1
|
Lewis CM, Hoffmann A, Helmchen F. Linking brain activity across scales with simultaneous opto- and electrophysiology. Neurophotonics 2024; 11:033403. [PMID: 37662552 PMCID: PMC10472193 DOI: 10.1117/1.nph.11.3.033403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023]
Abstract
The brain enables adaptive behavior via the dynamic coordination of diverse neuronal signals across spatial and temporal scales: from fast action potential patterns in microcircuits to slower patterns of distributed activity in brain-wide networks. Understanding principles of multiscale dynamics requires simultaneous monitoring of signals in multiple, distributed network nodes. Combining optical and electrical recordings of brain activity is promising for collecting data across multiple scales and can reveal aspects of coordinated dynamics invisible to standard, single-modality approaches. We review recent progress in combining opto- and electrophysiology, focusing on mouse studies that shed new light on the function of single neurons by embedding their activity in the context of brain-wide activity patterns. Optical and electrical readouts can be tailored to desired scales to tackle specific questions. For example, fast dynamics in single cells or local populations recorded with multi-electrode arrays can be related to simultaneously acquired optical signals that report activity in specified subpopulations of neurons, in non-neuronal cells, or in neuromodulatory pathways. Conversely, two-photon imaging can be used to densely monitor activity in local circuits while sampling electrical activity in distant brain areas at the same time. The refinement of combined approaches will continue to reveal previously inaccessible and under-appreciated aspects of coordinated brain activity.
Collapse
Affiliation(s)
| | - Adrian Hoffmann
- University of Zurich, Brain Research Institute, Zurich, Switzerland
- University of Zurich, Neuroscience Center Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- University of Zurich, Brain Research Institute, Zurich, Switzerland
- University of Zurich, Neuroscience Center Zurich, Zurich, Switzerland
- University of Zurich, University Research Priority Program, Adaptive Brain Circuits in Development and Learning, Zurich, Switzerland
| |
Collapse
|
2
|
Akter M, Li Y. Does astrocytic L-lactate enhance cognition through myelination? Neural Regen Res 2024; 19:1167-1168. [PMID: 37905847 DOI: 10.4103/1673-5374.385872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Mastura Akter
- Department of Neuroscience, City University of Hong Kong, Hong Kong Special Administrative Region, China (Akter M, Li Y)
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China (Akter M, Li Y)
| | - Ying Li
- Department of Neuroscience, City University of Hong Kong, Hong Kong Special Administrative Region, China (Akter M, Li Y)
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China (Akter M, Li Y)
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Special Administrative Region, China (Li Y)
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Hong Kong Special Administrative Region, China (Li Y)
| |
Collapse
|
3
|
Mourtzi T, Antoniou N, Dimitriou C, Gkaravelas P, Athanasopoulou G, Kostantzo PN, Stathi O, Theodorou E, Anesti M, Matsas R, Angelatou F, Kouroupi G, Kazanis I. Enhancement of endogenous midbrain neurogenesis by microneurotrophin BNN-20 after neural progenitor grafting in a mouse model of nigral degeneration. Neural Regen Res 2024; 19:1318-1324. [PMID: 37905881 DOI: 10.4103/1673-5374.385314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 08/28/2023] [Indexed: 11/02/2023] Open
Abstract
Abstract
JOURNAL/nrgr/04.03/01300535-202406000-00036/inline-graphic1/v/2023-10-30T152229Z/r/image-tiff
We have previously shown the neuroprotective and pro-neurogenic activity of microneurotrophin BNN-20 in the substantia nigra of the “weaver” mouse, a model of progressive nigrostriatal degeneration. Here, we extended our investigation in two clinically-relevant ways. First, we assessed the effects of BNN-20 on human induced pluripotent stem cell-derived neural progenitor cells and neurons derived from healthy and parkinsonian donors. Second, we assessed if BNN-20 can boost the outcome of mouse neural progenitor cell intranigral transplantations in weaver mice, at late stages of degeneration. We found that BNN-20 has limited direct effects on cultured human induced pluripotent stem cell-derived neural progenitor cells, marginally enhancing their differentiation towards neurons and partially reversing the pathological phenotype of dopaminergic neurons generated from parkinsonian donors. In agreement, we found no effects of BNN-20 on the mouse neural progenitor cells grafted in the substantia nigra of weaver mice. However, the graft strongly induced an endogenous neurogenic response throughout the midbrain, which was significantly enhanced by the administration of microneurotrophin BNN-20. Our results provide straightforward evidence of the existence of an endogenous midbrain neurogenic system that can be specifically strengthened by BNN-20. Interestingly, the lack of major similar activity on cultured human induced pluripotent stem cell-derived neural progenitors and their progeny reveals the in vivo specificity of the aforementioned pro-neurogenic effect.
Collapse
Affiliation(s)
- Theodora Mourtzi
- Laboratory of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Nasia Antoniou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - Christina Dimitriou
- Laboratory of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Panagiotis Gkaravelas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - Georgia Athanasopoulou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - Panagiota Nti Kostantzo
- Laboratory of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Olga Stathi
- Laboratory of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Efthymia Theodorou
- Laboratory of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Maria Anesti
- Laboratory of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - Fevronia Angelatou
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - Ilias Kazanis
- Laboratory of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| |
Collapse
|
4
|
Wei LN. CRABP1-mediated non-canonical retinoic acid signaling in motor neurons and neural stem cells. Neural Regen Res 2024; 19:1169-1170. [PMID: 37905848 DOI: 10.4103/1673-5374.385866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/24/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
5
|
Jia R, Solé-Guardia G, Kiliaan AJ. Blood-brain barrier pathology in cerebral small vessel disease. Neural Regen Res 2024; 19:1233-1240. [PMID: 37905869 DOI: 10.4103/1673-5374.385864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cerebral small vessel disease is a neurological disease that affects the brain microvasculature and which is commonly observed among the elderly. Although at first it was considered innocuous, small vessel disease is nowadays regarded as one of the major vascular causes of dementia. Radiological signs of small vessel disease include small subcortical infarcts, white matter magnetic resonance imaging hyperintensities, lacunes, enlarged perivascular spaces, cerebral microbleeds, and brain atrophy; however, great heterogeneity in clinical symptoms is observed in small vessel disease patients. The pathophysiology of these lesions has been linked to multiple processes, such as hypoperfusion, defective cerebrovascular reactivity, and blood-brain barrier dysfunction. Notably, studies on small vessel disease suggest that blood-brain barrier dysfunction is among the earliest mechanisms in small vessel disease and might contribute to the development of the hallmarks of small vessel disease. Therefore, the purpose of this review is to provide a new foundation in the study of small vessel disease pathology. First, we discuss the main structural domains and functions of the blood-brain barrier. Secondly, we review the most recent evidence on blood-brain barrier dysfunction linked to small vessel disease. Finally, we conclude with a discussion on future perspectives and propose potential treatment targets and interventions.
Collapse
Affiliation(s)
- Ruxue Jia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | | | | |
Collapse
|
6
|
Chu D, Yang X, Wang J, Zhou Y, Gu JH, Miao J, Wu F, Liu F. Tau truncation in the pathogenesis of Alzheimer's disease: a narrative review. Neural Regen Res 2024; 19:1221-1232. [PMID: 37905868 DOI: 10.4103/1673-5374.385853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Alzheimer's disease is characterized by two major neuropathological hallmarks-the extracellular β-amyloid plaques and intracellular neurofibrillary tangles consisting of aggregated and hyperphosphorylated Tau protein. Recent studies suggest that dysregulation of the microtubule-associated protein Tau, especially specific proteolysis, could be a driving force for Alzheimer's disease neurodegeneration. Tau physiologically promotes the assembly and stabilization of microtubules, whereas specific truncated fragments are sufficient to induce abnormal hyperphosphorylation and aggregate into toxic oligomers, resulting in them gaining prion-like characteristics. In addition, Tau truncations cause extensive impairments to neural and glial cell functions and animal cognition and behavior in a fragment-dependent manner. This review summarizes over 60 proteolytic cleavage sites and their corresponding truncated fragments, investigates the role of specific truncations in physiological and pathological states of Alzheimer's disease, and summarizes the latest applications of strategies targeting Tau fragments in the diagnosis and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xingyue Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Jin Miao
- Laboratory of Animal Center, Nantong University, Nantong, Jiangsu Province, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
7
|
Yang Y, Liu T, Li J, Yan D, Hu Y, Wu P, Fang F, McQuillan PM, Hang W, Leng J, Hu Z. General anesthetic agents induce neurotoxicity through astrocytes. Neural Regen Res 2024; 19:1299-1307. [PMID: 37905879 DOI: 10.4103/1673-5374.385857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/09/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Neuroscientists have recognized the importance of astrocytes in regulating neurological function and their influence on the release of glial transmitters. Few studies, however, have focused on the effects of general anesthetic agents on neuroglia or astrocytes. Astrocytes can also be an important target of general anesthetic agents as they exert not only sedative, analgesic, and amnesic effects but also mediate general anesthetic-induced neurotoxicity and postoperative cognitive dysfunction. Here, we analyzed recent advances in understanding the mechanism of general anesthetic agents on astrocytes, and found that exposure to general anesthetic agents will destroy the morphology and proliferation of astrocytes, in addition to acting on the receptors on their surface, which not only affect Ca2+ signaling, inhibit the release of brain-derived neurotrophic factor and lactate from astrocytes, but are even involved in the regulation of the pro- and anti-inflammatory processes of astrocytes. These would obviously affect the communication between astrocytes as well as between astrocytes and neighboring neurons, other neuroglia, and vascular cells. In this review, we summarize how general anesthetic agents act on neurons via astrocytes, and explore potential mechanisms of action of general anesthetic agents on the nervous system. We hope that this review will provide a new direction for mitigating the neurotoxicity of general anesthetic agents.
Collapse
Affiliation(s)
- Yanchang Yang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Tiantian Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Department of Anesthesiology, Ningbo Women and Children's Hospital, Ningbo, Zhejiang Province, China
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Department of Anesthesiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang Province, China
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yuhan Hu
- Cell Biology Department, Yale University, New Haven, CT, USA
| | - Pin Wu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Fuquan Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Patrick M McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Centre, Penn State College of Medicine, Hershey, PA, USA
| | - Wenxin Hang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jianhang Leng
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
8
|
Jia Q, Li J, Guo X, Li Y, Wu Y, Peng Y, Fang Z, Zhang X. Neuroprotective effects of chaperone-mediated autophagy in neurodegenerative diseases. Neural Regen Res 2024; 19:1291-1298. [PMID: 37905878 DOI: 10.4103/1673-5374.385848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/17/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Chaperone-mediated autophagy is one of three types of autophagy and is characterized by the selective degradation of proteins. Chaperone-mediated autophagy contributes to energy balance and helps maintain cellular homeostasis, while providing nutrients and support for cell survival. Chaperone-mediated autophagy activity can be detected in almost all cells, including neurons. Owing to the extreme sensitivity of neurons to their environmental changes, maintaining neuronal homeostasis is critical for neuronal growth and survival. Chaperone-mediated autophagy dysfunction is closely related to central nervous system diseases. It has been shown that neuronal damage and cell death are accompanied by chaperone-mediated autophagy dysfunction. Under certain conditions, regulation of chaperone-mediated autophagy activity attenuates neurotoxicity. In this paper, we review the changes in chaperone-mediated autophagy in neurodegenerative diseases, brain injury, glioma, and autoimmune diseases. We also summarize the most recent research progress on chaperone-mediated autophagy regulation and discuss the potential of chaperone-mediated autophagy as a therapeutic target for central nervous system diseases.
Collapse
Affiliation(s)
- Qi Jia
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jin Li
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
- Department of Critical Care Medicine, Air Force Medical Center, Beijing, China
| | - Xiaofeng Guo
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yi Li
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - You Wu
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yuliang Peng
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Zongping Fang
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xijing Zhang
- Department of Anesthesiology and Perioperative Medicine and Department of Intensive Care Unit, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
9
|
Wu K, Liu S. Hippocampal dopamine as a key regulator for learning deficits in Parkinson's disease. Neural Regen Res 2024; 19:1193-1194. [PMID: 37905860 DOI: 10.4103/1673-5374.385860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/18/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Kun Wu
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China (Wu K, Liu S)
| | - Shuai Liu
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China (Wu K, Liu S)
- Shanghai Changning Mental Health Center, Shanghai, China (Liu S)
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai, China (Liu S)
| |
Collapse
|
10
|
Huo A, Wang J, Li Q, Li M, Qi Y, Yin Q, Luo W, Shi J, Cong Q. Molecular mechanisms underlying microglial sensing and phagocytosis in synaptic pruning. Neural Regen Res 2024; 19:1284-1290. [PMID: 37905877 DOI: 10.4103/1673-5374.385854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/03/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Microglia are the main non-neuronal cells in the central nervous system that have important roles in brain development and functional connectivity of neural circuits. In brain physiology, highly dynamic microglial processes are facilitated to sense the surrounding environment and stimuli. Once the brain switches its functional states, microglia are recruited to specific sites to exert their immune functions, including the release of cytokines and phagocytosis of cellular debris. The crosstalk of microglia between neurons, neural stem cells, endothelial cells, oligodendrocytes, and astrocytes contributes to their functions in synapse pruning, neurogenesis, vascularization, myelination, and blood-brain barrier permeability. In this review, we highlight the neuron-derived "find-me," "eat-me," and "don't eat-me" molecular signals that drive microglia in response to changes in neuronal activity for synapse refinement during brain development. This review reveals the molecular mechanism of neuron-microglia interaction in synaptic pruning and presents novel ideas for the synaptic pruning of microglia in disease, thereby providing important clues for discovery of target drugs and development of nervous system disease treatment methods targeting synaptic dysfunction.
Collapse
Affiliation(s)
- Anran Huo
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiali Wang
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Qi Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Mengqi Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Yuwan Qi
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| | - Qiao Yin
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Weifeng Luo
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jijun Shi
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qifei Cong
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University; Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
11
|
Han T, Xu Y, Sun L, Hashimoto M, Wei J. Microglial response to aging and neuroinflammation in the development of neurodegenerative diseases. Neural Regen Res 2024; 19:1241-1248. [PMID: 37905870 DOI: 10.4103/1673-5374.385845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/17/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cellular senescence and chronic inflammation in response to aging are considered to be indicators of brain aging; they have a great impact on the aging process and are the main risk factors for neurodegeneration. Reviewing the microglial response to aging and neuroinflammation in neurodegenerative diseases will help understand the importance of microglia in neurodegenerative diseases. This review describes the origin and function of microglia and focuses on the role of different states of the microglial response to aging and chronic inflammation on the occurrence and development of neurodegenerative diseases, including Alzheimer's disease, Huntington's chorea, and Parkinson's disease. This review also describes the potential benefits of treating neurodegenerative diseases by modulating changes in microglial states. Therefore, inducing a shift from the neurotoxic to neuroprotective microglial state in neurodegenerative diseases induced by aging and chronic inflammation holds promise for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Tingting Han
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yuxiang Xu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lin Sun
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, Henan Province, China
| | - Makoto Hashimoto
- Department of Basic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| |
Collapse
|
12
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CTO, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T O Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
13
|
Misrani A, Tabassum S, Wang T, Huang H, Jiang J, Diao H, Zhao Y, Huang Z, Tan S, Long C, Yang L. Vibration-reduced anxiety-like behavior relies on ameliorating abnormalities of the somatosensory cortex and medial prefrontal cortex. Neural Regen Res 2024; 19:1351-1359. [PMID: 37905885 DOI: 10.4103/1673-5374.385840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/19/2023] [Indexed: 11/02/2023] Open
Abstract
Abstract
JOURNAL/nrgr/04.03/01300535-202406000-00040/inline-graphic1/v/2023-10-30T152229Z/r/image-tiff
Tibetan singing bowls emit low-frequency sounds and produce perceptible harmonic tones and vibrations through manual tapping. The sounds the singing bowls produce have been shown to enhance relaxation and reduce anxiety. However, the underlying mechanism remains unclear. In this study, we used chronic restraint stress or sleep deprivation to establish mouse models of anxiety that exhibit anxiety-like behaviors. We then supplied treatment with singing bowls in a bottomless cage placed on the top of a cushion. We found that unlike in humans, the combination of harmonic tones and vibrations did not improve anxiety-like behaviors in mice, while individual vibration components did. Additionally, the vibration of singing bowls increased the level of N-methyl-D-aspartate receptor 1 in the somatosensory cortex and prefrontal cortex of the mice, decreased the level of γ-aminobutyric acid A (GABA) receptor α 1 subtype, reduced the level of CaMKII in the prefrontal cortex, and increased the number of GABAergic interneurons. At the same time, electrophysiological tests showed that the vibration of singing bowls significantly reduced the abnormal low-frequency gamma oscillation peak frequency in the medial prefrontal cortex caused by stress restraint pressure and sleep deprivation. Results from this study indicate that the vibration of singing bowls can alleviate anxiety-like behaviors by reducing abnormal molecular and electrophysiological events in somatosensory and medial prefrontal cortex.
Collapse
Affiliation(s)
- Afzal Misrani
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Sidra Tabassum
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Tintin Wang
- Guangzhou Hongai Cultural Development, Inc., Guangzhou, Guangdong Province, China
- Yinguo Health Management Team, Guangzhou, Guangdong Province, China
| | - Huixian Huang
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Jinxiang Jiang
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Hongjun Diao
- Guangzhou Hongai Cultural Development, Inc., Guangzhou, Guangdong Province, China
- Yinguo Health Management Team, Guangzhou, Guangdong Province, China
| | - Yanping Zhao
- College of Biophotonics, South China Normal University, Guangzhou, Guangdong Province, China
| | - Zhen Huang
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
| | - Shaohua Tan
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
| | - Cheng Long
- South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, Guangdong Province, China
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong Province, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
| |
Collapse
|
14
|
Qiu X, Yang Y, Da X, Wang Y, Chen Z, Xu C. Satellite glial cells in sensory ganglia play a wider role in chronic pain via multiple mechanisms. Neural Regen Res 2024; 19:1056-1063. [PMID: 37862208 DOI: 10.4103/1673-5374.382986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
Satellite glial cells are unique glial cells that surround the cell body of primary sensory neurons. An increasing body of evidence suggests that in the presence of inflammation and nerve damage, a significant number of satellite glial cells become activated, thus triggering a series of functional changes. This suggests that satellite glial cells are closely related to the occurrence of chronic pain. In this review, we first summarize the morphological structure, molecular markers, and physiological functions of satellite glial cells. Then, we clarify the multiple key roles of satellite glial cells in chronic pain, including gap junction hemichannel Cx43, membrane channel Pannexin1, K channel subunit 4.1, ATP, purinergic P2 receptors, and a series of additional factors and their receptors, including tumor necrosis factor, glutamate, endothelin, and bradykinin. Finally, we propose that future research should focus on the specific sorting of satellite glial cells, and identify genomic differences between physiological and pathological conditions. This review provides an important perspective for clarifying mechanisms underlying the peripheral regulation of chronic pain and will facilitate the formulation of new treatment plans for chronic pain.
Collapse
Affiliation(s)
- Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yuanzhi Yang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaoli Da
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
15
|
Chen C. TDP-43 is a key molecule accelerating development of Alzheimer's disease following traumatic brain injury. Neural Regen Res 2024; 19:955-956. [PMID: 37862186 DOI: 10.4103/1673-5374.385301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Affiliation(s)
- Chu Chen
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
16
|
Jensen HH, Olsen A. Neurological consequences of human calmodulin mutations. Neural Regen Res 2024; 19:943-944. [PMID: 37862180 DOI: 10.4103/1673-5374.385299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Affiliation(s)
- Helene H Jensen
- Medical Biotechnology, Department of Chemistry and Biosciences, Aalborg University, Aalborg, Denmark
| | - Anders Olsen
- Medical Biotechnology, Department of Chemistry and Biosciences, Aalborg University, Aalborg, Denmark
| |
Collapse
|
17
|
Singh H, Das A, Khan MM, Pourmotabbed T. New insights into the therapeutic approaches for the treatment of tauopathies. Neural Regen Res 2024; 19:1020-1026. [PMID: 37862204 DOI: 10.4103/1673-5374.385288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
Tauopathies are a group of neurological disorders, including Alzheimer's disease and frontotemporal dementia, which involve progressive neurodegeneration, cognitive deficits, and aberrant tau protein accumulation. The development of tauopathies cannot currently be stopped or slowed down by treatment measures. Given the significant contribution of tau burden in primary tauopathies and the strong association between pathogenic tau accumulation and cognitive deficits, there has been a lot of interest in creating therapies that can alleviate tau pathology and render neuroprotective effects. Recently, small molecules, immunotherapies, and gene therapy have been used to reduce the pathological tau burden and prevent neurodegeneration in animal models of tauopathies. However, the major pitfall of the current therapeutic approach is the difficulty of drugs and gene-targeting modalities to cross the blood-brain barrier and their unintended side effects. In this review, the current therapeutic strategies used for tauopathies including the use of oligonucleotide-based gene therapy approaches that have shown a promising result for the treatment of tauopathies and Alzheimer's disease in preclinical animal models, have been discussed.
Collapse
Affiliation(s)
- Himanshi Singh
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India; Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India
| | - Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center; Neuroscience Institute, University of Tennessee Health Science Center; Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tayebeh Pourmotabbed
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
18
|
Wu E, Qi D, Nizamutdinov D, Huang JH. Astrocytic calcium waves: unveiling their roles in sleep and arousal modulation. Neural Regen Res 2024; 19:984-987. [PMID: 37862199 DOI: 10.4103/1673-5374.385287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
Neuron-astrocyte interactions are vital for the brain's connectome. Understanding astrocyte activities is crucial for comprehending the complex neural network, particularly the population-level functions of neurons in different cortical states and associated behaviors in mammals. Studies on animal sleep and wakefulness have revealed distinct cortical synchrony patterns between neurons. Astrocytes, outnumbering neurons by nearly fivefold, support and regulate neuronal and synaptic function. Recent research on astrocyte activation during cortical state transitions has emphasized the influence of norepinephrine as a neurotransmitter and calcium waves as key components of ion channel signaling. This summary focuses on a few recent studies investigating astrocyte-neuron interactions in mouse models during sleep, wakefulness, and arousal levels, exploring the involvement of noradrenaline signaling, ion channels, and glutamatergic signaling in different cortical states. These findings highlight the significant impact of astrocytes on large-scale neuronal networks, influencing brain activity and responsiveness. Targeting astrocytic signaling pathways shows promise for treating sleep disorders and arousal dysregulation. More research is needed to understand astrocytic calcium signaling in different brain regions and its implications for dysregulated brain states, requiring future human studies to comprehensively investigate neuron-astrocyte interactions and pave the way for therapeutic interventions in sleep- and arousal-related disorders.
Collapse
Affiliation(s)
- Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple; Texas A&M University School of Medicine, Temple; Texas A&M University School of Pharmacy, College Station; LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, the University of Texas at Austin, Austin; Department of Neurosurgery, Baylor College of Medicine, Temple, TX, USA
| | - Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, USA
| | - Damir Nizamutdinov
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health; Texas A&M University School of Medicine, Temple, TX, USA
| | - Jason H Huang
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health; Texas A&M University School of Medicine; Department of Neurosurgery, Baylor College of Medicine, Temple, TX, USA
| |
Collapse
|
19
|
Hu Y, Chen W, Wei C, Jiang S, Li S, Wang X, Xu R. Pathological mechanisms of amyotrophic lateral Sclerosis. Neural Regen Res 2024; 19:1036-1044. [PMID: 37862206 DOI: 10.4103/1673-5374.382985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
Amyotrophic lateral sclerosis refers to a neurodegenerative disease involving the motor system, the cause of which remains unexplained despite several years of research. Thus, the journey to understanding or treating amyotrophic lateral sclerosis is still a long one. According to current research, amyotrophic lateral sclerosis is likely not due to a single factor but rather to a combination of mechanisms mediated by complex interactions between molecular and genetic pathways. The progression of the disease involves multiple cellular processes and the interaction between different complex mechanisms makes it difficult to identify the causative factors of amyotrophic lateral sclerosis. Here, we review the most common amyotrophic lateral sclerosis-associated pathogenic genes and the pathways involved in amyotrophic lateral sclerosis, as well as summarize currently proposed potential mechanisms responsible for amyotrophic lateral sclerosis disease and their evidence for involvement in amyotrophic lateral sclerosis. In addition, we discuss current emerging strategies for the treatment of amyotrophic lateral sclerosis. Studying the emergence of these new therapies may help to further our understanding of the pathogenic mechanisms of the disease.
Collapse
Affiliation(s)
- Yushu Hu
- Department of Clinical Medicine, Nanchang University; Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province, China
| | - Wenzhi Chen
- Department of Clinical Medicine, Nanchang University; Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province, China
| | - Caihui Wei
- Department of Clinical Medicine, Nanchang University; Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province, China
| | - Shishi Jiang
- Department of Clinical Medicine, Nanchang University; Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province, China
| | - Shu Li
- Department of Clinical Medicine, Nanchang University; Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province, China
| | - Xinxin Wang
- Department of Clinical Medicine, Nanchang University; Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province, China
| | - Renshi Xu
- Department of Clinical Medicine, Nanchang University; Department of Neurology, Jiangxi Provincial People's Hospital; Department of Neurology, The First Affiliated Hospital of Nanchang Medical College; The Clinical College of Nanchang Medical College, Nanchang, Jiangxi Province, China
| |
Collapse
|
20
|
Liu T, Ma Z, Liu L, Pei Y, Wu Q, Xu S, Liu Y, Ding N, Guan Y, Zhang Y, Chen X. Conditioned medium from human dental pulp stem cells treats spinal cord injury by inhibiting microglial pyroptosis. Neural Regen Res 2024; 19:1105-1111. [PMID: 37862215 DOI: 10.4103/1673-5374.385309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
Human dental pulp stem cell transplantation has been shown to be an effective therapeutic strategy for spinal cord injury. However, whether the human dental pulp stem cell secretome can contribute to functional recovery after spinal cord injury remains unclear. In the present study, we established a rat model of spinal cord injury based on impact injury from a dropped weight and then intraperitoneally injected the rats with conditioned medium from human dental pulp stem cells. We found that the conditioned medium effectively promoted the recovery of sensory and motor functions in rats with spinal cord injury, decreased expression of the microglial pyroptosis markers NLRP3, GSDMD, caspase-1, and interleukin-1β, promoted axonal and myelin regeneration, and inhibited the formation of glial scars. In addition, in a lipopolysaccharide-induced BV2 microglia model, conditioned medium from human dental pulp stem cells protected cells from pyroptosis by inhibiting the NLRP3/caspase-1/interleukin-1β pathway. These results indicate that conditioned medium from human dental pulp stem cells can reduce microglial pyroptosis by inhibiting the NLRP3/caspase-1/interleukin-1β pathway, thereby promoting the recovery of neurological function after spinal cord injury. Therefore, conditioned medium from human dental pulp stem cells may become an alternative therapy for spinal cord injury.
Collapse
Affiliation(s)
- Tao Liu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Ziqian Ma
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Liang Liu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yilun Pei
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Qichao Wu
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Songjie Xu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yadong Liu
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Nan Ding
- Department of Stomatology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine; Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yan Zhang
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xueming Chen
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Zhang Y, Tian K, Wei W, Mi W, Lu F, Liu Z, Zhu Q, Zhang X, Geng P, Qiu J, Song Y, Zha D. Translocation of telomerase reverse transcriptase coincided with ATP release in postnatal cochlear supporting cells. Neural Regen Res 2024; 19:1119-1125. [PMID: 37862217 DOI: 10.4103/1673-5374.382862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Abstract
The spontaneous bursts of electrical activity in the developing auditory system are derived from the periodic release of adenosine triphosphate (ATP) by supporting cells in the Kölliker's organ. However, the mechanisms responsible for initiating spontaneous ATP release have not been determined. Our previous study revealed that telomerase reverse transcriptase (TERT) is expressed in the basilar membrane during the first postnatal week. Its role in cochlear development remains unclear. In this study, we investigated the expression and role of TERT in postnatal cochlea supporting cells. Our results revealed that in postnatal cochlear Kölliker's organ supporting cells, TERT shifts from the nucleus into the cytoplasm over time. We found that the TERT translocation tendency in postnatal cochlear supporting cells in vitro coincided with that observed in vivo. Further analysis showed that TERT in the cytoplasm was mainly located in mitochondria in the absence of oxidative stress or apoptosis, suggesting that TERT in mitochondria plays roles other than antioxidant or anti-apoptotic functions. We observed increased ATP synthesis, release and activation of purine signaling systems in supporting cells during the first 10 postnatal days. The phenomenon that TERT translocation coincided with changes in ATP synthesis, release and activation of the purine signaling system in postnatal cochlear supporting cells suggested that TERT may be involved in regulating ATP release and activation of the purine signaling system. Our study provides a new research direction for exploring the spontaneous electrical activity of the cochlea during the early postnatal period.
Collapse
Affiliation(s)
- Yukai Zhang
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Keyong Tian
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wei Wei
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wenjuan Mi
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Fei Lu
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Zhenzhen Liu
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Qingwen Zhu
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xinyu Zhang
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Panling Geng
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jianhua Qiu
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yongli Song
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Dingjun Zha
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
22
|
Coleman LG. From the dust: extracellular vesicles as regulators of development and neuroregeneration. Neural Regen Res 2024; 19:933-934. [PMID: 37862175 DOI: 10.4103/1673-5374.382243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023] Open
Affiliation(s)
- Leon G Coleman
- The University of North Carolina at Chapel Hill, School of Medicine, Department of Pharmacology, Bowles Center for Alcohol Studies, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Gao M, Dong Q, Yang Z, Zou D, Han Y, Chen Z, Xu R. Long non-coding RNA H19 regulates neurogenesis of induced neural stem cells in a mouse model of closed head injury. Neural Regen Res 2024; 19:872-880. [PMID: 37843223 PMCID: PMC10664125 DOI: 10.4103/1673-5374.382255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/08/2023] [Accepted: 07/04/2023] [Indexed: 10/17/2023] Open
Abstract
Stem cell-based therapies have been proposed as a potential treatment for neural regeneration following closed head injury. We previously reported that induced neural stem cells exert beneficial effects on neural regeneration via cell replacement. However, the neural regeneration efficiency of induced neural stem cells remains limited. In this study, we explored differentially expressed genes and long non-coding RNAs to clarify the mechanism underlying the neurogenesis of induced neural stem cells. We found that H19 was the most downregulated neurogenesis-associated lncRNA in induced neural stem cells compared with induced pluripotent stem cells. Additionally, we demonstrated that H19 levels in induced neural stem cells were markedly lower than those in induced pluripotent stem cells and were substantially higher than those in induced neural stem cell-derived neurons. We predicted the target genes of H19 and discovered that H19 directly interacts with miR-325-3p, which directly interacts with Ctbp2 in induced pluripotent stem cells and induced neural stem cells. Silencing H19 or Ctbp2 impaired induced neural stem cell proliferation, and miR-325-3p suppression restored the effect of H19 inhibition but not the effect of Ctbp2 inhibition. Furthermore, H19 silencing substantially promoted the neural differentiation of induced neural stem cells and did not induce apoptosis of induced neural stem cells. Notably, silencing H19 in induced neural stem cell grafts markedly accelerated the neurological recovery of closed head injury mice. Our results reveal that H19 regulates the neurogenesis of induced neural stem cells. H19 inhibition may promote the neural differentiation of induced neural stem cells, which is closely associated with neurological recovery following closed head injury.
Collapse
Affiliation(s)
- Mou Gao
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Qin Dong
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Zhijun Yang
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Dan Zou
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Yajuan Han
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Zhanfeng Chen
- Zhongsai Stem Cell Genetic Engineering Co., Ltd., Sanmenxia, Henan Province, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| |
Collapse
|
24
|
Amar Kumar P, Dulin JN. Implications of regional identity for neural stem and progenitor cell transplantation in the injured or diseased nervous system. Neural Regen Res 2024; 19:715-716. [PMID: 37843199 PMCID: PMC10664136 DOI: 10.4103/1673-5374.382236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/08/2023] [Accepted: 06/29/2023] [Indexed: 10/17/2023] Open
Affiliation(s)
| | - Jennifer N. Dulin
- Department of Biology, Texas A&M University, College Station, TX, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| |
Collapse
|
25
|
Chen Y, Kuang J, Niu Y, Zhu H, Chen X, So KF, Xu A, Shi L. Multiple factors to assist human-derived induced pluripotent stem cells to efficiently differentiate into midbrain dopaminergic neurons. Neural Regen Res 2024; 19:908-914. [PMID: 37843228 PMCID: PMC10664128 DOI: 10.4103/1673-5374.378203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/04/2023] [Accepted: 06/03/2023] [Indexed: 10/17/2023] Open
Abstract
Midbrain dopaminergic neurons play an important role in the etiology of neurodevelopmental and neurodegenerative diseases. They also represent a potential source of transplanted cells for therapeutic applications. In vitro differentiation of functional midbrain dopaminergic neurons provides an accessible platform to study midbrain neuronal dysfunction and can be used to examine obstacles to dopaminergic neuronal development. Emerging evidence and impressive advances in human induced pluripotent stem cells, with tuned neural induction and differentiation protocols, makes the production of induced pluripotent stem cell-derived dopaminergic neurons feasible. Using SB431542 and dorsomorphin dual inhibitor in an induced pluripotent stem cell-derived neural induction protocol, we obtained multiple subtypes of neurons, including 20% tyrosine hydroxylase-positive dopaminergic neurons. To obtain more dopaminergic neurons, we next added sonic hedgehog (SHH) and fibroblast growth factor 8 (FGF8) on day 8 of induction. This increased the proportion of dopaminergic neurons, up to 75% tyrosine hydroxylase-positive neurons, with 15% tyrosine hydroxylase and forkhead box protein A2 (FOXA2) co-expressing neurons. We further optimized the induction protocol by applying the small molecule inhibitor, CHIR99021 (CHIR).This helped facilitate the generation of midbrain dopaminergic neurons, and we obtained 31-74% midbrain dopaminergic neurons based on tyrosine hydroxylase and FOXA2 staining. Thus, we have established three induction protocols for dopaminergic neurons. Based on tyrosine hydroxylase and FOXA2 immunostaining analysis, the CHIR, SHH, and FGF8 combined protocol produces a much higher proportion of midbrain dopaminergic neurons, which could be an ideal resource for tackling midbrain-related diseases.
Collapse
Affiliation(s)
- Yalan Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Junxin Kuang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Yimei Niu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Hongyao Zhu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Xiaoxia Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Lingling Shi
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
- Department of Psychiatry, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
26
|
Frank MG, Baratta MV. Use of an immunocapture device to detect cytokine release in discrete brain regions. Neural Regen Res 2024; 19:703-704. [PMID: 37843193 PMCID: PMC10664115 DOI: 10.4103/1673-5374.382237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 10/17/2023] Open
Affiliation(s)
- Matthew G. Frank
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Michael V. Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
27
|
Jin M, Wang S, Gao X, Zou Z, Hirotsune S, Sun L. Pathological and physiological functional cross-talks of α-synuclein and tau in the central nervous system. Neural Regen Res 2024; 19:855-862. [PMID: 37843221 PMCID: PMC10664117 DOI: 10.4103/1673-5374.382231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/05/2023] [Accepted: 07/12/2023] [Indexed: 10/17/2023] Open
Abstract
α-Synuclein and tau are abundant multifunctional brain proteins that are mainly expressed in the presynaptic and axonal compartments of neurons, respectively. Previous works have revealed that intracellular deposition of α-synuclein and/or tau causes many neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. Despite intense investigation, the normal physiological functions and roles of α-synuclein and tau are still unclear, owing to the fact that mice with knockout of either of these proteins do not present apparent phenotypes. Interestingly, the co-occurrence of α-synuclein and tau aggregates was found in post-mortem brains with synucleinopathies and tauopathies, some of which share similarities in clinical manifestations. Furthermore, the direct interaction of α-synuclein with tau is considered to promote the fibrillization of each of the proteins in vitro and in vivo. On the other hand, our recent findings have revealed that α-synuclein and tau are cooperatively involved in brain development in a stage-dependent manner. These findings indicate strong cross-talk between the two proteins in physiology and pathology. In this review, we provide a summary of the recent findings on the functional roles of α-synuclein and tau in the physiological conditions and pathogenesis of neurodegenerative diseases. A deep understanding of the interplay between α-synuclein and tau in physiological and pathological conditions might provide novel targets for clinical diagnosis and therapeutic strategies to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Mingyue Jin
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Shengming Wang
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Xiaodie Gao
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Zhenyou Zou
- Department of Scientific Research, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou, Guangxi Zhuang Autonomous Region, China
| | - Shinji Hirotsune
- Department of Genetic Disease Research, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Liyuan Sun
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
28
|
Wang X, Hu Y, Xu R. The pathogenic mechanism of TAR DNA-binding protein 43 (TDP-43) in amyotrophic lateral sclerosis. Neural Regen Res 2024; 19:800-806. [PMID: 37843214 PMCID: PMC10664110 DOI: 10.4103/1673-5374.382233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 10/17/2023] Open
Abstract
The onset of amyotrophic lateral sclerosis is usually characterized by focal death of both upper and/or lower motor neurons occurring in the motor cortex, basal ganglia, brainstem, and spinal cord, and commonly involves the muscles of the upper and/or lower extremities, and the muscles of the bulbar and/or respiratory regions. However, as the disease progresses, it affects the adjacent body regions, leading to generalized muscle weakness, occasionally along with memory, cognitive, behavioral, and language impairments; respiratory dysfunction occurs at the final stage of the disease. The disease has a complicated pathophysiology and currently, only riluzole, edaravone, and phenylbutyrate/taurursodiol are licensed to treat amyotrophic lateral sclerosis in many industrialized countries. The TAR DNA-binding protein 43 inclusions are observed in 97% of those diagnosed with amyotrophic lateral sclerosis. This review provides a preliminary overview of the potential effects of TAR DNA-binding protein 43 in the pathogenesis of amyotrophic lateral sclerosis, including the abnormalities in nucleoplasmic transport, RNA function, post-translational modification, liquid-liquid phase separation, stress granules, mitochondrial dysfunction, oxidative stress, axonal transport, protein quality control system, and non-cellular autonomous functions (e.g., glial cell functions and prion-like propagation).
Collapse
Affiliation(s)
- Xinxin Wang
- Medical College of Nanchang University, Nanchang, Jiangxi Province, China
- Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, The Clinical College of Nanchang Medical College, Nanchang, Jiangxi Province, China
| | - Yushu Hu
- Medical College of Nanchang University, Nanchang, Jiangxi Province, China
- Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, The Clinical College of Nanchang Medical College, Nanchang, Jiangxi Province, China
| | - Renshi Xu
- Medical College of Nanchang University, Nanchang, Jiangxi Province, China
- Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, The Clinical College of Nanchang Medical College, Nanchang, Jiangxi Province, China
| |
Collapse
|
29
|
Cao P, Li J, Liu Z, Liang G. Current controversies in glia-to-neuron conversion therapy in neurodegenerative diseases. Neural Regen Res 2024; 19:723-724. [PMID: 37843203 PMCID: PMC10664116 DOI: 10.4103/1673-5374.382251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 10/17/2023] Open
Affiliation(s)
- Peng Cao
- Department of Neurosurgery, General Hospital of the Northern Theater Command of Chinese People’s Liberation Army, Shenyang, Liaoning Province, China
| | - Jianan Li
- Department of Neurosurgery, General Hospital of the Northern Theater Command of Chinese People’s Liberation Army, Shenyang, Liaoning Province, China
| | - Zhuxi Liu
- Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Guobiao Liang
- Department of Neurosurgery, General Hospital of the Northern Theater Command of Chinese People’s Liberation Army, Shenyang, Liaoning Province, China
| |
Collapse
|
30
|
Hashimoto S, Nagoshi N, Nakamura M, Okano H. Regenerative medicine strategies for chronic complete spinal cord injury. Neural Regen Res 2024; 19:818-824. [PMID: 37843217 PMCID: PMC10664101 DOI: 10.4103/1673-5374.382230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/30/2023] [Accepted: 06/27/2023] [Indexed: 10/17/2023] Open
Abstract
Spinal cord injury is a condition in which the parenchyma of the spinal cord is damaged by trauma or various diseases. While rapid progress has been made in regenerative medicine for spinal cord injury that was previously untreatable, most research in this field has focused on the early phase of incomplete injury. However, the majority of patients have chronic severe injuries; therefore, treatments for these situations are of fundamental importance. The reason why the treatment of complete spinal cord injury has not been studied is that, unlike in the early stage of incomplete spinal cord injury, there are various inhibitors of neural regeneration. Thus, we assumed that it is difficult to address all conditions with a single treatment in chronic complete spinal cord injury and that a combination of several treatments is essential to target severe pathologies. First, we established a combination therapy of cell transplantation and drug-releasing scaffolds, which contributes to functional recovery after chronic complete transection spinal cord injury, but we found that functional recovery was limited and still needs further investigation. Here, for the further development of the treatment of chronic complete spinal cord injury, we review the necessary approaches to the different pathologies based on our findings and the many studies that have been accumulated to date and discuss, with reference to the literature, which combination of treatments is most effective in achieving functional recovery.
Collapse
Affiliation(s)
- Shogo Hashimoto
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Narihito Nagoshi
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
King PH. Skeletal muscle as a molecular and cellular biomarker of disease progression in amyotrophic lateral sclerosis: a narrative review. Neural Regen Res 2024; 19:747-753. [PMID: 37843208 PMCID: PMC10664124 DOI: 10.4103/1673-5374.382226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/27/2023] [Accepted: 07/19/2023] [Indexed: 10/17/2023] Open
Abstract
Amyotrophic lateral sclerosis is a fatal multisystemic neurodegenerative disease with motor neurons being a primary target. Although progressive weakness is a hallmark feature of amyotrophic lateral sclerosis, there is considerable heterogeneity, including clinical presentation, progression, and the underlying triggers for disease initiation. Based on longitudinal studies with families harboring amyotrophic lateral sclerosis-associated gene mutations, it has become apparent that overt disease is preceded by a prodromal phase, possibly in years, where compensatory mechanisms delay symptom onset. Since 85-90% of amyotrophic lateral sclerosis is sporadic, there is a strong need for identifying biomarkers that can detect this prodromal phase as motor neurons have limited capacity for regeneration. Current Food and Drug Administration-approved therapies work by slowing the degenerative process and are most effective early in the disease. Skeletal muscle, including the neuromuscular junction, manifests abnormalities at the earliest stages of the disease, before motor neuron loss, making it a promising source for identifying biomarkers of the prodromal phase. The accessibility of muscle through biopsy provides a lens into the distal motor system at earlier stages and in real time. The advent of "omics" technology has led to the identification of numerous dysregulated molecules in amyotrophic lateral sclerosis muscle, ranging from coding and non-coding RNAs to proteins and metabolites. This technology has opened the door for identifying biomarkers of disease activity and providing insight into disease mechanisms. A major challenge is correlating the myriad of dysregulated molecules with clinical or histological progression and understanding their relevance to presymptomatic phases of disease. There are two major goals of this review. The first is to summarize some of the biomarkers identified in human amyotrophic lateral sclerosis muscle that have a clinicopathological correlation with disease activity, evidence of a similar dysregulation in the SOD1G93A mouse during presymptomatic stages, and evidence of progressive change during disease progression. The second goal is to review the molecular pathways these biomarkers reflect and their potential role in mitigating or promoting disease progression, and as such, their potential as therapeutic targets in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Peter H. King
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL, USA; Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| |
Collapse
|
32
|
Liu X, Liu Y, Liu J, Zhang H, Shan C, Guo Y, Gong X, Cui M, Li X, Tang M. Correlation between the gut microbiome and neurodegenerative diseases: a review of metagenomics evidence. Neural Regen Res 2024; 19:833-845. [PMID: 37843219 PMCID: PMC10664138 DOI: 10.4103/1673-5374.382223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/19/2023] [Accepted: 06/17/2023] [Indexed: 10/17/2023] Open
Abstract
A growing body of evidence suggests that the gut microbiota contributes to the development of neurodegenerative diseases via the microbiota-gut-brain axis. As a contributing factor, microbiota dysbiosis always occurs in pathological changes of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. High-throughput sequencing technology has helped to reveal that the bidirectional communication between the central nervous system and the enteric nervous system is facilitated by the microbiota's diverse microorganisms, and for both neuroimmune and neuroendocrine systems. Here, we summarize the bioinformatics analysis and wet-biology validation for the gut metagenomics in neurodegenerative diseases, with an emphasis on multi-omics studies and the gut virome. The pathogen-associated signaling biomarkers for identifying brain disorders and potential therapeutic targets are also elucidated. Finally, we discuss the role of diet, prebiotics, probiotics, postbiotics and exercise interventions in remodeling the microbiome and reducing the symptoms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu Province, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Chaofan Shan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | |
|