatment recently, mainly through the activation of related pathways to increase the differentiation of neural stem cells (NSCs) in the brain sub-ventricular zone (SVZ) and subgranular zone (SGZ) of hippocampal dentate gyrus (DG) areas into neurons, promoting neurogenesis. While there is still debate about the longevity of active adult neurogenesis in humans, the SVZ and SGZ have the capacity to upregulate neurogenesis in response to cerebral ischemia, which opens discussion about potential treatment strategies to harness this neuronal regenerative response. Wnt signaling pathway is one of the most important approaches potentially targeting on neurogenesis after cerebral ischemia, appropriate activation of which in NSCs may help to improve the sequelae of cerebral ischemia. Various therapeutic approaches are explored on preclinical stage to target endogenous neurogenesis induced by Wnt signaling after stroke onset. This article describes the composition of Wnt signaling pathway and the process of neurogenesis after cerebral ischemia, and emphatically introduces the recent studies on the mechanisms of this pathway for post-stroke neurogenesis and the therapeutic possibility of activating the pathway to improve neurogenesis after stroke.
Collapse
Affiliation(s)
- Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Gou Xue
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
37
|
Glia and Neural Stem and Progenitor Cells of the Healthy and Ischemic Brain: The Workplace for the Wnt Signaling Pathway. Genes (Basel) 2020; 11:genes11070804. [PMID: 32708801 PMCID: PMC7397164 DOI: 10.3390/genes11070804] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
Wnt signaling plays an important role in the self-renewal, fate-commitment and survival of the neural stem/progenitor cells (NS/PCs) of the adult central nervous system (CNS). Ischemic stroke impairs the proper functioning of the CNS and, therefore, active Wnt signaling may prevent, ameliorate, or even reverse the negative effects of ischemic brain injury. In this review, we provide the current knowledge of Wnt signaling in the adult CNS, its status in diverse cell types, and the Wnt pathway’s impact on the properties of NS/PCs and glial cells in the context of ischemic injury. Finally, we summarize promising strategies that might be considered for stroke therapy, and we outline possible future directions of the field.
Collapse
|
38
|
Zhang Y, Zhao Y, Song X, Luo H, Sun J, Han C, Gu X, Li J, Cai G, Zhu Y, Liu Z, Wei L, Wei ZZ. Modulation of Stem Cells as Therapeutics for Severe Mental Disorders and Cognitive Impairments. Front Psychiatry 2020; 11:80. [PMID: 32425815 PMCID: PMC7205035 DOI: 10.3389/fpsyt.2020.00080] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Severe mental illnesses (SMI) such as schizophrenia and bipolar disorder affect 2-4% of the world population. Current medications and diagnostic methods for mental illnesses are not satisfying. In animal studies, stem cell therapy is promising for some neuropsychiatric disorders and cognitive/social deficits, not only treating during development (targeting modulation and balancing) but also following neurodegeneration (cell replacement and regenerating support). We believe that novel interventions such as modulation of particular cell populations to develop cell-based treatment can improve cognitive and social functions in SMI. With pathological synaptic/myelin damage, oligodendrocytes seem to play a role. In this review, we have summarized oligodendrogenesis mechanisms and some related calcium signals in neural cells and stem/progenitor cells. The related benefits from endogenous stem/progenitor cells within the brain and exogenous stem cells, including multipotent mesenchymal-derived stromal cells (MSC), fetal neural stem cells (NSC), pluripotent stem cells (PSC), and differentiated progenitors, are discussed. These also include stimulating mechanisms of oligodendrocyte proliferation, maturation, and myelination, responsive to the regenerative effects by both endogenous stem cells and transplanted cells. Among the mechanisms, calcium signaling regulates the neuronal/glial progenitor cell (NPC/GPC)/oligodendrocyte precursor cell (OPC) proliferation, migration, and differentiation, dendrite development, and synaptic plasticity, which are involved in many neuropsychiatric diseases in human. On the basis of numerous protein annotation and protein-protein interaction databases, a total of 119 calcium-dependent/activated proteins that are related to neuropsychiatry in human are summarized in this investigation. One of the advanced methods, the calcium/cation-channel-optogenetics-based stimulation of stem cells and transplanted cells, can take advantage of calcium signaling regulations. Intranasal-to-brain delivery of drugs and stem cells or local delivery with the guidance of brain imaging techniques may provide a unique new approach for treating psychiatric disorders. It is also expected that preconditioning stem cell therapy following precise brain imaging as pathological confirmation has high potential if translated to cell clinic use. Generally, modulable cell transplantation followed by stimulations should provide paracrine protection, synaptic modulation, and myelin repair for the brain in SMI.
Collapse
Affiliation(s)
- Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yingying Zhao
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Xiaopeng Song
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| | - Hua Luo
- Emory Critical Care Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Jinmei Sun
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Chunyu Han
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jun Li
- Department of Biological Psychiatry, Peking University Sixth Hospital, Beijing, China
- Department of Biological Psychiatry, Peking University Institute of Mental Health, Beijing, China
- Department of Biological Psychiatry, NHC Key Laboratory of Mental Health (Peking University), Beijing, China
- Department of Biological Psychiatry, National Clinical Research Center for Mental Disorders, Beijing, China
| | - Guilan Cai
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanbing Zhu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhandong Liu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
39
|
Ruan W, Hu J, Zhou H, Li Y, Xu C, Luo Y, Chen T, Xu B, Yan F, Chen G. Intranasal wnt-3a alleviates neuronal apoptosis in early brain injury post subarachnoid hemorrhage via the regulation of wnt target PPAN mediated by the moonlighting role of aldolase C. Neurochem Int 2020; 134:104656. [DOI: 10.1016/j.neuint.2019.104656] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/22/2019] [Accepted: 12/29/2019] [Indexed: 01/01/2023]
|
40
|
Wang Y, Tian D, Wei C, Cui V, Wang H, Zhu Y, Wu A, Yue Y. Propofol Attenuates α-Synuclein Aggregation and Neuronal Damage in a Mouse Model of Ischemic Stroke. Neurosci Bull 2020; 36:289-298. [PMID: 31520398 PMCID: PMC7056784 DOI: 10.1007/s12264-019-00426-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/03/2019] [Indexed: 01/31/2023] Open
Abstract
α-Synuclein is a soluble monomer abundant in the central nervous system. Aggregates of α-synuclein, consisting of higher-level oligomers and insoluble fibrils, have been observed in many chronic neurological diseases and are implicated in neurotoxicity and neurodegeneration. α-Synuclein has recently been shown to aggregate following acute ischemic stroke, exacerbating neuronal damage. Propofol is an intravenous anesthetic that is commonly used during intravascular embolectomy following acute ischemic stroke. While propofol has demonstrated neuroprotective properties following brain injury, the mechanism of protection in the setting of ischemic stroke is unclear. In this study, propofol administration significantly reduced the neurotoxic aggregation of α-synuclein, decreased the infarct area, and attenuated the neurological deficits after ischemic stroke in a mouse model. We then demonstrated that the propofol-induced reduction of α-synuclein aggregation was associated with increased mammalian target of rapamycin/ribosomal protein S6 kinase beta-1 signaling pathway activity and reduction of the excessive autophagy occurring after acute ischemic stroke.
Collapse
Affiliation(s)
- Yuzhu Wang
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Dan Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Victoria Cui
- Washington University School of Medicine, St. Louis, MI, USA
| | - Huan Wang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yanbing Zhu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Yun Yue
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
41
|
Cheng J, Shen W, Jin L, Pan J, Zhou Y, Pan G, Xie Q, Hu Q, Wu S, Zhang H, Chen X. Treadmill exercise promotes neurogenesis and myelin repair via upregulating Wnt/β‑catenin signaling pathways in the juvenile brain following focal cerebral ischemia/reperfusion. Int J Mol Med 2020; 45:1447-1463. [PMID: 32323740 PMCID: PMC7138282 DOI: 10.3892/ijmm.2020.4515] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/20/2020] [Indexed: 12/15/2022] Open
Abstract
Physical exercise has a neuroprotective effect and is an important treatment after ischemic stroke. Promoting neurogenesis and myelin repair in the penumbra is an important method for the treatment of ischemic stroke. However, the role and potential mechanism of exercise in neurogenesis and myelin repair still needs to be clarified. The goal of the present study was to ascertain the possible effect of treadmill training on the neuroprotective signaling pathway in juvenile rats after ischemic stroke. The model of middle cerebral artery occlusion (MCAO) in juvenile rats was established and then the rats were randomly divided into 9 groups. XAV939 (an inhibitor of the Wnt/β‑catenin pathway) was used to confirm the effects of the Wnt/β‑catenin signaling pathway on exercise‑mediated neurogenesis and myelin repair. Neurological deficits were detected by modified neurological severity score, the injury of brain tissue and the morphology of neurons was detected by hematoxylin‑eosin staining and Nissl staining, and the infarct volume was detected by 2,3,5‑triphenyl tetrazolium chloride staining. The changes in myelin were observed by Luxol fast blue staining. The neuron ultrastructure was observed by transmission electron microscopy. Immunofluorescence and western blots analyzed the molecular mechanisms. The results showed that treadmill exercise improved neurogenesis, enhanced myelin repair, promoted neurological function recovery and reduced infarct volume. These were the results of the upregulation of Wnt3a and nucleus β‑catenin, brain‑derived neurotrophic factor (BDNF) and myelin basic protein (MBP). In addition, XAV939 inhibited treadmill exercise‑induced neurogenesis and myelin repair, which was consistent with the downregulation of Wnt3a, nucleus β‑catenin, BDNF and MBP expression, and the deterioration of neurological function. In summary, treadmill exercise promotes neurogenesis and myelin repair by upregulating the Wnt/β‑catenin signaling pathway, to improve the neurological deficit caused by focal cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Jingyan Cheng
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Weimin Shen
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Lingqin Jin
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Juanjuan Pan
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Yan Zhou
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Guoyuan Pan
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Qingfeng Xie
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Quan Hu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Shamin Wu
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Hongmei Zhang
- Nursing Department, Hangzhou Children's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Xiang Chen
- Physical Medicine and Rehabilitation Center, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| |
Collapse
|
42
|
Chang CY, Liang MZ, Wu CC, Huang PY, Chen HI, Yet SF, Tsai JW, Kao CF, Chen L. WNT3A Promotes Neuronal Regeneration upon Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21041463. [PMID: 32098078 PMCID: PMC7073099 DOI: 10.3390/ijms21041463] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 01/12/2023] Open
Abstract
The treatment of traumatic brain injury (TBI) remains a challenge due to limited knowledge about the mechanisms underlying neuronal regeneration. This current study compared the expression of WNT genes during regeneration of injured cortical neurons. Recombinant WNT3A showed positive effect in promoting neuronal regeneration via in vitro, ex vivo, and in vivo TBI models. Intranasal administration of WNT3A protein to TBI mice increased the number of NeuN+ neurons without affecting GFAP+ glial cells, compared to control mice, as well as retained motor function based on functional behavior analysis. Our findings demonstrated that WNT3A, 8A, 9B, and 10A promote regeneration of injured cortical neurons. Among these WNTs, WNT3A showed the most promising regenerative potential in vivo, ex vivo, and in vitro.
Collapse
Affiliation(s)
- Chu-Yuan Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Min-Zong Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Ching-Chih Wu
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Pei-Yuan Huang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Hong-I Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan;
| | - Jin-Wu Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11574, Taiwan
- Correspondence: (C.-F.K.); (L.C.); Tel.: +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: (C.-F.K.); (L.C.); Tel.: +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| |
Collapse
|
43
|
Gao K, Zhang Y, Niu J, Nie Z, Liu Q, Lv C. Zinc promotes cell apoptosis via activating the Wnt-3a/β-catenin signaling pathway in osteosarcoma. J Orthop Surg Res 2020; 15:57. [PMID: 32075661 PMCID: PMC7029609 DOI: 10.1186/s13018-020-01585-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The zinc content in the blood and tumor tissues of patients with osteosarcoma and the underlying regulation and molecular mechanism of zinc have not been reported. METHODS AND RESULTS This study showed that the zinc content in the blood and tumor tissues of patients with osteosarcoma significantly reduced. CCK-8 and Transwell chamber assays revealed that zinc treatment significantly inhibited the proliferation and invasion abilities of osteosarcoma cells. Western blot analysis indicated that the expression levels of caspase-3 and caspase-9 were significantly increased, suggesting that zinc inhibited the growth and promoted the apoptosis of osteosarcoma cells. In addition, the expression levels of Wnt-3a and β-catenin, the marker proteins of the Wnt/β-catenin signaling pathways, were significantly increased in osteosarcoma cells after zinc intervention, which demonstrated that the pathway was clearly activated. However, the effect of zinc on the apoptosis, proliferation, and invasion abilities of osteosarcoma cells was reversed when the Wnt/β-catenin signaling pathways was inhibited by XAV939 (Wnt antagonist) treatment. CONCLUSIONS This study is the first to report the changes in zinc levels in the blood and tumor tissues of patients with osteosarcoma and to preliminarily verify that zinc inhibits the proliferation and invasion and promote the apoptosis of osteosarcoma cells by inducing the Wnt/β-catenin signaling pathway, which ultimately inhibit cancer growth.
Collapse
Affiliation(s)
- Kai Gao
- Department of Orthopedics, Jining No.1 People's Hospital, Jining, China
| | - Yingchun Zhang
- Department of Interventional Radiology, Jining No.1 People's Hospital, Jining, China
| | - Jianbing Niu
- Department of Orthopedics, Jining No.1 People's Hospital, Jining, China
| | - Zhikui Nie
- Department of Orthopedics, Jining No.1 People's Hospital, Jining, China
| | - Qingsheng Liu
- Department of Orthopedics, Jining No.1 People's Hospital, Jining, China.
| | - Chaoliang Lv
- Department of Orthopedics, Jining No.1 People's Hospital, Jining, China.
| |
Collapse
|
44
|
Zhang Y, Lu P, Liang F, Liufu N, Dong Y, Zheng JC, Xie Z. Cyclophilin D Contributes to Anesthesia Neurotoxicity in the Developing Brain. Front Cell Dev Biol 2020; 7:396. [PMID: 32117955 PMCID: PMC7026027 DOI: 10.3389/fcell.2019.00396] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/30/2019] [Indexed: 11/13/2022] Open
Abstract
Anesthetic sevoflurane induces mitochondrial dysfunction, impairment of neurogenesis, and cognitive impairment in young mice, but the underlying mechanism remains to be determined. Cyclophilin D (CypD) is a modulatory factor for the mitochondrial permeability transition pore (mPTP). We, therefore, set out to evaluate the role of CypD in these sevoflurane-induced changes in vitro and in young mice. Wild-type (WT) and CypD knockout (KO) young (postnatal day 6, 7, and 8) mice received 3% sevoflurane 2 h daily and the neural progenitor cells (NPCs) harvested from the WT or CypD KO mice received 4.1% sevoflurane. We used immunohistochemistry and immunocytochemistry imaging, flow cytometry, Western blot, RT-PCR, co-immunoprecipitation, and Morris Water Maze to assess the interaction of sevoflurane and CypD on mitochondria function, neurogenesis, and cognition in vitro and in WT or CypD KO mice. We demonstrated that the sevoflurane anesthesia induced accumulation of CypD, mitochondrial dysfunction, impairment of neurogenesis, and cognitive impairment in WT mice or NPCs harvested from WT mice, but not in CypD KO mice or NPCs harvested from CypD KO mice. Furthermore, the sevoflurane anesthesia reduced the binding of CypD with Adenine nucleotide translocator, the other component of mPTP. These data suggest that the sevoflurane anesthesia might induce a CypD-dependent mitochondria dysfunction, impairment of neurogenesis, and cognitive impairment in young mice and NPCs.
Collapse
Affiliation(s)
- Yiying Zhang
- Center for Neuroimmunology and Regenerative Therapy, Shanghai Tenth People's Hospital, Anesthesia and Brain Research Institute, Tongji University School of Medicine, Shanghai, China.,Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Pan Lu
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Ning Liufu
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Jialin Charles Zheng
- Center for Neuroimmunology and Regenerative Therapy, Shanghai Tenth People's Hospital, Anesthesia and Brain Research Institute, Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neurosciences, Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| |
Collapse
|
45
|
Kim MS, Bang J, Jeon WK. The Involvement of Canonical Wnt Signaling in Memory Impairment Induced by Chronic Cerebral Hypoperfusion in Mice. Transl Stroke Res 2020; 11:734-746. [DOI: 10.1007/s12975-019-00748-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/17/2019] [Accepted: 10/21/2019] [Indexed: 01/25/2023]
|
46
|
Li Y, Zhu ZY, Lu BW, Huang TT, Zhang YM, Zhou NY, Xuan W, Chen ZA, Wen DX, Yu WF, Li PY. Rosiglitazone ameliorates tissue plasminogen activator-induced brain hemorrhage after stroke. CNS Neurosci Ther 2019; 25:1343-1352. [PMID: 31756041 PMCID: PMC6887660 DOI: 10.1111/cns.13260] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022] Open
Abstract
Objective Delayed thrombolytic therapy with recombinant tissue plasminogen activator (tPA) may exacerbate blood‐brain barrier (BBB) breakdown after ischemic stroke and lead to catastrophic hemorrhagic transformation (HT). Rosiglitazone(RSG), a widely used antidiabetic drug that activates peroxisome proliferator‐activated receptor‐γ (PPAR‐γ), has been shown to protect against cerebral ischemia through promoting poststroke microglial polarization toward the beneficial anti‐inflammatory phenotype. However, whether RSG can alleviate HT after delayed tPA treatment remains unknown. In this study, we sort to examine the role of RSG on tPA‐induced HT after stroke. Methods and results We used the murine suture middle cerebral artery occlusion (MCAO) models of stroke followed by delayed administration of tPA (10 mg/kg, 2 hours after suture occlusion) to investigate the therapeutic potential of RSG against tPA‐induced HT. When RSG(6 mg/kg) was intraperitoneally administered 1 hour before MCAO in tPA‐treated MCAO mice, HT in the ischemic territory was significantly attenuated 1 day after stroke. In the tPA‐treated MCAO mice, we found RSG significantly mitigated BBB disruption and hemorrhage development compared to tPA‐alone‐treated stroke mice. Using flow cytometry and immunostaining, we confirmed that the expression of CD206 was significantly upregulated while the expression of iNOS was down‐regulated in microglia of the RSG‐treated mice. We further found that the expression of Arg‐1 was also upregulated in those tPA and RSG‐treated stroke mice and the protection against tPA‐induced HT and BBB disruption in these mice were abolished in the presence of PPAR‐γ antagonist GW9662 (4 mg/kg, 1 hour before dMCAO through intraperitoneal injection). Conclusions RSG treatment protects against BBB damage and ameliorates HT in delayed tPA‐treated stroke mice by activating PPAR‐γ and favoring microglial polarization toward anti‐inflammatory phenotype.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Yu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Bing-Wei Lu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ting-Ting Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yue-Man Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Na-Ying Zhou
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zeng-Ai Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Da-Xiang Wen
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pei-Ying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
47
|
Wnt-3a alleviates neuroinflammation after ischemic stroke by modulating the responses of microglia/macrophages and astrocytes. Int Immunopharmacol 2019; 75:105760. [DOI: 10.1016/j.intimp.2019.105760] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
|
48
|
Xu D, Hou K, Li F, Chen S, Fang W, Li Y. XQ-1H alleviates cerebral ischemia in mice through inhibition of apoptosis and promotion of neurogenesis in a Wnt/β-catenin signaling dependent way. Life Sci 2019; 235:116844. [PMID: 31499069 DOI: 10.1016/j.lfs.2019.116844] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 11/27/2022]
Abstract
AIMS 10-O-(N,N-dimethylaminoethyl)-ginkgolide B methanesulfonate (XQ-1H), a new derivative of ginkgolide B, has drawn great attention for its potent bioactivities against ischemia-induced injury. The purpose of this study was to further investigate the effect of XQ-1H against acute ischemic stroke by inducing middle cerebral artery occlusion/reperfusion (MCAO/R) injuries in mice. MAIN METHODS Treatment of XQ-1H (78 or 39 mg/kg, i.g., bid) 2 h after MCAO improved motor skills and ameliorated the severity of brain infarction and apoptosis seen in the mice by diminishing pathological changes and the activation of a pro-apoptotic protein Cleaved-Caspase-3, which in turn induced anti-apoptotic Bcl-xL. Through introducing Wnt/β-catenin signaling inhibitor XAV-939, XQ-1H was proven to intensively promoted neurogenesis in the peri-infarct cortex, subventricular area (SVZ) and the dentate gyrus (DG) subgranular area (SGZ) in a Wnt signal dependent way by compromising the activation of GSK3β, which in turn upregulated Wnt1, β-catenin, Neuro D1 and Cyclin D1, most possibly through the activation of PI3K/Akt signaling via the upregulation of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). KEY FINDINGS We conclude that XQ-1H preserved the motor functions, limited apoptosis, and concomitantly promoted neurogenesis-related protein expression by Wnt signaling-dependently compromising GSK3β/Caspase-3 activity and enhancing the expression of Wnt1/β-catenin/Neuro D1/Cyclin D1 and Bcl-xL. SIGNIFICANCE This research may benefit the development of stroke therapeutics targeting neurogenesis through Wnt upregulation by XQ-1H.
Collapse
Affiliation(s)
- Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shijie Chen
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
49
|
Wei ZZ, Chen D, Liu LP, Gu X, Zhong W, Zhang YB, Wang Y, Yu SP, Wei L. Enhanced Neurogenesis and Collaterogenesis by Sodium Danshensu Treatment After Focal Cerebral Ischemia in Mice. Cell Transplant 2019; 27:622-636. [PMID: 29984620 PMCID: PMC7020234 DOI: 10.1177/0963689718771889] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ischemic stroke remains a serious threat to human life. There are limited effective
therapies for the treatment of stroke. We have previously demonstrated that angiogenesis
and neurogenesis in the brain play an important role in functional recovery following
ischemic stroke. Recent studies indicate that increased arteriogenesis and collateral
circulation are determining factors for restoring reperfusion and outcomes of stroke
patients. Danshensu, the Salvia miltiorrhiza root extract, is used in
treatments of various human ischemic events in traditional Chinese medicine. Its
therapeutic mechanism, however, is not well clarified. Due to its proposed effect on
angiogenesis and arteriogenesis, we hypothesized that danshensu could benefit stroke
recovery through stimulating neurogenesis and collaterogenesis in the post-ischemia brain.
Focal ischemic stroke targeting the right sensorimotor cortex was induced in wild-type
C57BL6 mice and transgenic mice expressing green fluorescent protein (GFP) to label smooth
muscle cells of brain arteries. Sodium danshensu (SDS, 700 mg/kg) was administered
intraperitoneally (i.p.) 10 min after stroke and once daily until animals were sacrificed.
To label proliferating cells, 5-bromo-2′-deoxyuridine (BrdU; 50 mg/kg, i.p.) was
administered, starting on day 3 after ischemia and continued once daily until sacrifice.
At 14 days after stroke, SDS significantly increased the expression of vascular
endothelial growth factor (VEGF), stromal-derived factor-1 (SDF-1), brain-derived
neurotrophic factor (BDNF), and endothelial nitric oxide synthase (eNOS) in the
peri-infarct region. SDS-treated animals showed increased number of doublecortin
(DCX)-positive cells. Greater numbers of proliferating endothelial cells and smooth muscle
cells were detected in SDS-treated mice 21 days after stroke in comparison with vehicle
controls. The number of newly formed neurons labeled by NeuN and BrdU antibodies increased
in SDS-treated mice 28 days after stroke. SDS significantly increased the newly formed
arteries and the diameter of collateral arteries, leading to enhanced local cerebral blood
flow recovery after stroke. These results suggest that systemic sodium danshensu treatment
shows significant regenerative effects in the post-ischemic brain, which may benefit
long-term functional recovery from ischemic stroke.
Collapse
Affiliation(s)
- Zheng Zachory Wei
- 1 Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,2 Experimental and Translational Research Center, Beijing Friendship Hospital, Beijing, China.,3 Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dongdong Chen
- 3 Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Li-Ping Liu
- 4 Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaohuan Gu
- 3 Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Weiwei Zhong
- 3 Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yong-Bo Zhang
- 1 Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yongjun Wang
- 4 Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shan Ping Yu
- 3 Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- 1 Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,2 Experimental and Translational Research Center, Beijing Friendship Hospital, Beijing, China.,3 Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA.,5 Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
50
|
Wei ZZ, Zhu YB, Zhang JY, McCrary MR, Wang S, Zhang YB, Yu SP, Wei L. Priming of the Cells: Hypoxic Preconditioning for Stem Cell Therapy. Chin Med J (Engl) 2018; 130:2361-2374. [PMID: 28937044 PMCID: PMC5634089 DOI: 10.4103/0366-6999.215324] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: Stem cell-based therapies are promising in regenerative medicine for protecting and repairing damaged brain tissues after injury or in the context of chronic diseases. Hypoxia can induce physiological and pathological responses. A hypoxic insult might act as a double-edged sword, it induces cell death and brain damage, but on the other hand, sublethal hypoxia can trigger an adaptation response called hypoxic preconditioning or hypoxic tolerance that is of immense importance for the survival of cells and tissues. Data Sources: This review was based on articles published in PubMed databases up to August 16, 2017, with the following keywords: “stem cells,” “hypoxic preconditioning,” “ischemic preconditioning,” and “cell transplantation.” Study Selection: Original articles and critical reviews on the topics were selected. Results: Hypoxic preconditioning has been investigated as a primary endogenous protective mechanism and possible treatment against ischemic injuries. Many cellular and molecular mechanisms underlying the protective effects of hypoxic preconditioning have been identified. Conclusions: In cell transplantation therapy, hypoxic pretreatment of stem cells and neural progenitors markedly increases the survival and regenerative capabilities of these cells in the host environment, leading to enhanced therapeutic effects in various disease models. Regenerative treatments can mobilize endogenous stem cells for neurogenesis and angiogenesis in the adult brain. Furthermore, transplantation of stem cells/neural progenitors achieves therapeutic benefits via cell replacement and/or increased trophic support. Combinatorial approaches of cell-based therapy with additional strategies such as neuroprotective protocols, anti-inflammatory treatment, and rehabilitation therapy can significantly improve therapeutic benefits. In this review, we will discuss the recent progress regarding cell types and applications in regenerative medicine as well as future applications.
Collapse
Affiliation(s)
- Zheng Z Wei
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Yan-Bing Zhu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - James Y Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Myles R McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Song Wang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Yong-Bo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shan-Ping Yu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Ling Wei
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University; Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|