1
|
Li Y, Hao P, Duan H, Hao F, Zhao W, Gao Y, Yang Z, So KF, Li X. Activation of adult endogenous neurogenesis by a hyaluronic acid collagen gel containing basic fibroblast growth factor promotes remodeling and functional recovery of the injured cerebral cortex. Neural Regen Res 2025; 20:2923-2937. [PMID: 39610105 PMCID: PMC11826446 DOI: 10.4103/nrr.nrr-d-23-01706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 02/02/2024] [Accepted: 04/20/2024] [Indexed: 11/30/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00024/figure1/v/2024-11-26T163120Z/r/image-tiff The presence of endogenous neural stem/progenitor cells in the adult mammalian brain suggests that the central nervous system can be repaired and regenerated after injury. However, whether it is possible to stimulate neurogenesis and reconstruct cortical layers II to VI in non-neurogenic regions, such as the cortex, remains unknown. In this study, we implanted a hyaluronic acid collagen gel loaded with basic fibroblast growth factor into the motor cortex immediately following traumatic injury. Our findings reveal that this gel effectively stimulated the proliferation and migration of endogenous neural stem/progenitor cells, as well as their differentiation into mature and functionally integrated neurons. Importantly, these new neurons reconstructed the architecture of cortical layers II to VI, integrated into the existing neural circuitry, and ultimately led to improved brain function. These findings offer novel insight into potential clinical treatments for traumatic cerebral cortex injuries.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Hao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Kwok-Fai So
- Guangdong–HongKong–Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong Province, China
- Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administration Region, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong–HongKong–Macao Greater Bay Area, Guangzhou, Guangdong Province, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
2
|
Tang Z, Li R, Guo X, Wang Z, Wu J. Regulation of blood-brain barrier integrity by brain microvascular endothelial cells in ischemic stroke: A therapeutic opportunity. Eur J Pharmacol 2025; 996:177553. [PMID: 40147580 DOI: 10.1016/j.ejphar.2025.177553] [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: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Stroke is the second leading cause of death from cardiovascular diseases. Brain microvascular endothelial cells (BMECs) are crucial in the treatment of cerebral ischemic stroke, as their functional status directly affects the integrity of the blood-brain barrier (BBB). This review systematically discusses the central role of BMECs in ischemia. The mitochondrial dysfunction and activation of apoptosis/necrosis pathways in BMECs directly disrupt the integrity of the BBB and the degradation of junctional complexes (such as TJs and AJs) further exacerbates its permeability. In the neurovascular unit (NVU), astrocytes, microglia, and pericytes regulate the function of BMECs by secreting cytokines (such as TGF-β and VEGF), showing dual effects of promoting repair and damage. The dynamic changes of transporters, including those from the ATP-binding cassette and solute carrier families, as well as ion channels and exchangers, such as potassium and calcium channels, offer novel insights for the development of targeted drug delivery systems.
Collapse
Affiliation(s)
- Ziqi Tang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| | - Ruoxi Li
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Xi Guo
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China.
| |
Collapse
|
3
|
Yang Q, Lu D, Wu J, Liang F, Wang H, Yang J, Zhang G, Wang C, Yang Y, Zhu L, Sun X. Nanoparticles for the treatment of spinal cord injury. Neural Regen Res 2025; 20:1665-1680. [PMID: 39104097 PMCID: PMC11688544 DOI: 10.4103/nrr.nrr-d-23-01848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/06/2024] [Accepted: 04/09/2024] [Indexed: 08/07/2024] Open
Abstract
Spinal cord injuries lead to significant loss of motor, sensory, and autonomic functions, presenting major challenges in neural regeneration. Achieving effective therapeutic concentrations at injury sites has been a slow process, partly due to the difficulty of delivering drugs effectively. Nanoparticles, with their targeted delivery capabilities, biocompatibility, and enhanced bioavailability over conventional drugs, are garnering attention for spinal cord injury treatment. This review explores the current mechanisms and shortcomings of existing treatments, highlighting the benefits and progress of nanoparticle-based approaches. We detail nanoparticle delivery methods for spinal cord injury, including local and intravenous injections, oral delivery, and biomaterial-assisted implantation, alongside strategies such as drug loading and surface modification. The discussion extends to how nanoparticles aid in reducing oxidative stress, dampening inflammation, fostering neural regeneration, and promoting angiogenesis. We summarize the use of various types of nanoparticles for treating spinal cord injuries, including metallic, polymeric, protein-based, inorganic non-metallic, and lipid nanoparticles. We also discuss the challenges faced, such as biosafety, effectiveness in humans, precise dosage control, standardization of production and characterization, immune responses, and targeted delivery in vivo. Additionally, we explore future directions, such as improving biosafety, standardizing manufacturing and characterization processes, and advancing human trials. Nanoparticles have shown considerable progress in targeted delivery and enhancing treatment efficacy for spinal cord injuries, presenting significant potential for clinical use and drug development.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Di Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jiuping Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Junjie Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ganggang Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinzhi Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
4
|
Samant V, Prabhu A. Exercise, exerkines and exercise mimetic drugs: Molecular mechanisms and therapeutics. Life Sci 2024; 359:123225. [PMID: 39522716 DOI: 10.1016/j.lfs.2024.123225] [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: 06/08/2024] [Revised: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Chronic diseases linked with sedentary lifestyles and poor dietary habits are increasingly common in modern society. Exercise is widely acknowledged to have a plethora of health benefits, including its role in primary prevention of various chronic conditions like type 2 diabetes mellitus, obesity, cardiovascular disease, and several musculoskeletal as well as degenerative disorders. Regular physical activity induces numerous physiological adaptations that contribute to these positive effects, primarily observed in skeletal muscle but also impacting other tissues. There is a growing interest among researchers in developing pharmaceutical interventions that mimic the beneficial effects of exercise for therapeutic applications. Exercise mimetic medications have the potential to be helpful aids in enhancing functional outcomes for patients with metabolic dysfunction, neuromuscular and musculoskeletal disorders. Some of the potential targets for exercise mimetics include pathways involved in metabolism, mitochondrial function, inflammation, and tissue regeneration. The present review aims to provide an exhaustive overview of the current understanding of exercise physiology, the role of exerkines and biomolecular pathways, and the potential applications of exercise mimetic drugs for the treatment of several diseases.
Collapse
Affiliation(s)
- Vedant Samant
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
5
|
Cai C, Gu C, Meng C, He S, Thashi L, Deji D, Zheng Z, Qiu Q. Therapeutic Effects of Metformin on Central Nervous System Diseases: A Focus on Protection of Neurovascular Unit. Pharm Res 2024; 41:1907-1920. [PMID: 39375240 DOI: 10.1007/s11095-024-03777-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024]
Abstract
Metformin is one of the most commonly used oral hypoglycemic drugs in clinical practice, with unique roles in neurodegeneration and vascular lesions. Neurodegeneration and vasculopathy coexist in many diseases and typically affect the neurovascular unit (NVU), a minimal structural and functional unit in the central nervous system. Its components interact with one another and are indispensable for maintaining tissue homeostasis. This review focuses on retinal (diabetic retinopathy, retinitis pigmentosa) and cerebral (ischemic stroke, Alzheimer's disease) diseases to explore the effects of metformin on the NVU. Metformin has a preliminarily confirmed therapeutic effect on the retinal NUV, affecting many of its components, such as photoreceptors (cones and rods), microglia, ganglion, Müller, and vascular endothelial cells. Since it rapidly penetrates the blood-brain barrier (BBB) and accumulates in the brain, metformin also has an extensively studied neuronal protective effect in neuronal diseases. Its mechanism affects various NVU components, including pericytes, astrocytes, microglia, and vascular endothelial cells, mainly serving to protect the BBB. Regulating the inflammatory response in NVU (especially neurons and microglia) may be the main mechanism of metformin in improving central nervous system related diseases. Metformin may be a potential drug for treating diseases associated with NVU deterioration, however, more trials are needed to validate its timing, duration, dose, clinical effects, and side effects.
Collapse
Affiliation(s)
- Chunyang Cai
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, PR China
| | - Chufeng Gu
- Department of Ophthalmology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Chunren Meng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Shuai He
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China
| | - Lhamo Thashi
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China
| | - Draga Deji
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, PR China.
| | - Qinghua Qiu
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China.
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China.
- High Altitude Ocular Disease Research Center of People's Hospital of Shigatse City and Tongren Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
6
|
Xing C, Liu S, Wang L, Ma H, Zhou M, Zhong H, Zhu S, Wu Q, Ning G. Metformin enhances endogenous neural stem cells proliferation, neuronal differentiation, and inhibits ferroptosis through activating AMPK pathway after spinal cord injury. J Transl Med 2024; 22:723. [PMID: 39103875 PMCID: PMC11302024 DOI: 10.1186/s12967-024-05436-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/25/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Inadequate nerve regeneration and an inhibitory local microenvironment are major obstacles to the repair of spinal cord injury (SCI). The activation and differentiation fate regulation of endogenous neural stem cells (NSCs) represent one of the most promising repair approaches. Metformin has been extensively studied for its antioxidative, anti-inflammatory, anti-aging, and autophagy-regulating properties in central nervous system diseases. However, the effects of metformin on endogenous NSCs remains to be elucidated. METHODS The proliferation and differentiation abilities of NSCs were evaluated using CCK-8 assay, EdU/Ki67 staining and immunofluorescence staining. Changes in the expression of key proteins related to ferroptosis in NSCs were detected using Western Blot and immunofluorescence staining. The levels of reactive oxygen species, glutathione and tissue iron were measured using corresponding assay kits. Changes in mitochondrial morphology and membrane potential were observed using transmission electron microscopy and JC-1 fluorescence probe. Locomotor function recovery after SCI in rats was assessed through BBB score, LSS score, CatWalk gait analysis, and electrophysiological testing. The expression of the AMPK pathway was examined using Western Blot. RESULTS Metformin promoted the proliferation and neuronal differentiation of NSCs both in vitro and in vivo. Furthermore, a ferroptosis model of NSCs using erastin treatment was established in vitro, and metformin treatment could reverse the changes in the expression of key ferroptosis-related proteins, increase glutathione synthesis, reduce reactive oxygen species production and improve mitochondrial membrane potential and morphology. Moreover, metformin administration improved locomotor function recovery and histological outcomes following SCI in rats. Notably, all the above beneficial effects of metformin were completely abolished upon addition of compound C, a specific inhibitor of AMP-activated protein kinase (AMPK). CONCLUSION Metformin, driven by canonical AMPK-dependent regulation, promotes proliferation and neuronal differentiation of endogenous NSCs while inhibiting ferroptosis, thereby facilitating recovery of locomotor function following SCI. Our study further elucidates the protective mechanism of metformin in SCI, providing new mechanistic insights for its candidacy as a therapeutic agent for SCI.
Collapse
Affiliation(s)
- Cong Xing
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Song Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Liyue Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Hongpeng Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Mi Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Hao Zhong
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Shibo Zhu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Qiang Wu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.
- International Science and Technology Cooperation Base of Spinal Cord lnjury, Tianjin, China.
- Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China.
| |
Collapse
|
7
|
Su C, Hou Y, Xu J, Xu Z, Zhou M, Ke A, Li H, Xu J, Brendel M, Maasch JRMA, Bai Z, Zhang H, Zhu Y, Cincotta MC, Shi X, Henchcliffe C, Leverenz JB, Cummings J, Okun MS, Bian J, Cheng F, Wang F. Identification of Parkinson's disease PACE subtypes and repurposing treatments through integrative analyses of multimodal data. NPJ Digit Med 2024; 7:184. [PMID: 38982243 PMCID: PMC11233682 DOI: 10.1038/s41746-024-01175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Parkinson's disease (PD) is a serious neurodegenerative disorder marked by significant clinical and progression heterogeneity. This study aimed at addressing heterogeneity of PD through integrative analysis of various data modalities. We analyzed clinical progression data (≥5 years) of individuals with de novo PD using machine learning and deep learning, to characterize individuals' phenotypic progression trajectories for PD subtyping. We discovered three pace subtypes of PD exhibiting distinct progression patterns: the Inching Pace subtype (PD-I) with mild baseline severity and mild progression speed; the Moderate Pace subtype (PD-M) with mild baseline severity but advancing at a moderate progression rate; and the Rapid Pace subtype (PD-R) with the most rapid symptom progression rate. We found cerebrospinal fluid P-tau/α-synuclein ratio and atrophy in certain brain regions as potential markers of these subtypes. Analyses of genetic and transcriptomic profiles with network-based approaches identified molecular modules associated with each subtype. For instance, the PD-R-specific module suggested STAT3, FYN, BECN1, APOA1, NEDD4, and GATA2 as potential driver genes of PD-R. It also suggested neuroinflammation, oxidative stress, metabolism, PI3K/AKT, and angiogenesis pathways as potential drivers for rapid PD progression (i.e., PD-R). Moreover, we identified repurposable drug candidates by targeting these subtype-specific molecular modules using network-based approach and cell line drug-gene signature data. We further estimated their treatment effects using two large-scale real-world patient databases; the real-world evidence we gained highlighted the potential of metformin in ameliorating PD progression. In conclusion, this work helps better understand clinical and pathophysiological complexity of PD progression and accelerate precision medicine.
Collapse
Grants
- R21 AG083003 NIA NIH HHS
- R01 AG082118 NIA NIH HHS
- R56 AG074001 NIA NIH HHS
- R01AG076448 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1AG072449 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- MJFF-023081 Michael J. Fox Foundation for Parkinson's Research (Michael J. Fox Foundation)
- R01AG080991 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P30 AG072959 NIA NIH HHS
- 3R01AG066707-01S1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R21AG083003 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG066707 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R35 AG071476 NIA NIH HHS
- RF1 AG082211 NIA NIH HHS
- R56AG074001 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG082118 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R25 AG083721 NIA NIH HHS
- RF1AG082211 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 NS093334 NINDS NIH HHS
- AG083721-01 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1NS133812 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P20GM109025 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1 NS133812 NINDS NIH HHS
- R35AG71476 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AG073323 NIA NIH HHS
- R01 AG066707 NIA NIH HHS
- R01AG053798 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG076234 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 AG076448 NIA NIH HHS
- R01 AG080991 NIA NIH HHS
- R01 AG076234 NIA NIH HHS
- U01NS093334 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P20 GM109025 NIGMS NIH HHS
- P30AG072959 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1 AG072449 NIA NIH HHS
- R01 AG053798 NIA NIH HHS
- 3R01AG066707-02S1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AG073323 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- ALZDISCOVERY-1051936 Alzheimer's Association
Collapse
Affiliation(s)
- Chang Su
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Yu Hou
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zhenxing Xu
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Manqi Zhou
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Alison Ke
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Haoyang Li
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jie Xu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Matthew Brendel
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jacqueline R M A Maasch
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computer Science, Cornell Tech, Cornell University, New York, NY, USA
| | - Zilong Bai
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Haotan Zhang
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Zhu
- Department of Computer Science, University of Texas at Arlington, Arlington, TX, USA
| | - Molly C Cincotta
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xinghua Shi
- Department of Computer and Information Sciences, Temple University, Philadelphia, PA, USA
| | - Claire Henchcliffe
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Michael S Okun
- Department of Neurology, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
8
|
Islam R, Noman H, Azimi A, Siu R, Chinchalongporn V, Schuurmans C, Morshead CM. Primitive and Definitive Neural Precursor Cells Are Present in Human Cerebral Organoids. Int J Mol Sci 2024; 25:6549. [PMID: 38928255 PMCID: PMC11203442 DOI: 10.3390/ijms25126549] [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: 05/01/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Activation of neural stem cells (NSCs) correlates with improved functional outcomes in mouse models of injury. In the murine brain, NSCs have been extensively characterized and comprise (1) primitive NSCs (pNSCs) and (2) definitive NSCs (dNSCs). pNSCs are the earliest cells in the NSC lineage giving rise to dNSCs in the embryonic and adult mouse brain. pNSCs are quiescent under baseline conditions and can be activated upon injury. Herein, we asked whether human pNSCs and dNSCs can be isolated during the maturation of human cerebral organoids (COs) and activated by drugs known to regulate mouse NSC behavior. We demonstrate that self-renewing, multipotent pNSC and dNSC populations are present in human COs and express genes previously characterized in mouse NSCs. The drug NWL283, an inhibitor of apoptosis, reduced cell death in COs but did not improve NSC survival. Metformin, a drug used to treat type II diabetes that is known to promote NSC activation in mice, was found to expand human NSC pools. Together, these findings are the first to identify and characterize human pNSCs, advancing our understanding of the human NSC lineage and highlighting drugs that enhance their activity.
Collapse
Affiliation(s)
- Rehnuma Islam
- Institute of Medical Science, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
| | - Humna Noman
- Institute of Medical Science, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
| | - Ashkan Azimi
- Department of Surgery, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
| | - Ricky Siu
- Department of Surgery, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
| | | | - Carol Schuurmans
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
| | - Cindi M. Morshead
- Institute of Medical Science, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
- Department of Surgery, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
9
|
Loan A, Syal C, Lui M, He L, Wang J. Promising use of metformin in treating neurological disorders: biomarker-guided therapies. Neural Regen Res 2024; 19:1045-1055. [PMID: 37862207 PMCID: PMC10749596 DOI: 10.4103/1673-5374.385286] [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: 02/09/2023] [Revised: 04/25/2023] [Accepted: 07/29/2023] [Indexed: 10/22/2023] Open
Abstract
Neurological disorders are a diverse group of conditions that affect the nervous system and include neurodegenerative diseases (Alzheimer's disease, multiple sclerosis, Parkinson's disease, Huntington's disease), cerebrovascular conditions (stroke), and neurodevelopmental disorders (autism spectrum disorder). Although they affect millions of individuals around the world, only a limited number of effective treatment options are available today. Since most neurological disorders express mitochondria-related metabolic perturbations, metformin, a biguanide type II antidiabetic drug, has attracted a lot of attention to be repurposed to treat neurological disorders by correcting their perturbed energy metabolism. However, controversial research emerges regarding the beneficial/detrimental effects of metformin on these neurological disorders. Given that most neurological disorders have complex etiology in their pathophysiology and are influenced by various risk factors such as aging, lifestyle, genetics, and environment, it is important to identify perturbed molecular functions that can be targeted by metformin in these neurological disorders. These molecules can then be used as biomarkers to stratify subpopulations of patients who show distinct molecular/pathological properties and can respond to metformin treatment, ultimately developing targeted therapy. In this review, we will discuss mitochondria-related metabolic perturbations and impaired molecular pathways in these neurological disorders and how these can be used as biomarkers to guide metformin-responsive treatment for the targeted therapy to treat neurological disorders.
Collapse
Affiliation(s)
- Allison Loan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON, Canada
| | - Charvi Syal
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Margarita Lui
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Ling He
- Department of Pediatrics and Medicine, Johns Hopkins Medical School, Baltimore, MD, USA
| | - Jing Wang
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
10
|
Pinoșanu EA, Pîrșcoveanu D, Albu CV, Burada E, Pîrvu A, Surugiu R, Sandu RE, Serb AF. Rhoa/ROCK, mTOR and Secretome-Based Treatments for Ischemic Stroke: New Perspectives. Curr Issues Mol Biol 2024; 46:3484-3501. [PMID: 38666949 PMCID: PMC11049286 DOI: 10.3390/cimb46040219] [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: 03/18/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Ischemic stroke triggers a complex cascade of cellular and molecular events leading to neuronal damage and tissue injury. This review explores the potential therapeutic avenues targeting cellular signaling pathways implicated in stroke pathophysiology. Specifically, it focuses on the articles that highlight the roles of RhoA/ROCK and mTOR signaling pathways in ischemic brain injury and their therapeutic implications. The RhoA/ROCK pathway modulates various cellular processes, including cytoskeletal dynamics and inflammation, while mTOR signaling regulates cell growth, proliferation, and autophagy. Preclinical studies have demonstrated the neuroprotective effects of targeting these pathways in stroke models, offering insights into potential treatment strategies. However, challenges such as off-target effects and the need for tissue-specific targeting remain. Furthermore, emerging evidence suggests the therapeutic potential of MSC secretome in stroke treatment, highlighting the importance of exploring alternative approaches. Future research directions include elucidating the precise mechanisms of action, optimizing treatment protocols, and translating preclinical findings into clinical practice for improved stroke outcomes.
Collapse
Affiliation(s)
- Elena Anca Pinoșanu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
- Doctoral School, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania
| | - Denisa Pîrșcoveanu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
| | - Emilia Burada
- Department of Physiology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania;
| | - Andrei Pîrvu
- Dolj County Regional Centre of Medical Genetics, Clinical Emergency County Hospital Craiova, St. Tabaci, No. 1, 200642 Craiova, Romania;
| | - Roxana Surugiu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania;
| | - Raluca Elena Sandu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania; (E.A.P.); (D.P.); (C.V.A.)
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, St. Petru Rares, No. 2-4, 200433 Craiova, Romania;
| | - Alina Florina Serb
- Department of Biochemistry and Pharmacology, Biochemistry Discipline, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq., No. 2, 300041 Timisoara, Romania;
| |
Collapse
|
11
|
Bakhashab S, O’Neill J, Barber R, Arden C, Weaver JU. Upregulation of Anti-Angiogenic miR-106b-3p Correlates Negatively with IGF-1 and Vascular Health Parameters in a Model of Subclinical Cardiovascular Disease: Study with Metformin Therapy. Biomedicines 2024; 12:171. [PMID: 38255276 PMCID: PMC10813602 DOI: 10.3390/biomedicines12010171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Well-controlled type 1 diabetes mellitus (T1DM) is regarded as a model of subclinical cardiovascular disease (CVD), characterized by inflammation and adverse vascular health. However, the underlying mechanisms are not fully understood. We investigated insulin-like growth factor-1 (IGF-1) and IGF-binding protein-3 (IGFBP-3) levels, their correlation to miR-106b-3p expression in a subclinical CVD model, and the cardioprotective effect of metformin. A total of 20 controls and 29 well-controlled T1DM subjects were studied. Plasma IGF-1, IGFBP-3 levels, and miR-106b-3p expression in colony-forming unit-Hills were analyzed and compared with vascular markers. miR-106b-3p was upregulated in T1DM (p < 0.05) and negatively correlated with pro-angiogenic markers CD34+/100-lymphocytes (p < 0.05) and IGF-1 (p < 0.05). IGF-1 was downregulated in T1DM (p < 0.01), which was associated with increased inflammatory markers TNF-α, CRP, and IL-10 and reduced CD34+/100-lymphocytes. IGFBP-3 had no significant results. Metformin had no effect on IGF-1 but significantly reduced miR-106b-3p (p < 0.0001). An Ingenuity Pathway analysis predicted miR-106b-3p to inhibit PDGFA, PIK3CG, GDNF, and ADAMTS13, which activated CVD. Metformin was predicted to be cardioprotective by inhibiting miR-106b-3p. In conclusion: Subclinical CVD is characterized by a cardio-adverse profile of low IGF-1 and upregulated miR-106b-3p. We demonstrated that the cardioprotective effect of metformin may be via downregulation of upregulated miR-106b-3p and its effect on downstream targets.
Collapse
Affiliation(s)
- Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia;
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| | - Josie O’Neill
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Rosie Barber
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Catherine Arden
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Jolanta U. Weaver
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (J.O.); (R.B.)
- Department of Diabetes, Queen Elizabeth Hospital, Newcastle upon Tyne NE9 6SH, UK
- Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
12
|
Ruan Z, Yin H, Wan TF, Lin ZR, Zhao SS, Long HT, Long C, Li ZH, Liu YQ, Luo H, Cheng L, Chen C, Zeng M, Lin ZY, Zhao RB, Chen CY, Wang ZX, Liu ZZ, Cao J, Wang YY, Jin L, Liu YW, Zhu GQ, Zou JT, Gong JS, Luo Y, Hu Y, Zhu Y, Xie H. Metformin accelerates bone fracture healing by promoting type H vessel formation through inhibition of YAP1/TAZ expression. Bone Res 2023; 11:45. [PMID: 37587136 PMCID: PMC10432554 DOI: 10.1038/s41413-023-00279-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 06/04/2023] [Accepted: 06/26/2023] [Indexed: 08/18/2023] Open
Abstract
Due to increasing morbidity worldwide, fractures are becoming an emerging public health concern. This study aimed to investigate the effect of metformin on the healing of osteoporotic as well as normal fractures. Type H vessels have recently been identified as a bone-specific vascular subtype that supports osteogenesis. Here, we show that metformin accelerated fracture healing in both osteoporotic and normal mice. Moreover, metformin promoted angiogenesis in vitro under hypoxia as well as type H vessel formation throughout fracture healing. Mechanistically, metformin increased the expression of HIF-1α, an important positive regulator of type H vessel formation, by inhibiting the expression of YAP1/TAZ in calluses and hypoxia-cultured human microvascular endothelial cells (HMECs). The results of HIF-1α or YAP1/TAZ interference in hypoxia-cultured HMECs using siRNA further suggested that the enhancement of HIF-1α and its target genes by metformin is primarily through YAP1/TAZ inhibition. Finally, overexpression of YAP1/TAZ partially counteracted the effect of metformin in promoting type H vessel-induced angiogenesis-osteogenesis coupling during fracture repair. In summary, our findings suggest that metformin has the potential to be a therapeutic agent for fractures by promoting type H vessel formation through YAP1/TAZ inhibition.
Collapse
Affiliation(s)
- Zhe Ruan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hao Yin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Teng-Fei Wan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Zhi-Rou Lin
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shu-Shan Zhao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hai-Tao Long
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Cheng Long
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhao-Hui Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yu-Qi Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hao Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Liang Cheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Can Chen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Min Zeng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhang-Yuan Lin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Rui-Bo Zhao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chun-Yuan Chen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhen-Xing Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zheng-Zhao Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jia Cao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yi-Yi Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Ling Jin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Yi-Wei Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Guo-Qiang Zhu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Jing-Tao Zou
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Jiang-Shan Gong
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Yi Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China
| | - Yin Hu
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yong Zhu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hui Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Angmedicine, Changsha, Hunan, 410008, China.
- Angmedicine Research Center of Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
13
|
Paro MR, Chakraborty AR, Angelo S, Nambiar S, Bulsara KR, Verma R. Molecular mediators of angiogenesis and neurogenesis after ischemic stroke. Rev Neurosci 2023; 34:425-442. [PMID: 36073599 PMCID: PMC12051358 DOI: 10.1515/revneuro-2022-0049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/22/2022] [Indexed: 11/15/2022]
Abstract
The mechanisms governing neurological and functional recovery after ischemic stroke are incompletely understood. Recent advances in knowledge of intrinsic repair processes of the CNS have so far translated into minimal improvement in outcomes for stroke victims. Better understanding of the processes underlying neurological recovery after stroke is necessary for development of novel therapeutic approaches. Angiogenesis and neurogenesis have emerged as central mechanisms of post-stroke recovery and potential targets for therapeutics. Frameworks have been developed for conceptualizing cerebral angiogenesis and neurogenesis at the tissue and cellular levels. These models highlight that angiogenesis and neurogenesis are linked to each other and to functional recovery. However, knowledge of the molecular framework linking angiogenesis and neurogenesis after stroke is limited. Studies of potential therapeutics typically focus on one mediator or pathway with minimal discussion of its role within these multifaceted biochemical processes. In this article, we briefly review the current understanding of the coupled processes of angiogenesis and neurogenesis after stroke. We then identify the molecular mediators and signaling pathways found in pre-clinical studies to upregulate both processes after stroke and contextualizes them within the current framework. This report thus contributes to a more-unified understanding of the molecular mediators governing angiogenesis and neurogenesis after stroke, which we hope will help guide the development of novel therapeutic approaches for stroke survivors.
Collapse
Affiliation(s)
- Mitch R. Paro
- UConn School of Medicine, 200 Academic Way, Farmington, Connecticut 06032, United States of America
- Department of Neuroscience, UConn School of Medicine, 263 Farmington Avenue, Farmington, Connecticut 06032, United States of America
| | - Arijit R. Chakraborty
- UConn School of Medicine, 200 Academic Way, Farmington, Connecticut 06032, United States of America
| | - Sophia Angelo
- UConn School of Medicine, 200 Academic Way, Farmington, Connecticut 06032, United States of America
| | - Shyam Nambiar
- University of Connecticut, 75 North Eagleville Rd, Storrs, Connecticut 06269, United States of America
| | - Ketan R. Bulsara
- UConn School of Medicine, 200 Academic Way, Farmington, Connecticut 06032, United States of America
- Division of Neurosurgery, UConn Health, 135 Dowling Way, Farmington, Connecticut 06030, United States of America
| | - Rajkumar Verma
- UConn School of Medicine, 200 Academic Way, Farmington, Connecticut 06032, United States of America
- Department of Neuroscience, UConn School of Medicine, 263 Farmington Avenue, Farmington, Connecticut 06032, United States of America
| |
Collapse
|
14
|
Metformin enhances neural precursor cells migration and functional recovery after ischemic stroke in mice. Exp Brain Res 2023; 241:505-515. [PMID: 36611122 DOI: 10.1007/s00221-023-06547-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Resident neural precursor cells (NPCs) activation is a promising therapeutic strategy for brain repair. This strategy involves stimulating multiple stages of NPCs development, including proliferation, self-renewal, migration, and differentiation. Metformin, an FDA-approved diabetes drug, has been shown to promote the proliferation and differentiation of NPCs. However, it is still unclear whether metformin promotes the migration of NPCs. EVOS living cell imaging system was used for observing the migration for primary NPCs dynamically in vitro after metformin treatment. For in vivo study, a mouse model of ischemic stroke was established through middle cerebral artery occlusion (MCAO). To label the proliferating cell in subventricular zone, BrdU was injected intraperitoneally into the mice. After co-staining with BrdU and doublecortin (DCX), a marker for NPCs, the migration of Brdu and DCX double positive NPCs was detected along the rostral migratory stream (RMS) and around the infarct area using frozen brain sections. Finally, the rotarod test, corner test and beam walking were performed to evaluate the motor functions of the mice after stroke in different groups. The results showed that metformin enhanced NPCs migration in vivo and in vitro by promoting F-actin assembly and lamellipodia formation. What's more, metformin treatment also significantly reduced the infarct volume and alleviated functional dysfunction after stroke. Mechanistically, metformin promoted NPCs migration via up-regulating the CDC42 expression. Taken together, metformin represents an optimal candidate agent for neural repair that is capable of not only expanding the adult NPC population but also subsequently driving them toward the destination for neuronal differentiation.
Collapse
|
15
|
Molecular mechanisms of exercise contributing to tissue regeneration. Signal Transduct Target Ther 2022; 7:383. [PMID: 36446784 PMCID: PMC9709153 DOI: 10.1038/s41392-022-01233-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Physical activity has been known as an essential element to promote human health for centuries. Thus, exercise intervention is encouraged to battle against sedentary lifestyle. Recent rapid advances in molecular biotechnology have demonstrated that both endurance and resistance exercise training, two traditional types of exercise, trigger a series of physiological responses, unraveling the mechanisms of exercise regulating on the human body. Therefore, exercise has been expected as a candidate approach of alleviating a wide range of diseases, such as metabolic diseases, neurodegenerative disorders, tumors, and cardiovascular diseases. In particular, the capacity of exercise to promote tissue regeneration has attracted the attention of many researchers in recent decades. Since most adult human organs have a weak regenerative capacity, it is currently a key challenge in regenerative medicine to improve the efficiency of tissue regeneration. As research progresses, exercise-induced tissue regeneration seems to provide a novel approach for fighting against injury or senescence, establishing strong theoretical basis for more and more "exercise mimetics." These drugs are acting as the pharmaceutical alternatives of those individuals who cannot experience the benefits of exercise. Here, we comprehensively provide a description of the benefits of exercise on tissue regeneration in diverse organs, mainly focusing on musculoskeletal system, cardiovascular system, and nervous system. We also discuss the underlying molecular mechanisms associated with the regenerative effects of exercise and emerging therapeutic exercise mimetics for regeneration, as well as the associated opportunities and challenges. We aim to describe an integrated perspective on the current advances of distinct physiological mechanisms associated with exercise-induced tissue regeneration on various organs and facilitate the development of drugs that mimics the benefits of exercise.
Collapse
|
16
|
Fu W, Tang Y, Che X, Tan J, Ma Y, He Z. Neuropharmacological efficacy of metformin for stroke in rodents: A meta-analysis of preclinical trials. Front Pharmacol 2022; 13:1009169. [PMID: 36408248 PMCID: PMC9669075 DOI: 10.3389/fphar.2022.1009169] [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: 08/01/2022] [Accepted: 10/18/2022] [Indexed: 09/28/2023] Open
Abstract
Background: Stroke, including ischemic stroke, intracerebral hemorrhage, and subarachnoid hemorrhage (SAH), remains a leading cause of mortality globally. Different stroke subtypes have similar detrimental effects in multiple fields of health. Previous research has shown that metformin plays a neuroprotective role in experimental animal models of stroke; however, a preclinical quantitative analysis on the ability of metformin to treat stroke is still lacking. This meta-analysis evaluates the efficacy of metformin in improving stroke prognosis in rodent models of stroke. Methods: Relevant preclinical trials were retrieved from PubMed, EMBASE, and the Web of Science. The neurological score (NS), brain water content (BWC), infarct size, rotarod test, TUNEL, neuron quantity, microglia quantity, and p-AMPK levels were compared between a control group and a metformin group using the standardized mean difference (SMD) and corresponding confidence interval (CI). Quality was assessed with SYRCLE's risk of bias tool. Results: Fifteen articles published from 2010 to 2022 were included in the meta-analysis. The metformin group had statistically significant differences compared to the control group in the following aspects: NS (SMD -1.45; 95% CI -2.32, -0.58; p = 0.001), BWC (SMD -3.22; 95% CI -4.69, -1.76; p < 0.0001), infarct size (SMD -2.90; 95% CI -3.95, -1.85; p < 0.00001), rotarod test (SMD 2.55; 95% CI 1.87, 3.23; p < 0.00001), TUNEL (SMD -3.63; 95% CI -5.77, -1.48; p = 0.0009), neuron quantity (SMD 3.42; 95% CI 2.51, 4.34; p < 0.00001), microglia quantity (SMD -3.06; 95% CI -4.69, -1.44; p = 0.0002), and p-AMPK levels (SMD 2.92; 95% CI 2.02, 3.82; p < 0.00001). Furthermore, sensitivity analysis and stratified analysis were conducted for heterogeneous outcome indicators. Conclusion: Overall, metformin treatment improves severe outcomes triggered by stroke. Despite the limitations intrinsic to animal studies, this systematic review may provide a vital reference for future high-quality preclinical trials and clinical use.
Collapse
Affiliation(s)
- Wenqiao Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yin Tang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xudong Che
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiahe Tan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yinrui Ma
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaohui He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Zhang G, Chen S, Jia J, Liu C, Wang W, Zhang H, Zhen X. Development and Evaluation of Novel Metformin Derivative Metformin Threonate for Brain Ischemia Treatment. Front Pharmacol 2022; 13:879690. [PMID: 35800435 PMCID: PMC9253272 DOI: 10.3389/fphar.2022.879690] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/11/2022] [Indexed: 11/25/2022] Open
Abstract
Epidemiologic data reveal that diabetes patients taking metformin exhibit lower incidence of stroke and better functional outcomes during post-stroke neurologic recovery. We previously demonstrated that chronic post-ischemic administration of metformin improved functional recovery in experimental cerebral ischemia. However, few beneficial effects of metformin on the acute phase of cerebral ischemia were reported either in experimental animals or in stroke patients, which limits the application of metformin in stroke. We hypothesized that slow cellular uptake of metformin hydrochloride may contribute to the lack of efficacy in acute stroke. We recently developed and patented a novel metformin derivative, metformin threonate (SHY-01). Pharmacokinetic profile in vivo and in cultured cells revealed that metformin is more rapidly uptaken and accumulated from SHY-01 than metformin hydrochloride. Accordingly, SHY-01 treatment exhibited more potent and rapid activation of AMP-activated protein kinase (AMPK). Furthermore, SHY-01 elicited a stronger inhibition of microglia activation and more potent neuroprotection when compared to metformin hydrochloride. SHY-01 administration also had superior beneficial effects on neurologic functional recovery in experimental stroke and offered strong protection against acute cerebral ischemia with reduced infarct volume and mortality, as well as the improved sensorimotor and cognitive functions in rats. Collectively, these results indicated that SHY-01 had an improved pharmacokinetic and pharmacological profile and produced more potent protective effects on acute stroke and long-term neurological damage. We propose that SHY-01 is a very promising therapeutic candidate for cerebral ischemic stroke.
Collapse
Affiliation(s)
- Gufang Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- *Correspondence: Xuechu Zhen, ; Gufang Zhang,
| | - Shuangshuang Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jia Jia
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chun Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Weipeng Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Hongjian Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- *Correspondence: Xuechu Zhen, ; Gufang Zhang,
| |
Collapse
|
18
|
Liu C, Zhang D, Lu Z, Man J, Zhang Z, Fu X, Cui K, Wang J. Metformin protects against pericyte apoptosis and promotes neurogenesis through suppressing JNK p38 MAPK signalling activation in ischemia/reperfusion injury. Neurosci Lett 2022; 783:136708. [PMID: 35660649 DOI: 10.1016/j.neulet.2022.136708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/12/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
Abstract
Metformin (MET) has been the subject of many classic studies in possessing antiapoptotic, anti-inflammatory, antioxidation activities and antiviral. Recently investigators have examined the anti-apoptosis effects of MET in acute myocardial infarction and Intracerebral hemorrhage, but very little is currently known about how it regulates ischemic stroke-induced pericytes apoptosis and neural stem cells (NSCs) proliferation. The present research explored the potential neuroprotective mechanisms of MET using transient middle cerebral artery occlusion(tMCAO) mice. The experimental work presented that tMCAO mice treated by metformin had better neurologic outcomes on days 1, 3, and 7 after operation, and alleviated blood-brain barrier (BBB) destruction, brain water content and infarct volume on 72 h after surgery. The data showed that MET alleviated BBB disruption by reducing PDGFRβ/ matrix metalloproteinase-9 (MMP9) positive cells, relieving zonula occludens-1 (ZO-1) drop away and increasing pericyte coverage through remarkably reducing the percentage of PDGFRβ/caspase-3 positive cells. In addition, MET induced antiapoptotic activity followed by downregulating cleaved caspase-3 and Bax expression. Moreover, JNK signaling pathway has been proved to be pivotal in mediating apoptosis in cerebral ischemia/reperfusion (I/R) injury. The results of this research illustrated that MET treatment downregulated the levels of phosphorylated JNK and P38 in vivo, however the use of JNK activator anisomycin (ANI) could reverse the neuroprotection effect of MET, demonstrating that the JNK pathway is associated with the anti-apoptosis mechanisms of MET. Finally, metformin remarkably increased the percentage of BrdU/DCX-positive cells in subventricular zone (SVZ) and up-regulated BDNF、VEGF and NGF expression after ischemia/reperfusion(I/R) injury on day 7. Our data illustrated that metformin provides an effective therapy for I/R injury.
Collapse
Affiliation(s)
- Chang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China
| | - Di Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China
| | - Zhengfang Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China
| | - Jiang Man
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China
| | - Zhen Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China
| | - Xiaojuan Fu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China
| | - Kefei Cui
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China
| | - Jianping Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan Province, China.
| |
Collapse
|
19
|
Al Dahhan NZ, Cox E, Nieman BJ, Mabbott DJ. Cross-translational models of late-onset cognitive sequelae and their treatment in pediatric brain tumor survivors. Neuron 2022; 110:2215-2241. [PMID: 35523175 DOI: 10.1016/j.neuron.2022.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/21/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Pediatric brain tumor treatments have a high success rate, but survivors are at risk of cognitive sequelae that impact long-term quality of life. We summarize recent clinical and animal model research addressing pathogenesis or evaluating candidate interventions for treatment-induced cognitive sequelae. Assayed interventions encompass a broad range of approaches, including modifications to radiotherapy, modulation of immune response, prevention of treatment-induced cell loss or promotion of cell renewal, manipulation of neuronal signaling, and lifestyle/environmental adjustments. We further emphasize the potential of neuroimaging as a key component of cross-translation to contextualize laboratory research within broader clinical findings. This cross-translational approach has the potential to accelerate discovery to improve pediatric cancer survivors' long-term quality of life.
Collapse
Affiliation(s)
- Noor Z Al Dahhan
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Elizabeth Cox
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Brian J Nieman
- Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada; Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Donald J Mabbott
- Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
20
|
Li T, Jing P, Yang L, Wan Y, Du X, Wei J, Zhou M, Liu Z, Lin Y, Zhong Z. CAQK modification enhances the targeted accumulation of metformin-loaded nanoparticles in rats with spinal cord injury. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 41:102526. [PMID: 35104674 DOI: 10.1016/j.nano.2022.102526] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) often causes neuronal membrane rupture and immediate death of neurons, followed by complicated secondary injuries. Treatment of SCI still remains a major challenge in clinical practice; thus, a great advance is urgently needed in this field. Metformin (MET) has anti-oxidant, anti-inflammatory, anti-apoptotic and neuroprotective properties, which may exert a potential therapeutic effect on SCI. In this study, we established a zein-based MET-loaded nanodrug system (CAQK-MET-NPs) for the targeted drug delivery for SCI. The results showed that MET could be effectively encapsulated into zein to obtain the zein-based spherical nanoparticles. Pharmacokinetic analysis indicated that CAQK-MET-NPs exhibited sustained-release and long-term therapeutic effects. The fluorescence imaging and tissue distribution experiments showed that CAQK-MET-NPs could efficiently accumulate at the lesion site of SCI rats. In conclusion, CAQK-MET-NPs may be a promising nanodrug for the treatment of SCI.
Collapse
Affiliation(s)
- Ting Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; The Second Hospital of Traditional Chinese Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Pei Jing
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lingling Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yujie Wan
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xingjie Du
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Wei
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Meiling Zhou
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhongbing Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yan Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
21
|
Regulating Endogenous Neural Stem Cell Activation to Promote Spinal Cord Injury Repair. Cells 2022; 11:cells11050846. [PMID: 35269466 PMCID: PMC8909806 DOI: 10.3390/cells11050846] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) affects millions of individuals worldwide. Currently, there is no cure, and treatment options to promote neural recovery are limited. An innovative approach to improve outcomes following SCI involves the recruitment of endogenous populations of neural stem cells (NSCs). NSCs can be isolated from the neuroaxis of the central nervous system (CNS), with brain and spinal cord populations sharing common characteristics (as well as regionally distinct phenotypes). Within the spinal cord, a number of NSC sub-populations have been identified which display unique protein expression profiles and proliferation kinetics. Collectively, the potential for NSCs to impact regenerative medicine strategies hinges on their cardinal properties, including self-renewal and multipotency (the ability to generate de novo neurons, astrocytes, and oligodendrocytes). Accordingly, endogenous NSCs could be harnessed to replace lost cells and promote structural repair following SCI. While studies exploring the efficacy of this approach continue to suggest its potential, many questions remain including those related to heterogeneity within the NSC pool, the interaction of NSCs with their environment, and the identification of factors that can enhance their response. We discuss the current state of knowledge regarding populations of endogenous spinal cord NSCs, their niche, and the factors that regulate their behavior. In an attempt to move towards the goal of enhancing neural repair, we highlight approaches that promote NSC activation following injury including the modulation of the microenvironment and parenchymal cells, pharmaceuticals, and applied electrical stimulation.
Collapse
|
22
|
Qiu-Yue X, Tian-Yuan Y, Xiao-Long W, Dong-Mei Q, Xiao-Rui C. Effects of Metformin on Modulating the Expression of Brain-related Genes of APP/PS1 Transgenic Mice based on Single Cell Sequencing. Curr Alzheimer Res 2022; 19:754-771. [PMID: 36464874 DOI: 10.2174/1567205020666221201143323] [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: 08/02/2022] [Revised: 10/26/2022] [Accepted: 11/12/2022] [Indexed: 12/07/2022]
Abstract
BACKGROUND Alzheimer's disease is the most common form of dementia, affecting millions of people worldwide. METHODS Here, we analyzed the effects of metformin on APP/PS1 transgenic mice by behavioral test and single-cell sequencing. RESULTS It showed that metformin can improve the spatial learning, memory function, and anxiety mood of APP/PS1 transgenic mice. We identified transcriptionally distinct subpopulations of nine major brain cell types. Metformin increased the differentiation of stem cells, decreased the proportion of cells in the G2 phase, enhanced the generation of neural stem cells and oligodendrocyte progenitor cells, and the tendency of neural stem cells to differentiate into astrocytes. Notably, 253 genes expressed abnormally in APP/PS1 transgenic mice and were reversed by metformin. Ttr, Uba52, and Rps21 are the top 3 genes in the cell-gene network with the highest node degree. Moreover, histochemistry showed the expressions of RPS15, Uba52, and RPL23a were consistent with the data from single-cell sequencing. Pathway and biological process enrichment analysis indicated metformin was involved in nervous system development and negative regulation of the apoptotic process. CONCLUSION Overall, metformin might play an important role in the differentiation and development and apoptotic process of the central nervous system by regulating the expression of Ttr, Uba52, Rps21, and other genes to improve cognition of APP/PS1 transgenic mice. These results provided a clue for elaborating on the molecular and cellular basis of metformin on AD.
Collapse
Affiliation(s)
- Xiao Qiu-Yue
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ye Tian-Yuan
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wang Xiao-Long
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qi Dong-Mei
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Cheng Xiao-Rui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
23
|
Xiang X, Zhou L, Lin Z, Qu X, Chen Y, Xia H. Metformin regulates macrophage polarization via the Shh signaling pathway to improve pulmonary vascular development in bronchopulmonary dysplasia. IUBMB Life 2021; 74:259-271. [PMID: 34910358 DOI: 10.1002/iub.2588] [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: 07/26/2021] [Accepted: 12/06/2021] [Indexed: 12/19/2022]
Abstract
Metformin has potential anti-inflammatory properties and accelerates wound healing by enhancing vascular development. In this study, we aimed to investigate the effects of metformin on pulmonary vascular development and the underlying mechanism. Newborn mice were subcutaneously injected with metformin from day 2 after exposure to hyperoxia. Pulmonary vascular development, inflammation, and Shh signaling pathway-related protein expression were evaluated by western blotting and immunofluorescence staining. M2 macrophage polarization was measured by flow cytometry. The effect of metformin on macrophage polarization was determined using RAW264.7 macrophages exposed to 90% oxygen in vitro. The role of metformin and purmorphamine on M1 and M2 polarization was observed by flow cytometry. M2 polarization of pulmonary macrophages was inhibited after hyperoxic exposure, and metformin increased the number of M2 macrophages in the lung on postnatal day 14. Metformin upregulated CD31 expression and suppressed inflammation in the lung of mice exposed to hyperoxia on postnatal days 7 and 14. Metformin downregulated the Gli1 expression in macrophages in the lung after exposure to hyperoxia on postnatal day 14. In vitro studies showed that metformin inhibited the Gli1 expression in RAW264.7 macrophages exposed to 90% oxygen, which was reversed after purmorphamine pretreatment. Exposure to 90% oxygen inhibited the polarization of M2 macrophages, whereas metformin increased the number of M2 macrophages. Purmorphamine reversed the effects of metformin on M2 polarization and vascular endothelial growth factor (VEGF) upregulation in RAW264.7 macrophages exposed to hyperoxia. In conclusion, metformin regulates macrophage polarization via the Shh signaling pathway to improve pulmonary vascular development in bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Xiaowen Xiang
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhou
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwei Lin
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Qu
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanru Chen
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Xia
- Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Ji Z, Liu GH, Qu J. Mitochondrial sirtuins, metabolism, and aging. J Genet Genomics 2021; 49:287-298. [PMID: 34856390 DOI: 10.1016/j.jgg.2021.11.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Maintaining metabolic homeostasis is essential for cellular and organismal health throughout life. Of the multiple signaling pathways that regulate metabolism, such as PI3K/AKT, mTOR, AMPK, and sirtuins, mammalian sirtuins also play unique roles in aging. By understanding how sirtuins regulate metabolic processes, we can start to understand how they slow down or accelerate biological aging. Here, we review the biology of SIRT3, SIRT4, and SIRT5, known as the mitochondrial sirtuins due to their localization in the mitochondrial matrix. First, we will focus on canonical pathways that regulate metabolism more broadly and how these are integrated with aging regulation. Then, we will summarize the current knowledge about functional differences between SIRT3, SIRT4, and SIRT5 in metabolic control and integration in signaling networks. Finally, we will discuss how mitochondrial sirtuins regulate processes associated with aging and oxidative stress, calorie restriction and disease.
Collapse
Affiliation(s)
- Zhejun Ji
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Guang-Hui Liu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
25
|
Exercise mimetics: harnessing the therapeutic effects of physical activity. Nat Rev Drug Discov 2021; 20:862-879. [PMID: 34103713 DOI: 10.1038/s41573-021-00217-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/05/2023]
Abstract
Exercise mimetics are a proposed class of therapeutics that specifically mimic or enhance the therapeutic effects of exercise. Increased physical activity has demonstrated positive effects in preventing and ameliorating a wide range of diseases, including brain disorders such as Alzheimer disease and dementia, cancer, diabetes and cardiovascular disease. This article discusses the molecular mechanisms and signalling pathways associated with the beneficial effects of physical activity, focusing on effects on brain function and cognitive enhancement. Emerging therapeutic targets and strategies for the development of exercise mimetics, particularly in the field of central nervous system disorders, as well as the associated opportunities and challenges, are discussed.
Collapse
|
26
|
Poor SR, Ettcheto M, Cano A, Sanchez-Lopez E, Manzine PR, Olloquequi J, Camins A, Javan M. Metformin a Potential Pharmacological Strategy in Late Onset Alzheimer's Disease Treatment. Pharmaceuticals (Basel) 2021; 14:ph14090890. [PMID: 34577590 PMCID: PMC8465337 DOI: 10.3390/ph14090890] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 08/28/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most devastating brain disorders. Currently, there are no effective treatments to stop the disease progression and it is becoming a major public health concern. Several risk factors are involved in the progression of AD, modifying neuronal circuits and brain cognition, and eventually leading to neuronal death. Among them, obesity and type 2 diabetes mellitus (T2DM) have attracted increasing attention, since brain insulin resistance can contribute to neurodegeneration. Consequently, AD has been referred to "type 3 diabetes" and antidiabetic medications such as intranasal insulin, glitazones, metformin or liraglutide are being tested as possible alternatives. Metformin, a first line antihyperglycemic medication, is a 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPK) activator hypothesized to act as a geroprotective agent. However, studies on its association with age-related cognitive decline have shown controversial results with positive and negative findings. In spite of this, metformin shows positive benefits such as anti-inflammatory effects, accelerated neurogenesis, strengthened memory, and prolonged life expectancy. Moreover, it has been recently demonstrated that metformin enhances synaptophysin, sirtuin-1, AMPK, and brain-derived neuronal factor (BDNF) immunoreactivity, which are essential markers of plasticity. The present review discusses the numerous studies which have explored (1) the neuropathological hallmarks of AD, (2) association of type 2 diabetes with AD, and (3) the potential therapeutic effects of metformin on AD and preclinical models.
Collapse
Affiliation(s)
- Saghar Rabiei Poor
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran;
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran 14117-13116, Iran
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (M.E.); (P.R.M.)
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (M.E.); (P.R.M.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 08028 Madrid, Spain; (A.C.); (E.S.-L.)
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 08028 Madrid, Spain; (A.C.); (E.S.-L.)
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), 08028 Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Elena Sanchez-Lopez
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 08028 Madrid, Spain; (A.C.); (E.S.-L.)
- Institute of Nanoscience and Nanotechnology (IN2UB), 08028 Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Patricia Regina Manzine
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (M.E.); (P.R.M.)
- Department of Gerontology, Federal University of São Carlos (UFSCar), São Carlos 13565-905, Brazil
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca 3467987, Chile;
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (M.E.); (P.R.M.)
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 08028 Madrid, Spain; (A.C.); (E.S.-L.)
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08028 Barcelona, Spain
- Laboratory of Cellular and Molecular Pathology, Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca 3467987, Chile;
- Correspondence: (A.C.); (M.J.)
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran;
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran 14117-13116, Iran
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 14117-13116, Iran
- Correspondence: (A.C.); (M.J.)
| |
Collapse
|
27
|
Yuen N, Szulc-Lerch KU, Li YQ, Morshead CM, Mabbott DJ, Wong CS, Nieman BJ. Metformin effects on brain development following cranial irradiation in a mouse model. Neuro Oncol 2021; 23:1523-1536. [PMID: 34042964 PMCID: PMC8408860 DOI: 10.1093/neuonc/noab131] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Cranial radiation therapy (CRT) is a mainstay of treatment for malignant pediatric brain tumors and high-risk leukemia. Although CRT improves survival, it has been shown to disrupt normal brain development and result in cognitive impairments in cancer survivors. Animal studies suggest that there is potential to promote brain recovery after injury using metformin. Our aim was to evaluate whether metformin can restore brain volume outcomes in a mouse model of CRT. METHODS C57BL/6J mice were irradiated with a whole-brain radiation dose of 7 Gy during infancy. Two weeks of metformin treatment started either on the day of or 3 days after irradiation. In vivo magnetic resonance imaging was performed prior to irradiation and at 3 subsequent time points to evaluate the effects of radiation and metformin on brain development. RESULTS Widespread volume loss in the irradiated brain appeared within 1 week of irradiation with limited subsequent recovery in volume outcomes. In many structures, metformin administration starting on the day of irradiation exacerbated radiation-induced injury, particularly in male mice. Metformin treatment starting 3 days after irradiation improved brain volume outcomes in subcortical regions, the olfactory bulbs, and structures of the brainstem and cerebellum. CONCLUSIONS Our results show that metformin treatment has the potential to improve neuroanatomical outcomes after CRT. However, both timing of metformin administration and subject sex affect structure outcomes, and metformin may also be deleterious. Our results highlight important considerations in determining the potential benefits of metformin treatment after CRT and emphasize the need for caution in repurposing metformin in clinical studies.
Collapse
Affiliation(s)
- Nili Yuen
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kamila U Szulc-Lerch
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Yu-Qing Li
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Cindi M Morshead
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Terrence Donelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Donald J Mabbott
- Department of Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - C Shun Wong
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Brian J Nieman
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Gupta R, Khan R, Cortes CJ. Forgot to Exercise? Exercise Derived Circulating Myokines in Alzheimer's Disease: A Perspective. Front Neurol 2021; 12:649452. [PMID: 34276532 PMCID: PMC8278015 DOI: 10.3389/fneur.2021.649452] [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: 01/11/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
Regular exercise plays an essential role in maintaining healthy neurocognitive function and central nervous system (CNS) immuno-metabolism in the aging CNS. Physical activity decreases the risk of developing Alzheimer's Disease (AD), is associated with better AD prognosis, and positively affects cognitive function in AD patients. Skeletal muscle is an important secretory organ, communicating proteotoxic and metabolic stress to distant tissues, including the CNS, through the secretion of bioactive molecules collectively known as myokines. Skeletal muscle undergoes significant physical and metabolic remodeling during exercise, including alterations in myokine expression profiles. This suggests that changes in myokine and myometabolite secretion may underlie the well-documented benefits of exercise in AD. However, to date, very few studies have focused on specific alterations in skeletal muscle-originating secreted factors and their potential neuroprotective effects in AD. In this review, we discuss exercise therapy for AD prevention and intervention, and propose the use of circulating myokines as novel therapeutic tools for modifying AD progression.
Collapse
Affiliation(s)
- Rajesh Gupta
- Department of Cell, Developmental and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rizwan Khan
- Department of Cell, Developmental and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Constanza J Cortes
- Department of Cell, Developmental and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Center for Neurodegeneration and Experimental Therapeutics (CNET), University of Alabama at Birmingham, Birmingham, AL, United States.,Center for Exercise Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,UAB Nathan Shock Center for the Excellence in the Study of Aging, University of Alabama at Birmingman, Birmingham, AL, United States
| |
Collapse
|
29
|
Sharma S, Nozohouri S, Vaidya B, Abbruscato T. Repurposing metformin to treat age-related neurodegenerative disorders and ischemic stroke. Life Sci 2021; 274:119343. [PMID: 33716063 PMCID: PMC8996678 DOI: 10.1016/j.lfs.2021.119343] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Aging is a risk factor for major central nervous system (CNS) disorders. More specifically, aging can be inked to neurodegenerative diseases (NDs) because of its deteriorating impact on neurovascular unit (NVU). Metformin, a first line FDA-approved anti-diabetic drug, has gained increasing interest among researchers for its role in improving aging-related neurodegenerative disorders. Additionally, numerous studies have illustrated metformin's role in ischemic stroke, a cerebrovascular disorder in which the NVU becomes dysfunctional which can lead to permanent life-threatening disabilities. Considering metformin's beneficial preclinical actions on various disorders, and the drug's role in alleviating severity of these conditions through involvement in commonly characterized cellular pathways, we discuss the potential of metformin as a suitable drug candidate for repurposing in CNS disorders.
Collapse
Affiliation(s)
- Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA.
| |
Collapse
|
30
|
Derkach D, Kehtari T, Renaud M, Heidari M, Lakshman N, Morshead CM. Metformin pretreatment rescues olfactory memory associated with subependymal zone neurogenesis in a juvenile model of cranial irradiation. Cell Rep Med 2021; 2:100231. [PMID: 33948569 PMCID: PMC8080112 DOI: 10.1016/j.xcrm.2021.100231] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/12/2020] [Accepted: 03/09/2021] [Indexed: 01/23/2023]
Abstract
Cranial irradiation (IR) is an effective adjuvant therapy in the treatment of childhood brain tumors but results in long-lasting cognitive deficits associated with impaired neurogenesis, as evidenced in rodent models. Metformin has been shown to expand the endogenous neural stem cell (NSC) pool and promote neurogenesis under physiological conditions and in response to neonatal brain injury, suggesting a potential role in neurorepair. Here, we assess whether metformin pretreatment, a clinically feasible treatment for children receiving cranial IR, promotes neurorepair in a mouse cranial IR model. Using immunofluorescence and the in vitro neurosphere assay, we show that NSCs are depleted by cranial IR but spontaneously recover, although deficits to proliferative neuroblasts persist. Metformin pretreatment enhances the recovery of neurogenesis, attenuates the microglial response, and promotes recovery of long-term olfactory memory. These findings indicate that metformin is a promising candidate for further preclinical and clinical investigations of neurorepair in childhood brain injuries.
Collapse
Affiliation(s)
- Daniel Derkach
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Tarlan Kehtari
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Matthew Renaud
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Mohsen Heidari
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Nishanth Lakshman
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Cindi M. Morshead
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Grissi M, Boudot C, Assem M, Candellier A, Lando M, Poirot-Leclercq S, Boullier A, Bennis Y, Lenglet G, Avondo C, Lalau JD, Choukroun G, Massy ZA, Kamel S, Chillon JM, Hénaut L. Metformin prevents stroke damage in non-diabetic female mice with chronic kidney disease. Sci Rep 2021; 11:7464. [PMID: 33811249 PMCID: PMC8018962 DOI: 10.1038/s41598-021-86905-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) worsens ischemic stroke severity in both patients and animals. In mice, these poorer functional outcomes are associated with decreased brain activity of AMP-activated protein kinase (AMPK), a molecule that recently emerged as a potential therapeutic target for ischemic stroke. The antidiabetic drug metformin, a well-known activator of AMPK, has improved stroke outcomes in diabetic patients with normal renal function. We investigated whether chronic metformin pre-conditioning can rescue AMPK activity and prevent stroke damage in non-diabetic mice with CKD. Eight-week-old female C57BL/6J mice were assigned to CKD or SHAM groups. CKD was induced through right kidney cortical electrocautery, followed by left total nephrectomy. Mice were then allocated to receive metformin (200 mg/kg/day) or vehicle for 5 weeks until stroke induction by transient middle cerebral artery occlusion (tMCAO). The infarct volumes were lower in CKD mice exposed to metformin than in vehicle-treated CKD mice 24 h after tMCAO. Metformin pre-conditioning of CKD mice improved their neurological score, grip strength, and prehensile abilities. It also enhanced AMPK activation, reduced apoptosis, increased neuron survival and decreased microglia/macrophage M1 signature gene expression as well as CKD-induced activation of the canonical NF-κB pathway in the ischemic lesions of CKD mice.
Collapse
MESH Headings
- Adenylate Kinase/metabolism
- Animals
- Apoptosis/drug effects
- Body Weight
- Brain Infarction/blood
- Brain Infarction/complications
- Brain Infarction/drug therapy
- Brain Infarction/genetics
- Enzyme Activation/drug effects
- Female
- Gene Expression Regulation
- Gliosis/blood
- Gliosis/complications
- Gliosis/drug therapy
- Infarction, Middle Cerebral Artery/blood
- Infarction, Middle Cerebral Artery/complications
- Infarction, Middle Cerebral Artery/drug therapy
- Infarction, Middle Cerebral Artery/genetics
- Ischemic Preconditioning
- Macrophages/drug effects
- Macrophages/pathology
- Metformin/blood
- Metformin/pharmacology
- Metformin/therapeutic use
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/pathology
- Models, Biological
- NF-kappa B/metabolism
- Neurons/drug effects
- Neurons/pathology
- Renal Insufficiency, Chronic/blood
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/genetics
- Stroke/drug therapy
- Stroke/genetics
- Stroke/prevention & control
- Mice
Collapse
Affiliation(s)
- Maria Grissi
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
| | - Cédric Boudot
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
| | - Maryam Assem
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Faculty of Medicine, University of Picardie Jules Verne, 80000, Amiens, France
- Division of Nephrology, Amiens University Hospital, 80054, Amiens, France
| | - Alexandre Candellier
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Faculty of Medicine, University of Picardie Jules Verne, 80000, Amiens, France
- Division of Nephrology, Amiens University Hospital, 80054, Amiens, France
| | - Mathilde Lando
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Faculty of Medicine, University of Picardie Jules Verne, 80000, Amiens, France
- Division of Nephrology, Amiens University Hospital, 80054, Amiens, France
| | - Sabrina Poirot-Leclercq
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
| | - Agnès Boullier
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Faculty of Medicine, University of Picardie Jules Verne, 80000, Amiens, France
- Department of Biochemistry, Amiens University Hospital, 80054, Amiens, France
| | - Youssef Bennis
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Department of Clinical Pharmacology, Amiens University Hospital, 80054, Amiens, France
| | - Gaëlle Lenglet
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
| | - Carine Avondo
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
| | - Jean-Daniel Lalau
- Faculty of Medicine, University of Picardie Jules Verne, 80000, Amiens, France
- Department of Endocrinology-Diabetology-Nutrition, Amiens University Hospital, 80054, Amiens, France
- UMR_I 01, PériTox, CURS, 80054, Amiens, France
| | - Gabriel Choukroun
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Faculty of Medicine, University of Picardie Jules Verne, 80000, Amiens, France
- Division of Nephrology, Amiens University Hospital, 80054, Amiens, France
| | - Ziad A Massy
- Department of Nephrology, Ambroise Paré University Hospital, APHP, 92104, Boulogne-Billancourt, France
- Inserm U1018-Team 5, CESP, UVSQ, University Paris Saclay, 94807, Villejuif, France
- University Versailles-Saint Quentin, University Paris-Saclay, 91190, Villejuif, France
| | - Saïd Kamel
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Department of Biochemistry, Amiens University Hospital, 80054, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, 80000, Amiens, France
| | - Jean-Marc Chillon
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, 80000, Amiens, France
- Direction of Clinical Research, Amiens University Hospital, 80054, Amiens, France
| | - Lucie Hénaut
- UR UPJV 7517, MP3CV, CURS, Université de Picardie Jules Verne, Avenue René Laennec, 80054, Amiens, France.
| |
Collapse
|
32
|
Ren Y, Luo H. Metformin: The next angiogenesis panacea? SAGE Open Med 2021; 9:20503121211001641. [PMID: 33796300 PMCID: PMC7970164 DOI: 10.1177/20503121211001641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/17/2021] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis, the development of new blood vessels from existing ones, is
a critical process in wound healing and skeletal muscle hypertrophy.
It also leads to pathological conditions such as retinopathy and tumor
genesis. Metformin, the first-line treatment for type 2 diabetic
mellitus, has a specific regulatory effect on the process of
angiogenesis. Anti-angiogenesis can inhibit the occurrence and
metastasis of tumors and alleviate patients’ symptoms with polycystic
ovary syndrome. Moreover, promoting angiogenesis effect can accelerate
wound healing and promote stroke recovery and limb ischemia
reconstruction. This review reorganizes metformin in angiogenesis, and
the underlying mechanism in alleviating disease to bring some
inspiration to relevant researchers.
Collapse
Affiliation(s)
- Yu Ren
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Hua Luo
- Department of Orthopaedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| |
Collapse
|
33
|
Bressan C, Saghatelyan A. Intrinsic Mechanisms Regulating Neuronal Migration in the Postnatal Brain. Front Cell Neurosci 2021; 14:620379. [PMID: 33519385 PMCID: PMC7838331 DOI: 10.3389/fncel.2020.620379] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/08/2020] [Indexed: 01/19/2023] Open
Abstract
Neuronal migration is a fundamental brain development process that allows cells to move from their birthplaces to their sites of integration. Although neuronal migration largely ceases during embryonic and early postnatal development, neuroblasts continue to be produced and to migrate to a few regions of the adult brain such as the dentate gyrus and the subventricular zone (SVZ). In the SVZ, a large number of neuroblasts migrate into the olfactory bulb (OB) along the rostral migratory stream (RMS). Neuroblasts migrate in chains in a tightly organized micro-environment composed of astrocytes that ensheath the chains of neuroblasts and regulate their migration; the blood vessels that are used by neuroblasts as a physical scaffold and a source of molecular factors; and axons that modulate neuronal migration. In addition to diverse sets of extrinsic micro-environmental cues, long-distance neuronal migration involves a number of intrinsic mechanisms, including membrane and cytoskeleton remodeling, Ca2+ signaling, mitochondria dynamics, energy consumption, and autophagy. All these mechanisms are required to cope with the different micro-environment signals and maintain cellular homeostasis in order to sustain the proper dynamics of migrating neuroblasts and their faithful arrival in the target regions. Neuroblasts in the postnatal brain not only migrate into the OB but may also deviate from their normal path to migrate to a site of injury induced by a stroke or by certain neurodegenerative disorders. In this review, we will focus on the intrinsic mechanisms that regulate long-distance neuroblast migration in the adult brain and on how these pathways may be modulated to control the recruitment of neuroblasts to damaged/diseased brain areas.
Collapse
Affiliation(s)
- Cedric Bressan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| | - Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
34
|
Wang G, Lin F, Wan Q, Wu J, Luo M. Mechanisms of action of metformin and its regulatory effect on microRNAs related to angiogenesis. Pharmacol Res 2020; 164:105390. [PMID: 33352227 DOI: 10.1016/j.phrs.2020.105390] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023]
Abstract
Angiogenesis is rapidly initiated in response to pathological conditions and is a key target for pharmaceutical intervention in various malignancies. Anti-angiogenic therapy has emerged as a potential and effective therapeutic strategy for treating cancer and cardiovascular-related diseases. Metformin, a first-line oral antidiabetic agent for type 2 diabetes mellitus (T2DM), not only reduces blood glucose levels and improves insulin sensitivity and exerts cardioprotective effects but also shows benefits against cancers, cardiovascular diseases, and other diverse diseases and regulates angiogenesis. MicroRNAs (miRNAs) are endogenous noncoding RNA molecules with a length of approximately 19-25 bases that are widely involved in controlling various human biological processes. A large number of miRNAs are involved in the regulation of cardiovascular cell function and angiogenesis, of which miR-21 not only regulates vascular cell proliferation, migration and apoptosis but also plays an important role in angiogenesis. The relationship between metformin and abnormal miRNA expression has gradually been revealed in the context of numerous diseases and has received increasing attention. This paper reviews the drug-target interactions and drug repositioning events of metformin that influences vascular cells and has benefits on angiogenesis-mediated effects. Furthermore, we use miR-21 as an example to explain the specific molecular mechanism underlying metformin-mediated regulation of the miRNA signaling pathway controlling angiogenesis and vascular protective effects. These findings may provide a new therapeutic target and theoretical basis for the clinical prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Fang Lin
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Qin Wan
- Department of Endocrinology, Nephropathy Clinical Medical Research Center of Sichuan Province, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
35
|
Fan YY, Wang YJ, Guo J, Wu MN, Zhang MS, Niu BL, Li Y, Zhao J, Yang CH, Li Y, Chen M, Jiao XY. Delayed metformin treatment improves functional recovery following traumatic brain injury via central AMPK-dependent brain tissue repair. Brain Res Bull 2020; 164:146-156. [DOI: 10.1016/j.brainresbull.2020.08.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 11/15/2022]
|
36
|
Acute administration of metformin prior to cardiac ischemia/reperfusion injury protects brain injury. Eur J Pharmacol 2020; 885:173418. [DOI: 10.1016/j.ejphar.2020.173418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 11/19/2022]
|
37
|
Zhang S, Lachance BB, Moiz B, Jia X. Optimizing Stem Cell Therapy after Ischemic Brain Injury. J Stroke 2020; 22:286-305. [PMID: 33053945 PMCID: PMC7568970 DOI: 10.5853/jos.2019.03048] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Stem cells have been used for regenerative and therapeutic purposes in a variety of diseases. In ischemic brain injury, preclinical studies have been promising, but have failed to translate results to clinical trials. We aimed to explore the application of stem cells after ischemic brain injury by focusing on topics such as delivery routes, regeneration efficacy, adverse effects, and in vivo potential optimization. PUBMED and Web of Science were searched for the latest studies examining stem cell therapy applications in ischemic brain injury, particularly after stroke or cardiac arrest, with a focus on studies addressing delivery optimization, stem cell type comparison, or translational aspects. Other studies providing further understanding or potential contributions to ischemic brain injury treatment were also included. Multiple stem cell types have been investigated in ischemic brain injury treatment, with a strong literature base in the treatment of stroke. Studies have suggested that stem cell administration after ischemic brain injury exerts paracrine effects via growth factor release, blood-brain barrier integrity protection, and allows for exosome release for ischemic injury mitigation. To date, limited studies have investigated these therapeutic mechanisms in the setting of cardiac arrest or therapeutic hypothermia. Several delivery modalities are available, each with limitations regarding invasiveness and safety outcomes. Intranasal delivery presents a potentially improved mechanism, and hypoxic conditioning offers a potential stem cell therapy optimization strategy for ischemic brain injury. The use of stem cells to treat ischemic brain injury in clinical trials is in its early phase; however, increasing preclinical evidence suggests that stem cells can contribute to the down-regulation of inflammatory phenotypes and regeneration following injury. The safety and the tolerability profile of stem cells have been confirmed, and their potent therapeutic effects make them powerful therapeutic agents for ischemic brain injury patients.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brittany Bolduc Lachance
- Program in Trauma, Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bilal Moiz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Livingston JM, Syeda T, Christie T, Gilbert EA, Morshead CM. Subacute metformin treatment reduces inflammation and improves functional outcome following neonatal hypoxia ischemia. Brain Behav Immun Health 2020; 7:100119. [PMID: 34589876 PMCID: PMC8474427 DOI: 10.1016/j.bbih.2020.100119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/03/2023] Open
Abstract
Hypoxia-ischemia (HI) injury is a leading cause of neonatal death and long-term disability, and existing treatment options for HI offer only modest benefit. Early intervention with the drug metformin has been shown to promote functional improvement in numerous rodent models of injury and has pleiotropic cellular effects in the brain. We have previously shown that 1 week of metformin treatment initiated 24 h after HI in neonatal mice resulted in improved motor and cognitive performance, activation of endogenous neural precursor cells (NPCs), and increased oligodendrogenesis. While promising, a limitation to this work is that immediate pharmacological intervention is not always possible in the clinic. Herein, we investigated whether delaying metformin treatment to begin in the subacute phase post-HI would still effectively promote recovery. Male and female C57/BL6 mice received HI injury postnatally, and metformin treatment began 7 days post-HI for up to 4 weeks. Motor and cognitive performance was assessed across time using behavioural tests (cylinder, foot fault, puzzle box). We found that metformin improved motor and cognitive behaviour, decreased inflammation, and increased oligodendrocytes in the motor cortex. Our present findings demonstrate that a clinically relevant subacute metformin treatment paradigm affords the potential to treat neonatal HI, and that improved outcomes occur through modulation of the inflammatory response and oligodendrogenesis. Subacute metformin treatment improves functional recovery after neonatal hypoxia ischemia. Metformin reduces the number of microglia present in the brain early after injury. Metformin increases the number of oligodendrocytes present in the chronic post-injury phase. Metformin treatment has therapeutic potential in the treatment of hypoxic ischemic brain damage.
Collapse
Affiliation(s)
- Jessica M. Livingston
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Tasfia Syeda
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Taryn Christie
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Emily A.B. Gilbert
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Cindi M. Morshead
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
- Institute of Medical Science, University of Toronto, Toronto, M5S1A8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S3E1, Canada
- Corresponding author. Faculty of Medicine, Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada.
| |
Collapse
|
39
|
El-Ghaiesh SH, Bahr HI, Ibrahiem AT, Ghorab D, Alomar SY, Farag NE, Zaitone SA. Metformin Protects From Rotenone-Induced Nigrostriatal Neuronal Death in Adult Mice by Activating AMPK-FOXO3 Signaling and Mitigation of Angiogenesis. Front Mol Neurosci 2020; 13:84. [PMID: 32625061 PMCID: PMC7314970 DOI: 10.3389/fnmol.2020.00084] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease that affects substantia nigra dopamine neurons. Many studies have documented the role of oxidative stress and angiogenesis in the pathogenesis of PD. Metformin (MTF) is an antidiabetic medication and AMP-activated protein kinase (AMPK) regulator that has shown antioxidant and antiangiogenic properties in many disorders. The aim of this study is to investigate the neuroprotective effect of MTF in a mouse model of rotenone-prompted PD with a highlight on its influence on the AMPK/forkhead box transcription factor O3 (FOXO3) pathway and striatal angiogenesis. In the running study, PD was induced in mice using repeated doses of rotenone and concomitantly treated with MTF 100 or 200 mg/kg/day for 18 days. Rotarod and pole tests were used to examine the animals’ motor functionality. After that, animals were sacrificed, and brains were isolated and processed for immunohistochemical investigations or biochemical analyses. Oxidant stress and angiogenic markers were measured, including reduced glutathione, malondialdehyde, the nuclear factor erythroid 2–related factor 2 (Nrf2), hemoxygenase-1, thioredoxin, AMPK, FOXO3, and vascular endothelial growth factor (VEGF). Results indicated that MTF improved animals’ motor function, improved striatal glutathione, Nrf2, hemoxygenase-1, and thioredoxin. Furthermore, MTF upregulated AMPK-FOXO3 proteins and reduced VEGF and cleaved caspase 3. MTF also increased the number of tyrosine hydroxylase (TH)–stained neurons in the substantia nigra neurons and in striatal neuronal terminals. This study is the first to highlight that the neuroprotective role of MTF is mediated through activation of AMPK-FOXO3 signaling and inhibition of the proangiogenic factor, VEGF. Further studies are warranted to confirm this mechanism in other models of PD and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hoda I Bahr
- Department of Biochemistry, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Afaf T Ibrahiem
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa Ghorab
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Suliman Y Alomar
- Doping Research Chair, Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Noha E Farag
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,Department of Physiology, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
40
|
Tang BL. Could metformin be therapeutically useful in Huntington's disease? Rev Neurosci 2020; 31:297-317. [PMID: 31751298 DOI: 10.1515/revneuro-2019-0072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggest that dimethylbiguanide (metformin), a first-line drug for type 2 diabetes mellitus, could be neuroprotective in a range of brain pathologies, which include neurodegenerative diseases and brain injury. However, there are also contraindications that associate metformin treatment with cognitive impairment as well as adverse outcomes in Alzheimer's disease and Parkinson's disease animal models. Recently, a beneficial effect of metformin in animal models of Huntington's disease (HD) has been strengthened by multiple reports. In this brief review, the findings associated with the effects of metformin in attenuating neurodegenerative diseases are discussed, focusing on HD-associated pathology and the potential underlying mechanisms highlighted by these studies. The mechanism of action of metformin is complex, and its therapeutic efficacy is therefore expected to be dependent on the disease context. The key metabolic pathways that are effectively affected by metformin, such as AMP-activated protein kinase activation, may be altered in the later decades of the human lifespan. In this regard, metformin may nonetheless be therapeutically useful for neurological diseases with early pathological onsets, such as HD.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, Singapore 117596, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Medical Drive, Singapore 119077, Singapore
| |
Collapse
|
41
|
Kim D, Liu QF, Jeong HJ, Han SH, Kim DI, Jeon S. A Modified Formulation of Sutaehwan Ameliorates Menopausal Anxiety, Depression and Heart Hypertrophy in the VCD-Induced Menopausal Mouse Model. Biol Pharm Bull 2020; 42:1471-1481. [PMID: 31474708 DOI: 10.1248/bpb.b19-00056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sutaehwan (STH) has been used in Korean medicine for the treatment of abortus habitualis such as fetal restlessness in the uterus. Previously, we reported that a modified formulation of STH, Sutaehwan-Gami, has phytoestrogen-like properties in an ovariectomized menopausal rat model. However, the therapeutic effects of STH and the precise mechanisms by which STH affects various menopausal symptoms remain poorly understood. The current study was designed to explore the effects of a modified form of STH on menopausal anxiety, depression and heart hypertrophy and its mechanisms in 4-vinylcyclohexene diepoxide (VCD)-induced menopausal mouse models. VCD-induced menopausal model mice were fed a modified form of STH, which contained water extract of 3 herbs (called STH_KP17001) at a dose of 100 or 300 mg/kg/d or as a positive control, estradiol at a dose of 0.2 mg/kg/d with standard mouse pellets for 13 weeks. The results show that STH_KP17001 significantly restored the VCD-induced weight reduction of uterine and ovary through the phosphorylation of extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) in the uterus and ovary. Moreover, STH_KP17001 showed slight proliferative effects and estrogen receptor α phosphorylation in MCF-7 cells. Treatment with STH_KP17001 reversed VCD-induced anxiety and depression through AMP-activated protein kinase (AMPK) activation and brain-derived neurotrophic factor (BDNF) expression in the cerebral cortex, while improving heart hypertrophy through inactivation of inhibitor of kappaB α (IκBα) in the heart. The results indicate that STH_KP17001 improves menopause-induced anxiety, depression and heart hypertrophy, implying its protective role for the management of menopausal symptoms.
Collapse
Affiliation(s)
- Deokho Kim
- Department of Korean Medicine, Graduate School of Dongguk University
| | - Quan Feng Liu
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University
| | - Ha Jin Jeong
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University
| | | | - Dong-Il Kim
- Department of Obstetrics & Gynecology, College of Korean Medicine, Dongguk University
| | - Songhee Jeon
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University
| |
Collapse
|
42
|
Metformin reduces neuronal damage and promotes neuroblast proliferation and differentiation in a cerebral ischemia/reperfusion rat model. Neuroreport 2019; 30:232-240. [PMID: 30614910 DOI: 10.1097/wnr.0000000000001190] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
According to the previous research, metformin, a medication utilized for type 2 diabetes management, inhibits neural aging and reduces infarct size by enhancing angiogenesis in a mouse stroke model. What is more, metformin administration also promotes neural precursor cells proliferation, migration, as well as differentiation for newborn mice with hypoxia-ischemia brain injury. However, whether metformin regulates neurogenesis in an adult rat ischemia/reperfusion (I/R) model remains unclear. The current research found that metformin administration reduced neuronal damage in the CA1 area of hippocampus in a rat model of I/R. The number of neuronal nuclei positive neuron was significantly increased and glial fibrillary acidic protein positive astrocyte became obviously declined in the CA1 region in I/R rats treated with metformin. It was further demonstrated that metformin promoted neuroblasts proliferation and neuronal differentiation in the subgranular zone of the dentate gyrus and inhibited the formation of astrocyte. Our study indicates that activation of endogenous neuroblasts using metformin will become a favorable target in therapeutic intervention of cerebral ischemia injury models.
Collapse
|
43
|
Khacho M, Harris R, Slack RS. Mitochondria as central regulators of neural stem cell fate and cognitive function. Nat Rev Neurosci 2019; 20:34-48. [PMID: 30464208 DOI: 10.1038/s41583-018-0091-3] [Citation(s) in RCA: 254] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Emerging evidence now indicates that mitochondria are central regulators of neural stem cell (NSC) fate decisions and are crucial for both neurodevelopment and adult neurogenesis, which in turn contribute to cognitive processes in the mature brain. Inherited mutations and accumulated damage to mitochondria over the course of ageing serve as key factors underlying cognitive defects in neurodevelopmental disorders and neurodegenerative diseases, respectively. In this Review, we explore the recent findings that implicate mitochondria as crucial regulators of NSC function and cognition. In this respect, mitochondria may serve as targets for stem-cell-based therapies and interventions for cognitive defects.
Collapse
Affiliation(s)
- Mireille Khacho
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology (OISB), University of Ottawa, Ottawa, Ontario, Canada
| | - Richard Harris
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
44
|
Pharmaceutical, clinical, and immunohistochemical studies of metformin hydrochloride topical hydrogel for wound healing application. Arch Dermatol Res 2019; 312:113-121. [DOI: 10.1007/s00403-019-01982-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/03/2019] [Accepted: 09/21/2019] [Indexed: 12/23/2022]
|
45
|
Ruddy RM, Adams KV, Morshead CM. Age- and sex-dependent effects of metformin on neural precursor cells and cognitive recovery in a model of neonatal stroke. SCIENCE ADVANCES 2019; 5:eaax1912. [PMID: 31535024 PMCID: PMC6739114 DOI: 10.1126/sciadv.aax1912] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/15/2019] [Indexed: 05/30/2023]
Abstract
Resident neural stem and progenitor cells, collectively termed neural precursor cells (NPCs), reside in a well-defined neurogenic niche in the subventricular zone (SVZ) and contribute to ongoing postnatal neurogenesis. It is well established that the NPC niche can alter the behavior of NPCs. NPC activation is a promising therapeutic strategy for brain repair. The drug metformin has been shown to activate neural stem cells, promote differentiation, and lead to functional motor recovery in a neonatal stroke model. We demonstrate that metformin-induced NPC expansion and functional recovery is sex hormone dependent. Metformin increases the size of the NPC pool in adult females, but not males, and promotes cognitive recovery in a model of brain injury in females, but not males. Our data demonstrate that metformin has age- and sex-dependent effects on NPCs that correlate with functional recovery, which has important implications for neural repair.
Collapse
Affiliation(s)
- Rebecca M. Ruddy
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Kelsey V. Adams
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Cindi M. Morshead
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
46
|
Ahn S, Liu QF, Jang JH, Park J, Jeong HJ, Kim Y, Kim DH, Jeong G, Oh ST, Park SU, Cho SY, Park HJ, Jeon S. Gami-Chunggan Formula Prevents Motor Dysfunction in MPTP/p-Induced and A53T α-Synuclein Overexpressed Parkinson's Disease Mouse Model Though DJ-1 and BDNF Expression. Front Aging Neurosci 2019; 11:230. [PMID: 31555122 PMCID: PMC6724569 DOI: 10.3389/fnagi.2019.00230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 08/13/2019] [Indexed: 12/25/2022] Open
Abstract
The Gami–Chunggan formula (GCF) is a modification of the Chunggan (CG) decoction, which has been used to treat movement disorders such as Parkinson’s disease (PD) in Traditional East Asian Medicine. To evaluate the neuroprotective effects of GCF in chronic PD animal models, we used either a 5-week treatment of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine with probenecid (MPTP/p) or the α-synuclein A53T overexpressed PD mouse model. C57BL/6 mice were treated with MPTP, in combination with probenecid, for 5 weeks. GCF was administered simultaneously with MPTP injection for 38 days. The A53T α-synuclein overexpressed mice were also fed with GCF for 60 days. Using behavioral readouts and western blot analyses, it was observed that GCF prevents motor dysfunction in the MPTP/p-induced and A53T α-synuclein overexpressed mice. Moreover, GCF inhibited the reduction of dopaminergic neurons in the substantia nigra (SN) and fibers in the striatum (ST) against MPTP/p challenge. The expression of DJ-1 was increased but that of α-synuclein was decreased in the SN of PD-like brains by GCF administration. In vitro experiments also showed that GCF inhibited 6-OHDA-induced neurotoxicity in SH-SY5Y neuroblastoma cell lines and that it did so to a greater degree than CG. Furthermore, GCF induced BDNF expression through phosphorylation of Akt, ERK, CREB, and AMPK in the SN of PD-like brains. Therefore, use of the herbal medicine GCF offers a potential remedy for neurodegenerative disorders, including Parkinson’s disease.
Collapse
Affiliation(s)
- Sora Ahn
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Quan Feng Liu
- Department of Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Gyeongju, South Korea
| | - Jae-Hwan Jang
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jeonghun Park
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Ha Jin Jeong
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea
| | - Youngman Kim
- Research Institute, Dong Kwang Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Dong-Hee Kim
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Guwon Jeong
- Research Institute, Dong Kwang Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Seung Tack Oh
- Research Institute, Dong Kwang Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Seong-Uk Park
- Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Seung-Yeon Cho
- Stroke and Neurological Disorders Center, Kyung Hee University Hospital at Gangdong, Kyung Hee University, Seoul, South Korea
| | - Hi-Joon Park
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, Seoul, South Korea.,Studies of Translational Acupuncture Research (STAR), Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, Seoul, South Korea
| | - Songhee Jeon
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
47
|
Muri L, Le ND, Zemp J, Grandgirard D, Leib SL. Metformin mediates neuroprotection and attenuates hearing loss in experimental pneumococcal meningitis. J Neuroinflammation 2019; 16:156. [PMID: 31351490 PMCID: PMC6660697 DOI: 10.1186/s12974-019-1549-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/18/2019] [Indexed: 12/16/2022] Open
Abstract
Background Pneumococcal meningitis is associated with high risk of neurological sequelae such as cognitive impairment and hearing loss. These sequelae are due to parenchymal brain and inner ear damage primarily induced by the excessive inflammatory reaction in response to bacterial brain invasion. Metformin—a biguanide drug to treat diabetes mellitus type 2—was recently found to suppress neuroinflammation and induce neuroregeneration. This study evaluated the effect of metformin adjunctive to antibiotics on neuroinflammation, brain and inner ear damage, and neurofunctional outcome in experimental pediatric pneumococcal meningitis. Methods Eleven-day-old Wistar rats were infected intracisternally with 5.22 ± 1.27 × 103 CFU Streptococcus pneumoniae and randomized for treatment with metformin (50 mg/kg, i.p., once daily for 3 weeks) plus ceftriaxone (100 mg/kg, i.p., bid, n = 61) or ceftriaxone monotherapy (n = 79). Cortical damage and hippocampal apoptosis were evaluated histomorphometrically 42 h post infection. Cerebrospinal fluid cytokine levels were analyzed during acute infection. Five weeks post infection, auditory brainstem responses were measured to determine hearing thresholds. Spiral ganglion neuron density and abundance of recently proliferated and integrated hippocampal granule neurons were assessed histologically. Additionally, the anti-inflammatory effect of metformin was studied in primary rat astroglial cells in vitro. Results Upon pneumococcal infection, metformin treatment significantly reduced levels of inflammatory cytokines and nitric oxide production in cerebrospinal fluid and in astroglial cell cultures in vitro (p < 0.05). Compared to animals receiving ceftriaxone monotherapy, adjunctive metformin significantly reduced cortical necrosis (p < 0.02) during acute infection and improved median click-induced hearing thresholds (60 dB vs. 100 dB, p < 0.002) 5 weeks after infection. Adjuvant metformin significantly improved pure tone hearing thresholds at all assessed frequencies compared to ceftriaxone monotherapy (p < 0.05) and protected from PM-induced spiral ganglion neuron loss in the inner ear (p < 0.05). Conclusion Adjuvant metformin reduces brain injury during pneumococcal meningitis by decreasing the excessive neuroinflammatory response. Furthermore, it protects spiral ganglion neurons in the inner ear and improves hearing impairments after experimental pneumococcal meningitis. These results identify adjuvant metformin as a promising therapeutic option to improve the outcome after pediatric pneumococcal meningitis. Electronic supplementary material The online version of this article (10.1186/s12974-019-1549-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lukas Muri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Mittelstrasse 43, 3012, Bern, Switzerland
| | - Ngoc Dung Le
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Mittelstrasse 43, 3012, Bern, Switzerland
| | - Jonas Zemp
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland
| | - Denis Grandgirard
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland
| | - Stephen L Leib
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3010, Bern, Switzerland.
| |
Collapse
|
48
|
Pereira AF, Pereira LMS, Silva CMP, Freitas Alves BW, Barbosa JS, Pinto FMM, Pereira AC, Silva KO, Pontes RB, Alencar NMN, Lima-Júnior RCP, Vale ML. Metformin reduces c-Fos and ATF3 expression in the dorsal root ganglia and protects against oxaliplatin-induced peripheral sensory neuropathy in mice. Neurosci Lett 2019; 709:134378. [PMID: 31325582 DOI: 10.1016/j.neulet.2019.134378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022]
Abstract
Oxaliplatin is a third-generation platinum drug commonly used as the first line treatment of metastatic colorectal cancer. Oxaliplatin-based anticancer regimens course with dose-limiting neurotoxicity. The pharmacological strategies used to manage such side effect are not totally effective. Metformin is an anti-diabetic drug that is described to negatively modulate painful diabetic neuropathy. Then, this study aimed to assess the effect of metformin in the oxaliplatin-induced peripheral sensory neuropathy in mice. For that purpose, Swiss male mice were injected with oxaliplatin (1, 2 or 4 mg/kg, i.v., twice a week with a total of nine injections) alone or in combination with daily administration of metformin (250 mg/kg, p.o.). Thermal and mechanical nociceptive tests were performed once a week for five weeks. Then, the animals were euthanized on day 35 post-first injection of oxaliplatin and the dorsal root ganglia were harvested for the assessment of c-Fos and ATF3 expressions. Oxaliplatin caused a nociceptive response accompanied by the increased expression of c-Fos and ATF3 in the dorsal root ganglia and spinal cord. In addition, the oxaliplatin-associated nociception was significantly attenuated by metformin (P < 0.05), which also reduced the expression of c-Fos and ATF3 (P < 0.05). Therefore, metformin protected from the peripheral sensory neuropathy induced by oxaliplatin, which was confirmed by the reduction of c-Fos and ATF3 expression, two known neuronal activation and damage markers, respectively.
Collapse
Affiliation(s)
- Anamaria Falcão Pereira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lus Mário Silva Pereira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Bruno Wesley Freitas Alves
- Department of Morphology, Morpho-functional Sciences Post Graduation Program, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jéssica Sales Barbosa
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Ana Carolina Pereira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Karla Oliveira Silva
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Renata Bessa Pontes
- Department of Physical Therapy, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Nylane Maria Nunes Alencar
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Roberto César Pereira Lima-Júnior
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Mariana Lima Vale
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Department of Morphology, Morpho-functional Sciences Post Graduation Program, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
49
|
Wang Q, Guo X, Li L, Gao Z, Ji M. Treatment with metformin and sorafenib alleviates endometrial hyperplasia in polycystic ovary syndrome by promoting apoptosis via synergically regulating autophagy. J Cell Physiol 2019; 235:1339-1348. [PMID: 31256441 DOI: 10.1002/jcp.29051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/29/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Qian‐qing Wang
- Gynecologic Oncology Department Xinxiang City Central Hospital in Henan Province Xinxiang Henan China
| | - Xiang‐cui Guo
- Gynecologic Oncology Department Xinxiang City Central Hospital in Henan Province Xinxiang Henan China
| | - Li Li
- Gynecologic Oncology Department Xinxiang City Central Hospital in Henan Province Xinxiang Henan China
| | - Zhi‐hui Gao
- Gynecologic Oncology Department Xinxiang City Central Hospital in Henan Province Xinxiang Henan China
| | - Mei Ji
- Gynecology Department Zhengzhou University First Affiliated Hospital Zhengzhou Henan China
| |
Collapse
|
50
|
Leech T, Chattipakorn N, Chattipakorn SC. The beneficial roles of metformin on the brain with cerebral ischaemia/reperfusion injury. Pharmacol Res 2019; 146:104261. [PMID: 31170502 DOI: 10.1016/j.phrs.2019.104261] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Cerebral ischaemia/reperfusion (I/R) injury is the transient loss, followed by rapid return, of blood flow to the brain. This condition is often caused by strokes and heart attacks. The underlying mechanisms resulting in brain damage during cerebral I/R injury include mitochondrial dysregulation, increased oxidative stress/reactive oxygen species, blood-brain-barrier breakdown, inflammation of the brain, and increased neuronal apoptosis. Metformin is the first-line antidiabetic drug which has recently been shown to be capable of acting through the aforementioned pathways to improve recovery following cerebral I/R injury. However, some studies have suggested that metformin therapy may have no effect or even worsen recovery following cerebral I/R injury. The present review will compile and examine the available in vivo, in vitro, and clinical data concerning the neuroprotective effects of metformin following cerebral I/R injury. Any contradictory evidence will also be assessed and presented to determine the actual effectiveness of metformin treatment in stroke recovery.
Collapse
Affiliation(s)
- Tom Leech
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, United Kingdom; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|