1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Wu H, Dong L, Jin S, Zhao Y, Zhu L. Innovative gene delivery systems for retinal disease therapy. Neural Regen Res 2026; 21:542-552. [PMID: 39665817 DOI: 10.4103/nrr.nrr-d-24-00797] [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: 08/13/2024] [Accepted: 11/10/2024] [Indexed: 12/13/2024] Open
Abstract
The human retina, a complex and highly specialized structure, includes multiple cell types that work synergistically to generate and transmit visual signals. However, genetic predisposition or age-related degeneration can lead to retinal damage that severely impairs vision or causes blindness. Treatment options for retinal diseases are limited, and there is an urgent need for innovative therapeutic strategies. Cell and gene therapies are promising because of the efficacy of delivery systems that transport therapeutic genes to targeted retinal cells. Gene delivery systems hold great promise for treating retinal diseases by enabling the targeted delivery of therapeutic genes to affected cells or by converting endogenous cells into functional ones to facilitate nerve regeneration, potentially restoring vision. This review focuses on two principal categories of gene delivery vectors used in the treatment of retinal diseases: viral and non-viral systems. Viral vectors, including lentiviruses and adeno-associated viruses, exploit the innate ability of viruses to infiltrate cells, which is followed by the introduction of therapeutic genetic material into target cells for gene correction. Lentiviruses can accommodate exogenous genes up to 8 kb in length, but their mechanism of integration into the host genome presents insertion mutation risks. Conversely, adeno-associated viruses are safer, as they exist as episomes in the nucleus, yet their limited packaging capacity constrains their application to a narrower spectrum of diseases, which necessitates the exploration of alternative delivery methods. In parallel, progress has also occurred in the development of novel non-viral delivery systems, particularly those based on liposomal technology. Manipulation of the ratios of hydrophilic and hydrophobic molecules within liposomes and the development of new lipid formulations have led to the creation of advanced non-viral vectors. These innovative systems include solid lipid nanoparticles, polymer nanoparticles, dendrimers, polymeric micelles, and polymeric nanoparticles. Compared with their viral counterparts, non-viral delivery systems offer markedly enhanced loading capacities that enable the direct delivery of nucleic acids, mRNA, or protein molecules into cells. This bypasses the need for DNA transcription and processing, which significantly enhances therapeutic efficiency. Nevertheless, the immunogenic potential and accumulation toxicity associated with non-viral particulate systems necessitates continued optimization to reduce adverse effects in vivo . This review explores the various delivery systems for retinal therapies and retinal nerve regeneration, and details the characteristics, advantages, limitations, and clinical applications of each vector type. By systematically outlining these factors, our goal is to guide the selection of the optimal delivery tool for a specific retinal disease, which will enhance treatment efficacy and improve patient outcomes while paving the way for more effective and targeted therapeutic interventions.
Collapse
Affiliation(s)
- Hongguang Wu
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
3
|
Duan Y, Yang F, Zhang Y, Zhang M, Shi Y, Lang Y, Sun H, Wang X, Jin H, Kang X. Role of mitophagy in spinal cord ischemia-reperfusion injury. Neural Regen Res 2026; 21:598-611. [PMID: 39665804 DOI: 10.4103/nrr.nrr-d-24-00668] [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: 06/18/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024] Open
Abstract
Spinal cord ischemia-reperfusion injury, a severe form of spinal cord damage, can lead to sensory and motor dysfunction. This injury often occurs after traumatic events, spinal cord surgeries, or thoracoabdominal aortic surgeries. The unpredictable nature of this condition, combined with limited treatment options, poses a significant burden on patients, their families, and society. Spinal cord ischemia-reperfusion injury leads to reduced neuronal regenerative capacity and complex pathological processes. In contrast, mitophagy is crucial for degrading damaged mitochondria, thereby supporting neuronal metabolism and energy supply. However, while moderate mitophagy can be beneficial in the context of spinal cord ischemia-reperfusion injury, excessive mitophagy may be detrimental. Therefore, this review aims to investigate the potential mechanisms and regulators of mitophagy involved in the pathological processes of spinal cord ischemia-reperfusion injury. The goal is to provide a comprehensive understanding of recent advancements in mitophagy related to spinal cord ischemia-reperfusion injury and clarify its potential clinical applications.
Collapse
Affiliation(s)
- Yanni Duan
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Fengguang Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yibao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Mingtao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yujun Shi
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yun Lang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongli Sun
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xin Wang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongyun Jin
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xuewen Kang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
4
|
Wen J, Tang H, Tian M, Wang L, Yang Q, Zhao Y, Li X, Ren Y, Wang J, Zhou L, Tan Y, Wu H, Cai X, Wang Y, Cao H, Xu J, Yang Q. Fibrotic scar formation after cerebral ischemic stroke: Targeting the Sonic hedgehog signaling pathway for scar reduction. Neural Regen Res 2026; 21:756-768. [PMID: 40183351 DOI: 10.4103/nrr.nrr-d-24-00999] [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: 08/28/2024] [Accepted: 12/30/2024] [Indexed: 04/05/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00044/figure1/v/2025-05-05T160104Z/r/image-tiff Recent studies have shown that fibrotic scar formation following cerebral ischemic injury has varying effects depending on the microenvironment. However, little is known about how fibrosis is induced and regulated after cerebral ischemic injury. Sonic hedgehog signaling participates in fibrosis in the heart, liver, lung, and kidney. Whether Shh signaling modulates fibrotic scar formation after cerebral ischemic stroke and the underlying mechanisms are unclear. In this study, we found that Sonic Hedgehog expression was upregulated in patients with acute ischemic stroke and in a middle cerebral artery occlusion/reperfusion injury rat model. Both Sonic hedgehog and Mitofusin 2 showed increased expression in the middle cerebral artery occlusion rat model and in vitro fibrosis cell model induced by transforming growth factor-beta 1. Activation of the Sonic hedgehog signaling pathway enhanced the expression of phosphorylated Smad 3 and Mitofusin 2 proteins, promoted the formation of fibrotic scars, protected synapses or promoted synaptogenesis, alleviated neurological deficits following middle cerebral artery occlusion/reperfusion injury, reduced cell apoptosis, facilitated the transformation of meninges fibroblasts into myofibroblasts, and enhanced the proliferation and migration of meninges fibroblasts. The Smad3 phosphorylation inhibitor SIS3 reversed the effects induced by Sonic hedgehog signaling pathway activation. Bioinformatics analysis revealed significant correlations between Sonic hedgehog and Smad3, between Sonic hedgehog and Mitofusin 2, and between Smad3 and Mitofusin 2. These findings suggest that Sonic hedgehog signaling may influence Mitofusin 2 expression by regulating Smad3 phosphorylation, thereby modulating the formation of early fibrotic scars following cerebral ischemic stroke and affecting prognosis. The Sonic Hedgehog signaling pathway may serve as a new therapeutic target for stroke treatment.
Collapse
Affiliation(s)
- Jun Wen
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Tang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingfen Tian
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Wang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghuan Yang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Zhao
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuemei Li
- Department of Neurology, Second People's Hospital of Chongqing Banan District, Chongqing, China
| | - Yu Ren
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiani Wang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhou
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjun Tan
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyun Wu
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinrui Cai
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yilin Wang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Neurosurgery, Third Hospital of Mianyang, Mianyang, Sichuan Province, China
| | - Jianfeng Xu
- Department of Neurosurgery, Third Hospital of Mianyang, Mianyang, Sichuan Province, China
| | - Qin Yang
- Department of Neurology, The Frist Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Cao H, Shang L, Hu D, Huang J, Wang Y, Li M, Song Y, Yang Q, Luo Y, Wang Y, Cai X, Liu J. Neuromodulation techniques for modulating cognitive function: Enhancing stimulation precision and intervention effects. Neural Regen Res 2026; 21:491-501. [PMID: 39665818 DOI: 10.4103/nrr.nrr-d-24-00836] [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: 09/10/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024] Open
Abstract
Neuromodulation techniques effectively intervene in cognitive function, holding considerable scientific and practical value in fields such as aerospace, medicine, life sciences, and brain research. These techniques utilize electrical stimulation to directly or indirectly target specific brain regions, modulating neural activity and influencing broader brain networks, thereby regulating cognitive function. Regulating cognitive function involves an understanding of aspects such as perception, learning and memory, attention, spatial cognition, and physical function. To enhance the application of cognitive regulation in the general population, this paper reviews recent publications from the Web of Science to assess the advancements and challenges of invasive and non-invasive stimulation methods in modulating cognitive functions. This review covers various neuromodulation techniques for cognitive intervention, including deep brain stimulation, vagus nerve stimulation, and invasive methods using microelectrode arrays. The non-invasive techniques discussed include transcranial magnetic stimulation, transcranial direct current stimulation, transcranial alternating current stimulation, transcutaneous electrical acupoint stimulation, and time interference stimulation for activating deep targets. Invasive stimulation methods, which are ideal for studying the pathogenesis of neurological diseases, tend to cause greater trauma and have been less researched in the context of cognitive function regulation. Non-invasive methods, particularly newer transcranial stimulation techniques, are gentler and more appropriate for regulating cognitive functions in the general population. These include transcutaneous acupoint electrical stimulation using acupoints and time interference methods for activating deep targets. This paper also discusses current technical challenges and potential future breakthroughs in neuromodulation technology. It is recommended that neuromodulation techniques be combined with neural detection methods to better assess their effects and improve the accuracy of non-invasive neuromodulation. Additionally, researching closed-loop feedback neuromodulation methods is identified as a promising direction for future development.
Collapse
Affiliation(s)
- Hanwen Cao
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Li Shang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Deheng Hu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Jianbing Huang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yu Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Ming Li
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Qianzi Yang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Liu Z, Guo Y, Zhang Y, Gao Y, Ning B. Metabolic reprogramming of astrocytes: Emerging roles of lactate. Neural Regen Res 2026; 21:421-432. [PMID: 39688570 DOI: 10.4103/nrr.nrr-d-24-00776] [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: 07/16/2024] [Accepted: 10/25/2024] [Indexed: 12/18/2024] Open
Abstract
Lactate serves as a key energy metabolite in the central nervous system, facilitating essential brain functions, including energy supply, signaling, and epigenetic modulation. Moreover, it links epigenetic modifications with metabolic reprogramming. Nonetheless, the specific mechanisms and roles of this connection in astrocytes remain unclear. Therefore, this review aims to explore the role and specific mechanisms of lactate in the metabolic reprogramming of astrocytes in the central nervous system. The close relationship between epigenetic modifications and metabolic reprogramming was discussed. Therapeutic strategies for targeting metabolic reprogramming in astrocytes in the central nervous system were also outlined to guide future research in central nervous system diseases. In the nervous system, lactate plays an essential role. However, its mechanism of action as a bridge between metabolic reprogramming and epigenetic modifications in the nervous system requires future investigation. The involvement of lactate in epigenetic modifications is currently a hot research topic, especially in lactylation modification, a key determinant in this process. Lactate also indirectly regulates various epigenetic modifications, such as N6-methyladenosine, acetylation, ubiquitination, and phosphorylation modifications, which are closely linked to several neurological disorders. In addition, exploring the clinical applications and potential therapeutic strategies of lactic acid provides new insights for future neurological disease treatments.
Collapse
Affiliation(s)
- Zeyu Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yijian Guo
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yulei Gao
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Bin Ning
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
7
|
Yue Q, Li S, Lei CL, Wan H, Zhang Z, Hoi MPM. Insights into the transcriptomic heterogeneity of brain endothelial cells in normal aging and Alzheimer's disease. Neural Regen Res 2026; 21:569-576. [PMID: 39688567 DOI: 10.4103/nrr.nrr-d-24-00695] [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: 06/23/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Drug development for Alzheimer's disease is extremely challenging, as demonstrated by the repeated failures of amyloid-β-targeted therapeutics and the controversies surrounding the amyloid-β cascade hypothesis. More recently, advances in the development of Lecanemab, an anti-amyloid-β monoclonal antibody, have shown positive results in reducing brain A burden and slowing cognitive decline in patients with early-stage Alzheimer's disease in the Phase III clinical trial (Clarity Alzheimer's disease). Despite these promising results, side effects such as amyloid-related imaging abnormalities (ARIA) may limit its usage. ARIA can manifest as ARIA-E (cerebral edema or effusions) and ARIA-H (microhemorrhages or superficial siderosis) and is thought to be caused by increased vascular permeability due to inflammatory responses, leading to leakages of blood products and protein-rich fluid into brain parenchyma. Endothelial dysfunction is an early pathological feature of Alzheimer's disease, and the blood-brain barrier becomes increasingly leaky as the disease progresses. In addition, APOE4, the strongest genetic risk factor for Alzheimer's disease, is associated with higher vascular amyloid burden, increased ARIA incidence, and accelerated blood-brain barrier disruptions. These interconnected vascular abnormalities highlight the importance of vascular contributions to the pathophysiology of Alzheimer's disease. Here, we will closely examine recent research evaluating the heterogeneity of brain endothelial cells in the microvasculature of different brain regions and their relationships with Alzheimer's disease progression.
Collapse
Affiliation(s)
- Qian Yue
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Heyuan, Guangdong Province, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao Special Administrative Region, China
| | - Shang Li
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Chon Lok Lei
- Department of Biological Sciences, Faculty of Health Sciences, University of Macau, Macao Special Administrative Region, China
| | - Huaibin Wan
- The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Heyuan, Guangdong Province, China
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University, Guangzhou, Guangdong Province, China
- Guangdong-Hong Kong-Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University College of Pharmacy, Guangzhou, Guangdong Province, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University College of Pharmacy, Guangzhou, Guangdong Province, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao Special Administrative Region, China
| |
Collapse
|
8
|
Wu S, Chen J. Is age-related myelinodegenerative change an initial risk factor of neurodegenerative diseases? Neural Regen Res 2026; 21:648-658. [PMID: 40326982 DOI: 10.4103/nrr.nrr-d-24-00848] [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: 07/29/2024] [Accepted: 11/25/2024] [Indexed: 05/07/2025] Open
Abstract
Myelination, the continuous ensheathment of neuronal axons, is a lifelong process in the nervous system that is essential for the precise, temporospatial conduction of action potentials between neurons. Myelin also provides intercellular metabolic support to axons. Even minor disruptions in the integrity of myelin can impair neural performance and increase susceptibility to neurological diseases. In fact, myelin degeneration is a well-known neuropathological condition that is associated with normal aging and several neurodegenerative diseases, including multiple sclerosis and Alzheimer's disease. In the central nervous system, compact myelin sheaths are formed by fully mature oligodendrocytes. However, the entire oligodendrocyte lineage is susceptible to changes in the biological microenvironment and other risk factors that arise as the brain ages. In addition to their well-known role in action potential propagation, oligodendrocytes also provide intercellular metabolic support to axons by transferring energy metabolites and delivering exosomes. Therefore, myelin degeneration in the aging central nervous system is a significant contributor to the development of neurodegenerative diseases. Interventions that mitigate age-related myelin degeneration can improve neurological function in aging individuals. In this review, we investigate the changes in myelin that are associated with aging and their underlying mechanisms. We also discuss recent advances in understanding how myelin degeneration in the aging brain contributes to neurodegenerative diseases and explore the factors that can prevent, slow down, or even reverse age-related myelin degeneration. Future research will enhance our understanding of how reducing age-related myelin degeneration can be used as a therapeutic target for delaying or preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuangchan Wu
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, Guangdong Province, China
| | - Jun Chen
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
9
|
Cercel AM, Boboc IK, Surugiu R, Doeppner TR, Hermann DM, Catalin B, Gresita A, Popa-Wagner A. Grafts of hydrogel-embedded electrically stimulated subventricular stem cells into the stroke cavity improves functional recovery of mice. Neural Regen Res 2026; 21:695-703. [PMID: 39589177 DOI: 10.4103/nrr.nrr-d-23-02092] [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: 12/27/2023] [Accepted: 10/29/2024] [Indexed: 11/27/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00039/figure1/v/2025-05-05T160104Z/r/image-tiff The major aim of stroke therapy is to stimulate brain repair and improve behavioral recovery after cerebral ischemia. One option is to stimulate endogenous neurogenesis in the subventricular zone and direct the newly formed neurons to the damaged area. However, only a small percentage of these neurons survive, and many do not reach the damaged area, possibly because the corpus callosum impedes the migration of subventricular zone-derived stem cells into the lesioned cortex. A second major obstacle to stem cell therapy is the strong inflammatory reaction induced by cerebral ischemia, whereby the associated phagocytic activity of brain macrophages removes both therapeutic cells and/or cell-based drug carriers. To address these issues, neurogenesis was electrically stimulated in the subventricular zone, followed by isolation of proliferating cells, including newly formed neurons, which were subsequently mixed with a nutritional hydrogel. This mixture was then transferred to the stroke cavity of day 14 post-stroke mice. We found that the performance of the treated animals improved in behavioral tests, including novel object, open field, hole board, grooming, and "time-to-feel" adhesive tape tests. Furthermore, immunostaining revealed that the stem cell marker nestin, the neuroepithelial marker Mash1, and the immature neuronal marker doublecortin-positive cells survived in the transplanted area for 2 weeks, possibly due to reduced phagocytic activity and supportive angiogenesis. These results clearly indicate that the transplantation of committed subventricular zone stem cells combined with a protective nutritional gel directly into the infarct cavity after the peak of stroke-induced neuroinflammation represents a feasible approach to improve neurorestoration after cerebral ischemia.
Collapse
Affiliation(s)
- Andreea-Mihaela Cercel
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, Craiova, Romania
- Doctoral School, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Ianis Ks Boboc
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Roxana Surugiu
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, Craiova, Romania
- Chair of Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, Essen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Department of Neurology, University of Giessen Medical School, Giessen, Germany
| | - Dirk M Hermann
- Chair of Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, Essen, Germany
| | - Bogdan Catalin
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Andrei Gresita
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Aurel Popa-Wagner
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, Craiova, Romania
- Chair of Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, Essen, Germany
| |
Collapse
|
10
|
Jöe M, Williams PA. Targeting Wallerian degeneration in glaucoma. Neural Regen Res 2026; 21:693-694. [PMID: 39820324 DOI: 10.4103/nrr.nrr-d-24-01160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/01/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Melissa Jöe
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
11
|
Fu H, Li J, Zhang C, Gao G, Ge Q, Guan X, Cui D. Pathological axonal enlargement in connection with amyloidosis, lysosome destabilization, and bleeding is a major defect in Alzheimer's disease. Neural Regen Res 2026; 21:790-799. [PMID: 40326989 DOI: 10.4103/nrr.nrr-d-24-01440] [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: 11/18/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00047/figure1/v/2025-05-05T160104Z/r/image-tiff Alzheimer's disease is a multi-amyloidosis disease characterized by amyloid-β deposits in brain blood vessels, microaneurysms, and senile plaques. How amyloid-β deposition affects axon pathology has not been examined extensively. We used immunohistochemistry and immunofluorescence staining to analyze the forebrain tissue slices of Alzheimer's disease patients. Widespread axonal amyloidosis with distinctive axonal enlargement was observed in patients with Alzheimer's disease. On average, amyloid-β-positive axon diameters in Alzheimer's disease brains were 1.72 times those of control brain axons. Furthermore, axonal amyloidosis was associated with microtubule-associated protein 2 reduction, tau phosphorylation, lysosome destabilization, and several blood-related markers, such as apolipoprotein E, alpha-hemoglobin, glycosylated hemoglobin type A1C, and hemin. Lysosome destabilization in Alzheimer's disease was also clearly identified in the neuronal soma, where it was associated with the co-expression of amyloid-β, Cathepsin D, alpha-hemoglobin, actin alpha 2, and collagen type IV. This suggests that exogenous hemorrhagic protein intake influences neural lysosome stability. Additionally, the data showed that amyloid-β-containing lysosomes were 2.23 times larger than control lysosomes. Furthermore, under rare conditions, axonal breakages were observed, which likely resulted in Wallerian degeneration. In summary, axonal enlargement associated with amyloidosis, micro-bleeding, and lysosome destabilization is a major defect in patients with Alzheimer's disease. This finding suggests that, in addition to the well-documented neural soma and synaptic damage, axonal damage is a key component of neuronal defects in Alzheimer's disease.
Collapse
Affiliation(s)
- Hualin Fu
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Marine Equipment, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jilong Li
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chunlei Zhang
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guo Gao
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiqi Ge
- Institute of Marine Equipment, Shanghai Jiao Tong University, Shanghai, China
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China
| | - Xinping Guan
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China
- The Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
12
|
Eldar D, Albert S, Tatyana A, Galina S, Albert R, Yana M. Optogenetic approaches for neural tissue regeneration: A review of basic optogenetic principles and target cells for therapy. Neural Regen Res 2026; 21:521-533. [PMID: 39995064 DOI: 10.4103/nrr.nrr-d-24-00685] [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: 06/21/2024] [Accepted: 10/17/2024] [Indexed: 02/26/2025] Open
Abstract
Optogenetics has revolutionized the field of neuroscience by enabling precise control of neural activity through light-sensitive proteins known as opsins. This review article discusses the fundamental principles of optogenetics, including the activation of both excitatory and inhibitory opsins, as well as the development of optogenetic models that utilize recombinant viral vectors. A considerable portion of the article addresses the limitations of optogenetic tools and explores strategies to overcome these challenges. These strategies include the use of adeno-associated viruses, cell-specific promoters, modified opsins, and methodologies such as bioluminescent optogenetics. The application of viral recombinant vectors, particularly adeno-associated viruses, is emerging as a promising avenue for clinical use in delivering opsins to target cells. This trend indicates the potential for creating tools that offer greater flexibility and accuracy in opsin delivery. The adaptations of these viral vectors provide advantages in optogenetic studies by allowing for the restricted expression of opsins through cell-specific promoters and various viral serotypes. The article also examines different cellular targets for optogenetics, including neurons, astrocytes, microglia, and Schwann cells. Utilizing specific promoters for opsin expression in these cells is essential for achieving precise and efficient stimulation. Research has demonstrated that optogenetic stimulation of both neurons and glial cells-particularly the distinct phenotypes of microglia, astrocytes, and Schwann cells-can have therapeutic effects in neurological diseases. Glial cells are increasingly recognized as important targets for the treatment of these disorders. Furthermore, the article emphasizes the emerging field of bioluminescent optogenetics, which combines optogenetic principles with bioluminescent proteins to visualize and manipulate neural activity in real time. By integrating molecular genetics techniques with bioluminescence, researchers have developed methods to monitor neuronal activity efficiently and less invasively, enhancing our understanding of central nervous system function and the mechanisms of plasticity in neurological disorders beyond traditional neurobiological methods. Evidence has shown that optogenetic modulation can enhance motor axon regeneration, achieve complete sensory reinnervation, and accelerate the recovery of neuromuscular function. This approach also induces complex patterns of coordinated motor neuron activity and promotes neural reorganization. Optogenetic approaches hold immense potential for therapeutic interventions in the central nervous system. They enable precise control of neural circuits and may offer new treatments for neurological disorders, particularly spinal cord injuries, peripheral nerve injuries, and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Davletshin Eldar
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Sufianov Albert
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Research and Educational Institute of Neurosurgery, Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Ageeva Tatyana
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Sufianova Galina
- Department of Pharmacology, Tyumen State Medical University, Tyumen, Russia
| | - Rizvanov Albert
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Mukhamedshina Yana
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| |
Collapse
|
13
|
Shi R, Ye J, Liu Z, Wang C, Wu S, Shen H, Suo Q, Li W, He X, Zhang Z, Tang Y, Yang GY, Wang Y. Tropism-shifted AAV-PHP.eB-mediated bFGF gene therapy promotes varied neurorestoration after ischemic stroke in mice. Neural Regen Res 2026; 21:704-714. [PMID: 38993123 DOI: 10.4103/nrr.nrr-d-23-01802] [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: 11/02/2023] [Accepted: 03/26/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00040/figure1/v/2025-05-05T160104Z/r/image-tiff AAV-PHP.eB is an artificial adeno-associated virus (AAV) that crosses the blood-brain barrier and targets neurons more efficiently than other AAVs when administered systematically. While AAV-PHP.eB has been used in various disease models, its cellular tropism in cerebrovascular diseases remains unclear. In the present study, we aimed to elucidate the tropism of AAV-PHP.eB for different cell types in the brain in a mouse model of ischemic stroke and evaluate its effectiveness in mediating basic fibroblast growth factor ( bFGF ) gene therapy. Mice were injected intravenously with AAV-PHP.eB either 14 days prior to (pre-stroke) or 1 day following (post-stroke) transient middle cerebral artery occlusion. Notably, we observed a shift in tropism from neurons to endothelial cells with post-stroke administration of AAV-PHP.eB-mNeonGreen (mNG). This endothelial cell tropism correlated strongly with expression of the endothelial membrane receptor lymphocyte antigen 6 family member A (Ly6A). Furthermore, AAV-PHP.eB-mediated overexpression of bFGF markedly improved neurobehavioral outcomes and promoted long-term neurogenesis and angiogenesis post-ischemic stroke. Our findings underscore the significance of considering potential tropism shifts when utilizing AAV-PHP.eB-mediated gene therapy in neurological diseases and suggest a promising new strategy for bFGF gene therapy in stroke treatment.
Collapse
Affiliation(s)
- Rubing Shi
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Ye
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ze Liu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Cheng Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shengju Wu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Shen
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Suo
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wanlu Li
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaosong He
- Department of Emergency, the Second Affiliated Hospital, Department of Human Anatomy, School of Basic Science, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Zhijun Zhang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yaohui Tang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongting Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
Cui Z, He J, Li A, Wang J, Yang Y, Wang K, Liu Z, Ouyang Q, Su Z, Hu P, Xiao G. Novel insights into non-coding RNAs and their role in hydrocephalus. Neural Regen Res 2026; 21:636-647. [PMID: 39688559 DOI: 10.4103/nrr.nrr-d-24-00963] [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: 08/20/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
A large body of evidence has highlighted the role of non-coding RNAs in neurodevelopment and neuroinflammation. This evidence has led to increasing speculation that non-coding RNAs may be involved in the pathophysiological mechanisms underlying hydrocephalus, one of the most common neurological conditions worldwide. In this review, we first outline the basic concepts and incidence of hydrocephalus along with the limitations of existing treatments for this condition. Then, we outline the definition, classification, and biological role of non-coding RNAs. Subsequently, we analyze the roles of non-coding RNAs in the formation of hydrocephalus in detail. Specifically, we have focused on the potential significance of non-coding RNAs in the pathophysiology of hydrocephalus, including glymphatic pathways, neuroinflammatory processes, and neurological dysplasia, on the basis of the existing evidence. Lastly, we review the potential of non-coding RNAs as biomarkers of hydrocephalus and for the creation of innovative treatments.
Collapse
Affiliation(s)
- Zhiyue Cui
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan Province, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jian He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - An Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Junqiang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Kaiyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qian Ouyang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Neurosurgery, Zhuzhou Hospital, Central South University Xiangya School of Medicine, Zhuzhou, Hunan Province, China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke 's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, UK
| | - Pingsheng Hu
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan Province, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
15
|
Desai M, Gulati K, Agrawal M, Ghumra S, Sahoo PK. Stress granules: Guardians of cellular health and triggers of disease. Neural Regen Res 2026; 21:588-597. [PMID: 39995077 DOI: 10.4103/nrr.nrr-d-24-01196] [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: 10/07/2024] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
Stress granules are membraneless organelles that serve as a protective cellular response to external stressors by sequestering non-translating messenger RNAs (mRNAs) and regulating protein synthesis. Stress granules formation mechanism is conserved across species, from yeast to mammals, and they play a critical role in minimizing cellular damage during stress. Composed of heterogeneous ribonucleoprotein complexes, stress granules are enriched not only in mRNAs but also in noncoding RNAs and various proteins, including translation initiation factors and RNA-binding proteins. Genetic mutations affecting stress granule assembly and disassembly can lead to abnormal stress granule accumulation, contributing to the progression of several diseases. Recent research indicates that stress granule dynamics are pivotal in determining their physiological and pathological functions, with acute stress granule formation offering protection and chronic stress granule accumulation being detrimental. This review focuses on the multifaceted roles of stress granules under diverse physiological conditions, such as regulation of mRNA transport, mRNA translation, apoptosis, germ cell development, phase separation processes that govern stress granule formation, and their emerging implications in pathophysiological scenarios, such as viral infections, cancer, neurodevelopmental disorders, neurodegeneration, and neuronal trauma.
Collapse
Affiliation(s)
- Meghal Desai
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Keya Gulati
- College of Science and Liberal Arts, New Jersey Institute of Technology, Newark, NJ, USA
| | - Manasi Agrawal
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Shruti Ghumra
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Pabitra K Sahoo
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| |
Collapse
|
16
|
Solomon B, Voronov-Goldman M. Amyloid-β is NOT only the most promising target for Alzheimer's disease. Neural Regen Res 2026; 21:681-682. [PMID: 39665791 DOI: 10.4103/nrr.nrr-d-24-00829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/27/2024] [Indexed: 12/13/2024] Open
Affiliation(s)
- Beka Solomon
- Department of Molecular Microbiology and Biotechnology, The Shmunis School of Biomedicine and Cancer Research, the George S. Wise Faculty of Life Sciences Tel-Aviv University, Ramat Aviv, Israel
| | | |
Collapse
|
17
|
Lin AL, Aware C. Rapamycin as a preventive intervention for Alzheimer's disease in APOE4 carriers: Targeting brain metabolic and vascular restoration. Neural Regen Res 2026; 21:685-686. [PMID: 40326985 DOI: 10.4103/nrr.nrr-d-24-01006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/23/2024] [Indexed: 05/07/2025] Open
Affiliation(s)
- Ai-Ling Lin
- Department of Radiology, University of Missouri, Columbia, MO, USA (Lin AL, Aware C)
- NextGen Precision Health, University of Missouri, Columbia, MO, USA (Lin AL, Aware C)
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA (Lin AL)
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA (Lin AL)
| | - Chetan Aware
- Department of Radiology, University of Missouri, Columbia, MO, USA (Lin AL, Aware C)
- NextGen Precision Health, University of Missouri, Columbia, MO, USA (Lin AL, Aware C)
| |
Collapse
|
18
|
Liu M, Meng Y, Ouyang S, Zhai M, Yang L, Yang Y, Wang Y. Neuromodulation technologies improve functional recovery after brain injury: From bench to bedside. Neural Regen Res 2026; 21:506-520. [PMID: 39851132 DOI: 10.4103/nrr.nrr-d-24-00652] [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: 06/12/2024] [Accepted: 11/05/2024] [Indexed: 01/26/2025] Open
Abstract
Spontaneous recovery frequently proves maladaptive or insufficient because the plasticity of the injured adult mammalian central nervous system is limited. This limited plasticity serves as a primary barrier to functional recovery after brain injury. Neuromodulation technologies represent one of the fastest-growing fields in medicine. These techniques utilize electricity, magnetism, sound, and light to restore or optimize brain functions by promoting reorganization or long-term changes that support functional recovery in patients with brain injury. Therefore, this review aims to provide a comprehensive overview of the effects and underlying mechanisms of neuromodulation technologies in supporting motor function recovery after brain injury. Many of these technologies are widely used in clinical practice and show significant improvements in motor function across various types of brain injury. However, studies report negative findings, potentially due to variations in stimulation protocols, differences in observation periods, and the severity of functional impairments among participants across different clinical trials. Additionally, we observed that different neuromodulation techniques share remarkably similar mechanisms, including promoting neuroplasticity, enhancing neurotrophic factor release, improving cerebral blood flow, suppressing neuroinflammation, and providing neuroprotection. Finally, considering the advantages and disadvantages of various neuromodulation techniques, we propose that future development should focus on closed-loop neural circuit stimulation, personalized treatment, interdisciplinary collaboration, and precision stimulation.
Collapse
Affiliation(s)
- Mei Liu
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA), Wuxi, Jiangsu Province, China
| | - Yijing Meng
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA), Wuxi, Jiangsu Province, China
| | - Siguang Ouyang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA), Wuxi, Jiangsu Province, China
| | - Meng'ai Zhai
- Department of Neurosurgery, The 904 Hospital of PLA, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Likun Yang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA), Wuxi, Jiangsu Province, China
| | - Yang Yang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA), Wuxi, Jiangsu Province, China
| | - Yuhai Wang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA), Wuxi, Jiangsu Province, China
| |
Collapse
|
19
|
Sha S, Ren L, Xing X, Guo W, Wang Y, Li Y, Cao Y, Qu L. Recent advances in immunotherapy targeting amyloid-beta and tauopathies in Alzheimer's disease. Neural Regen Res 2026; 21:577-587. [PMID: 39885674 DOI: 10.4103/nrr.nrr-d-24-00846] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/28/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease, a devastating neurodegenerative disorder, is characterized by progressive cognitive decline, primarily due to amyloid-beta protein deposition and tau protein phosphorylation. Effectively reducing the cytotoxicity of amyloid-beta42 aggregates and tau oligomers may help slow the progression of Alzheimer's disease. Conventional drugs, such as donepezil, can only alleviate symptoms and are not able to prevent the underlying pathological processes or cognitive decline. Currently, active and passive immunotherapies targeting amyloid-beta and tau have shown some efficacy in mice with asymptomatic Alzheimer's disease and other transgenic animal models, attracting considerable attention. However, the clinical application of these immunotherapies demonstrated only limited efficacy before the discovery of lecanemab and donanemab. This review first discusses the advancements in the pathogenesis of Alzheimer's disease and active and passive immunotherapies targeting amyloid-beta and tau proteins. Furthermore, it reviews the advantages and disadvantages of various immunotherapies and considers their future prospects. Although some antibodies have shown promise in patients with mild Alzheimer's disease, substantial clinical data are still lacking to validate their effectiveness in individuals with moderate Alzheimer's disease.
Collapse
Affiliation(s)
- Sha Sha
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Lina Ren
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaona Xing
- Department of Neurology, Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Wanshu Guo
- Department of Neurology, People's Hospital of Liaoning Province, Shenyang, Liaoning Province, China
| | - Yan Wang
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ying Li
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Le Qu
- Department of Dermatology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
20
|
Payne JL, Sabunciyan S. Liquid biopsies in psychiatric disorders: Identifying peripheral biomarkers of brain health. Neural Regen Res 2026; 21:691-692. [PMID: 39819865 DOI: 10.4103/nrr.nrr-d-24-00962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Jennifer L Payne
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA (Payne JL)
| | - Sarven Sabunciyan
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA (Sabunciyan S)
| |
Collapse
|
21
|
Hohman G, Shahid M, Eldeeb MA. Stress signaling caused by mitochondrial import malfunction can be terminated by SIFI: Importance of stress response silencing. Neural Regen Res 2026; 21:673-674. [PMID: 39820346 DOI: 10.4103/nrr.nrr-d-24-01169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/02/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Grace Hohman
- Department of Chemistry, Illinois State University, Normal, IL, USA
| | | | | |
Collapse
|
22
|
Saiyisan A, Zeng S, Zhang H, Wang Z, Wang J, Cai P, Huang J. Chemical exchange saturation transfer MRI for neurodegenerative diseases: An update on clinical and preclinical studies. Neural Regen Res 2026; 21:553-568. [PMID: 39885672 DOI: 10.4103/nrr.nrr-d-24-01246] [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: 10/15/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025] Open
Abstract
Chemical exchange saturation transfer magnetic resonance imaging is an advanced imaging technique that enables the detection of compounds at low concentrations with high sensitivity and spatial resolution and has been extensively studied for diagnosing malignancy and stroke. In recent years, the emerging exploration of chemical exchange saturation transfer magnetic resonance imaging for detecting pathological changes in neurodegenerative diseases has opened up new possibilities for early detection and repetitive scans without ionizing radiation. This review serves as an overview of chemical exchange saturation transfer magnetic resonance imaging with detailed information on contrast mechanisms and processing methods and summarizes recent developments in both clinical and preclinical studies of chemical exchange saturation transfer magnetic resonance imaging for Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. A comprehensive literature search was conducted using databases such as PubMed and Google Scholar, focusing on peer-reviewed articles from the past 15 years relevant to clinical and preclinical applications. The findings suggest that chemical exchange saturation transfer magnetic resonance imaging has the potential to detect molecular changes and altered metabolism, which may aid in early diagnosis and assessment of the severity of neurodegenerative diseases. Although promising results have been observed in selected clinical and preclinical trials, further validations are needed to evaluate their clinical value. When combined with other imaging modalities and advanced analytical methods, chemical exchange saturation transfer magnetic resonance imaging shows potential as an in vivo biomarker, enhancing the understanding of neuropathological mechanisms in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ahelijiang Saiyisan
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Shihao Zeng
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Huabin Zhang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Ziyan Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jiawen Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Pei Cai
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jianpan Huang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Tam Wing Fan Neuroimaging Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
23
|
Palumbo L, Nuzzo D, Girgenti A, Picone P. Mitochondria-derived vesicles: New players in the game of neurodegeneration. Neural Regen Res 2026; 21:679-680. [PMID: 39819868 DOI: 10.4103/nrr.nrr-d-24-01220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/05/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Laura Palumbo
- Istituto per la Ricerca e l'Innovazione Biomedica, CNR, Via Ugo La Malfa, Palermo, Italy
| | | | | | | |
Collapse
|
24
|
Jiang T, Li Y, Liu H, Sun Y, Zhang H, Zhang Q, Tang S, Niu X, Du H, Yu Y, Yue H, Guo Y, Chen Y, Xu F. Blood-brain barrier disruption and neuroinflammation in the hippocampus of a cardiac arrest porcine model: Single-cell RNA sequencing analysis. Neural Regen Res 2026; 21:742-755. [PMID: 40146000 DOI: 10.4103/nrr.nrr-d-24-01269] [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: 10/20/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00043/figure1/v/2025-05-05T160104Z/r/image-tiff Global brain ischemia and neurological deficit are consequences of cardiac arrest that lead to high mortality. Despite advancements in resuscitation science, our limited understanding of the cellular and molecular mechanisms underlying post-cardiac arrest brain injury have hindered the development of effective neuroprotective strategies. Previous studies primarily focused on neuronal death, potentially overlooking the contributions of non-neuronal cells and intercellular communication to the pathophysiology of cardiac arrest-induced brain injury. To address these gaps, we hypothesized that single-cell transcriptomic analysis could uncover previously unidentified cellular subpopulations, altered cell communication networks, and novel molecular mechanisms involved in post-cardiac arrest brain injury. In this study, we performed a single-cell transcriptomic analysis of the hippocampus from pigs with ventricular fibrillation-induced cardiac arrest at 6 and 24 hours following the return of spontaneous circulation, and from sham control pigs. Sequencing results revealed changes in the proportions of different cell types, suggesting post-arrest disruption in the blood-brain barrier and infiltration of neutrophils. These results were validated through western blotting, quantitative reverse transcription-polymerase chain reaction, and immunofluorescence staining. We also identified and validated a unique subcluster of activated microglia with high expression of S100A8, which increased over time following cardiac arrest. This subcluster simultaneously exhibited significant M1/M2 polarization and expressed key functional genes related to chemokines and interleukins. Additionally, we revealed the post-cardiac arrest dysfunction of oligodendrocytes and the differentiation of oligodendrocyte precursor cells into oligodendrocytes. Cell communication analysis identified enhanced post-cardiac arrest communication between neutrophils and microglia that was mediated by neutrophil-derived resistin, driving pro-inflammatory microglial polarization. Our findings provide a comprehensive single-cell map of the post-cardiac arrest hippocampus, offering potential novel targets for neuroprotection and repair following cardiac arrest.
Collapse
Affiliation(s)
- Tangxing Jiang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yaning Li
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hehui Liu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yijun Sun
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Huidan Zhang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Qirui Zhang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Shuyao Tang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Xu Niu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Han Du
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yinxia Yu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hongwei Yue
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yunyun Guo
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
25
|
Yao C, Xie D, Zhang Y, Shen Y, Sun P, Ma Z, Li J, Tao J, Fang M. Tryptophan metabolism and ischemic stroke: An intricate balance. Neural Regen Res 2026; 21:466-477. [PMID: 40326980 DOI: 10.4103/nrr.nrr-d-24-00777] [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: 07/16/2024] [Accepted: 11/27/2024] [Indexed: 05/07/2025] Open
Abstract
Ischemic stroke, which is characterized by hypoxia and ischemia, triggers a cascade of injury responses, including neurotoxicity, inflammation, oxidative stress, disruption of the blood-brain barrier, and neuronal death. In this context, tryptophan metabolites and enzymes, which are synthesized through the kynurenine and 5-hydroxytryptamine pathways, play dual roles. The delicate balance between neurotoxic and neuroprotective substances is a crucial factor influencing the progression of ischemic stroke. Neuroprotective metabolites, such as kynurenic acid, exert their effects through various mechanisms, including competitive blockade of N-methyl-D-aspartate receptors, modulation of α7 nicotinic acetylcholine receptors, and scavenging of reactive oxygen species. In contrast, neurotoxic substances such as quinolinic acid can hinder the development of vascular glucose transporter proteins, induce neurotoxicity mediated by reactive oxygen species, and disrupt mitochondrial function. Additionally, the enzymes involved in tryptophan metabolism play major roles in these processes. Indoleamine 2,3-dioxygenase in the kynurenine pathway and tryptophan hydroxylase in the 5-hydroxytryptamine pathway influence neuroinflammation and brain homeostasis. Consequently, the metabolites generated through tryptophan metabolism have substantial effects on the development and progression of ischemic stroke. Stroke treatment aims to restore the balance of various metabolite levels; however, precise regulation of tryptophan metabolism within the central nervous system remains a major challenge for the treatment of ischemic stroke. Therefore, this review aimed to elucidate the complex interactions between tryptophan metabolites and enzymes in ischemic stroke and develop targeted therapies that can restore the delicate balance between neurotoxicity and neuroprotection.
Collapse
Affiliation(s)
- Chongjie Yao
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Xie
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuchen Zhang
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanhao Shen
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pingping Sun
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhao Ma
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jin Li
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiming Tao
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Fang
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Research Institute of Tuina, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Chen H, Li N, Cai Y, Ma C, Ye Y, Shi X, Guo J, Han Z, Liu Y, Wei X. Exosomes in neurodegenerative diseases: Therapeutic potential and modification methods. Neural Regen Res 2026; 21:478-490. [PMID: 40326981 DOI: 10.4103/nrr.nrr-d-24-00720] [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: 08/06/2024] [Accepted: 10/14/2024] [Indexed: 05/07/2025] Open
Abstract
In recent years, exosomes have garnered extensive attention as therapeutic agents and early diagnostic markers in neurodegenerative disease research. Exosomes are small and can effectively cross the blood-brain barrier, allowing them to target deep brain lesions. Recent studies have demonstrated that exosomes derived from different cell types may exert therapeutic effects by regulating the expression of various inflammatory cytokines, mRNAs, and disease-related proteins, thereby halting the progression of neurodegenerative diseases and exhibiting beneficial effects. However, exosomes are composed of lipid bilayer membranes and lack the ability to recognize specific target cells. This limitation can lead to side effects and toxicity when they interact with non-specific cells. Growing evidence suggests that surface-modified exosomes have enhanced targeting capabilities and can be used as targeted drug-delivery vehicles that show promising results in the treatment of neurodegenerative diseases. In this review, we provide an up-to-date overview of existing research aimed at devising approaches to modify exosomes and elucidating their therapeutic potential in neurodegenerative diseases. Our findings indicate that exosomes can efficiently cross the blood-brain barrier to facilitate drug delivery and can also serve as early diagnostic markers for neurodegenerative diseases. We introduce the strategies being used to enhance exosome targeting, including genetic engineering, chemical modifications (both covalent, such as click chemistry and metabolic engineering, and non-covalent, such as polyvalent electrostatic and hydrophobic interactions, ligand-receptor binding, aptamer-based modifications, and the incorporation of CP05-anchored peptides), and nanomaterial modifications. Research into these strategies has confirmed that exosomes have significant therapeutic potential for neurodegenerative diseases. However, several challenges remain in the clinical application of exosomes. Improvements are needed in preparation, characterization, and optimization methods, as well as in reducing the adverse reactions associated with their use. Additionally, the range of applications and the safety of exosomes require further research and evaluation.
Collapse
Affiliation(s)
- Hongli Chen
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
| | - Na Li
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
| | - Yuanhao Cai
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
- School of Intelligent Information Engineering, Medicine & Technology College of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Chunyan Ma
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
| | - Yutong Ye
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
| | - Xinyu Shi
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
| | - Jun Guo
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
| | - Zhibo Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceuticals, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China
| | - Yi Liu
- State Key Laboratory of Separation Membrane and Membrane Process & Tianjin Key Laboratory of Optoelectronic Detection Technology and Systems, School of Life Sciences, Tiangong University, Tianjin, China
| | - Xunbin Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Cancer Hospital & Institute, International Cancer Institute, Institute of Medical Technology, Peking University Health Science Center, Department of Biomedical Engineering, Peking University, Beijing, China
| |
Collapse
|
27
|
Sun Y, Pang X, Huang X, Liu D, Huang J, Zheng P, Wei Y, Pang C. Potential mechanisms of non-coding RNA regulation in Alzheimer's disease. Neural Regen Res 2026; 21:265-280. [PMID: 39851253 PMCID: PMC12094571 DOI: 10.4103/nrr.nrr-d-24-00696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/02/2024] [Accepted: 10/19/2024] [Indexed: 01/26/2025] Open
Abstract
Alzheimer's disease, a progressively degenerative neurological disorder, is the most common cause of dementia in the elderly. While its precise etiology remains unclear, researchers have identified diverse pathological characteristics and molecular pathways associated with its progression. Advances in scientific research have increasingly highlighted the crucial role of non-coding RNAs in the progression of Alzheimer's disease. These non-coding RNAs regulate several biological processes critical to the advancement of the disease, offering promising potential as therapeutic targets and diagnostic biomarkers. Therefore, this review aims to investigate the underlying mechanisms of Alzheimer's disease onset, with a particular focus on microRNAs, long non-coding RNAs, and circular RNAs associated with the disease. The review elucidates the potential pathogenic processes of Alzheimer's disease and provides a detailed description of the synthesis mechanisms of the three aforementioned non-coding RNAs. It comprehensively summarizes the various non-coding RNAs that have been identified to play key regulatory roles in Alzheimer's disease, as well as how these non-coding RNAs influence the disease's progression by regulating gene expression and protein functions. For example, miR-9 targets the UBE4B gene, promoting autophagy-mediated degradation of Tau protein, thereby reducing Tau accumulation and delaying Alzheimer's disease progression. Conversely, the long non-coding RNA BACE1-AS stabilizes BACE1 mRNA, promoting the generation of amyloid-β and accelerating Alzheimer's disease development. Additionally, circular RNAs play significant roles in regulating neuroinflammatory responses. By integrating insights from these regulatory mechanisms, there is potential to discover new therapeutic targets and potential biomarkers for early detection and management of Alzheimer's disease. This review aims to enhance the understanding of the relationship between Alzheimer's disease and non-coding RNAs, potentially paving the way for early detection and novel treatment strategies.
Collapse
Affiliation(s)
- Yue Sun
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Xinping Pang
- School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu Province, China
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Dinglu Liu
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Jingyue Huang
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Pengtao Zheng
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Yanyu Wei
- National Key Laboratory of Science and Technology on Vacuum Electronics, School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| |
Collapse
|
28
|
Chen Y, Hu J, Zhang Y, Peng L, Li X, Li C, Wu X, Wang C. Epilepsy therapy beyond neurons: Unveiling astrocytes as cellular targets. Neural Regen Res 2026; 21:23-38. [PMID: 39819836 PMCID: PMC12094549 DOI: 10.4103/nrr.nrr-d-24-01035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 10/16/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025] Open
Abstract
Epilepsy is a leading cause of disability and mortality worldwide. However, despite the availability of more than 20 antiseizure medications, more than one-third of patients continue to experience seizures. Given the urgent need to explore new treatment strategies for epilepsy, recent research has highlighted the potential of targeting gliosis, metabolic disturbances, and neural circuit abnormalities as therapeutic strategies. Astrocytes, the largest group of nonneuronal cells in the central nervous system, play several crucial roles in maintaining ionic and energy metabolic homeostasis in neurons, regulating neurotransmitter levels, and modulating synaptic plasticity. This article briefly reviews the critical role of astrocytes in maintaining balance within the central nervous system. Building on previous research, we discuss how astrocyte dysfunction contributes to the onset and progression of epilepsy through four key aspects: the imbalance between excitatory and inhibitory neuronal signaling, dysregulation of metabolic homeostasis in the neuronal microenvironment, neuroinflammation, and the formation of abnormal neural circuits. We summarize relevant basic research conducted over the past 5 years that has focused on modulating astrocytes as a therapeutic approach for epilepsy. We categorize the therapeutic targets proposed by these studies into four areas: restoration of the excitation-inhibition balance, reestablishment of metabolic homeostasis, modulation of immune and inflammatory responses, and reconstruction of abnormal neural circuits. These targets correspond to the pathophysiological mechanisms by which astrocytes contribute to epilepsy. Additionally, we need to consider the potential challenges and limitations of translating these identified therapeutic targets into clinical treatments. These limitations arise from interspecies differences between humans and animal models, as well as the complex comorbidities associated with epilepsy in humans. We also highlight valuable future research directions worth exploring in the treatment of epilepsy and the regulation of astrocytes, such as gene therapy and imaging strategies. The findings presented in this review may help open new therapeutic avenues for patients with drug-resistant epilepsy and for those suffering from other central nervous system disorders associated with astrocytic dysfunction.
Collapse
Affiliation(s)
- Yuncan Chen
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiayi Hu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lulu Peng
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cong Li
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xunyi Wu
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Cong Wang
- Shanghai Fifth People’s Hospital, School of Pharmacy, MOE Key Laboratory of Smart Drug Delivery, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Shanghai, China
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
29
|
Terzo S, Amato A, Calvi P, Giardina M, Nuzzo D, Picone P, Palumbo-Piccionello A, Amata S, Giardina IC, Massaro A, Restivo I, Attanzio A, Tesoriere L, Allegra M, Mulè F. Positive impact of indicaxanthin from Opuntia ficus-indica fruit on high-fat diet-induced neuronal damage and gut microbiota dysbiosis. Neural Regen Res 2026; 21:324-332. [PMID: 39314163 PMCID: PMC12094550 DOI: 10.4103/nrr.nrr-d-23-02039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 07/26/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202601000-00036/figure1/v/2025-06-09T151831Z/r/image-tiff Indicaxanthin is a betalain that is abundant in Opuntia ficus-indica orange fruit and has antioxidative and anti-inflammatory effects. Nevertheless, very little is known about the neuroprotective potential of indicaxanthin. This study investigated the impact of indicaxanthin on neuronal damage and gut microbiota dysbiosis induced by a high-fat diet in mice. The mice were divided into three groups according to different diets: the negative control group was fed a standard diet; the high-fat diet group was fed a high-fat diet; and the high-fat diet + indicaxanthin group was fed a high-fat diet and received indicaxanthin orally (0.86 mg/kg per day) for 4 weeks. Brain apoptosis, redox status, inflammation, and the gut microbiota composition were compared among the different animal groups. The results demonstrated that indicaxanthin treatment reduced neuronal apoptosis by downregulating the expression of proapoptotic genes and increasing the expression of antiapoptotic genes. Indicaxanthin also markedly decreased the expression of neuroinflammatory proteins and genes and inhibited high-fat diet-induced neuronal oxidative stress by reducing reactive oxygen and nitrogen species, malondialdehyde, and nitric oxide levels. In addition, indicaxanthin treatment improved the microflora composition by increasing the abundance of healthy bacterial genera, known as producers of short-chain fatty acids ( Lachnospiraceae , Alloprovetella , and Lactobacillus ), and by reducing bacteria related to unhealthy profiles ( Blautia , Faecalibaculum , Romboutsia and Bilophila ). In conclusion, indicaxanthin has a positive effect on high-fat diet-induced neuronal damage and on the gut microbiota composition in obese mice.
Collapse
Affiliation(s)
- Simona Terzo
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Antonella Amato
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation – IRIB, Palermo, Italy
| | - Pasquale Calvi
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, Palermo, Italy
| | - Marta Giardina
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Domenico Nuzzo
- Institute for Biomedical Research and Innovation – IRIB, Palermo, Italy
| | - Pasquale Picone
- Institute for Biomedical Research and Innovation – IRIB, Palermo, Italy
| | - Antonio Palumbo-Piccionello
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Sara Amata
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Ilenia Concetta Giardina
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Alessandro Massaro
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Ignazio Restivo
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Alessandro Attanzio
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Mario Allegra
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| | - Flavia Mulè
- Department of Biological- Chemical- Pharmaceutical Science and Technology, University of Palermo, Palermo, Italy
| |
Collapse
|
30
|
Zeng F, Li Y, Li X, Gu X, Cao Y, Cheng S, Tian H, Mei R, Mei X. Microglia overexpressing brain-derived neurotrophic factor promote vascular repair and functional recovery in mice after spinal cord injury. Neural Regen Res 2026; 21:365-376. [PMID: 39435607 PMCID: PMC12094574 DOI: 10.4103/nrr.nrr-d-24-00381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/26/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202601000-00040/figure1/v/2025-06-09T151831Z/r/image-tiff Spinal cord injury represents a severe form of central nervous system trauma for which effective treatments remain limited. Microglia is the resident immune cells of the central nervous system, play a critical role in spinal cord injury. Previous studies have shown that microglia can promote neuronal survival by phagocytosing dead cells and debris and by releasing neuroprotective and anti-inflammatory factors. However, excessive activation of microglia can lead to persistent inflammation and contribute to the formation of glial scars, which hinder axonal regeneration. Despite this, the precise role and mechanisms of microglia during the acute phase of spinal cord injury remain controversial and poorly understood. To elucidate the role of microglia in spinal cord injury, we employed the colony-stimulating factor 1 receptor inhibitor PLX5622 to deplete microglia. We observed that sustained depletion of microglia resulted in an expansion of the lesion area, downregulation of brain-derived neurotrophic factor, and impaired functional recovery after spinal cord injury. Next, we generated a transgenic mouse line with conditional overexpression of brain-derived neurotrophic factor specifically in microglia. We found that brain-derived neurotrophic factor overexpression in microglia increased angiogenesis and blood flow following spinal cord injury and facilitated the recovery of hindlimb motor function. Additionally, brain-derived neurotrophic factor overexpression in microglia reduced inflammation and neuronal apoptosis during the acute phase of spinal cord injury. Furthermore, through using specific transgenic mouse lines, TMEM119, and the colony-stimulating factor 1 receptor inhibitor PLX73086, we demonstrated that the neuroprotective effects were predominantly due to brain-derived neurotrophic factor overexpression in microglia rather than macrophages. In conclusion, our findings suggest the critical role of microglia in the formation of protective glial scars. Depleting microglia is detrimental to recovery of spinal cord injury, whereas targeting brain-derived neurotrophic factor overexpression in microglia represents a promising and novel therapeutic strategy to enhance motor function recovery in patients with spinal cord injury.
Collapse
Affiliation(s)
- Fanzhuo Zeng
- Department of Orthopedics, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
- Department of Neurobiology, School of Basic Medical Sciences, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuxin Li
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xiaoyu Li
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xinyang Gu
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yue Cao
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shuai Cheng
- Department of Orthopedics, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - He Tian
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Rongcheng Mei
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Xifan Mei
- Department of Orthopedics, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| |
Collapse
|
31
|
Qu X, Lai X, He M, Zhang J, Xiang B, Liu C, Huang R, Shi Y, Qiao J. Investigation of epilepsy-related genes in a Drosophila model. Neural Regen Res 2026; 21:195-211. [PMID: 39688550 PMCID: PMC12094548 DOI: 10.4103/nrr.nrr-d-24-00877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/15/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Complex genetic architecture is the major cause of heterogeneity in epilepsy, which poses challenges for accurate diagnosis and precise treatment. A large number of epilepsy candidate genes have been identified from clinical studies, particularly with the widespread use of next-generation sequencing. Validating these candidate genes is emerging as a valuable yet challenging task. Drosophila serves as an ideal animal model for validating candidate genes associated with neurogenetic disorders such as epilepsy, due to its rapid reproduction rate, powerful genetic tools, and efficient use of ethological and electrophysiological assays. Here, we systematically summarize the advantageous techniques of the Drosophila model used to investigate epilepsy genes, including genetic tools for manipulating target gene expression, ethological assays for seizure-like behaviors, electrophysiological techniques, and functional imaging for recording neural activity. We then introduce several typical strategies for identifying epilepsy genes and provide new insights into gene‒gene interactions in epilepsy with polygenic causes. We summarize well-established precision medicine strategies for epilepsy and discuss prospective treatment options, including drug therapy and gene therapy for genetic epilepsy based on the Drosophila model. Finally, we also address genetic counseling and assisted reproductive technology as potential approaches for the prevention of genetic epilepsy.
Collapse
Affiliation(s)
- Xiaochong Qu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xiaodan Lai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Mingfeng He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Jinyuan Zhang
- School of Health Management, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Binbin Xiang
- The First Clinical Medicine School of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Chuqiao Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Ruina Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yiwu Shi
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Jingda Qiao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
32
|
Cai X, Cai X, Xie Q, Xiao X, Li T, Zhou T, Sun H. NLRP3 inflammasome and gut microbiota-brain axis: A new perspective on white matter injury after intracerebral hemorrhage. Neural Regen Res 2026; 21:62-80. [PMID: 39885662 PMCID: PMC12094575 DOI: 10.4103/nrr.nrr-d-24-00917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/09/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025] Open
Abstract
Intracerebral hemorrhage is the most dangerous subtype of stroke, characterized by high mortality and morbidity rates, and frequently leads to significant secondary white matter injury. In recent decades, studies have revealed that gut microbiota can communicate bidirectionally with the brain through the gut microbiota-brain axis. This axis indicates that gut microbiota is closely related to the development and prognosis of intracerebral hemorrhage and its associated secondary white matter injury. The NACHT, LRR, and pyrin domain-containing protein 3 (NLRP3) inflammasome plays a crucial role in this context. This review summarizes the dysbiosis of gut microbiota following intracerebral hemorrhage and explores the mechanisms by which this imbalance may promote the activation of the NLRP3 inflammasome. These mechanisms include metabolic pathways (involving short-chain fatty acids, lipopolysaccharides, lactic acid, bile acids, trimethylamine-N-oxide, and tryptophan), neural pathways (such as the vagus nerve and sympathetic nerve), and immune pathways (involving microglia and T cells). We then discuss the relationship between the activated NLRP3 inflammasome and secondary white matter injury after intracerebral hemorrhage. The activation of the NLRP3 inflammasome can exacerbate secondary white matter injury by disrupting the blood-brain barrier, inducing neuroinflammation, and interfering with nerve regeneration. Finally, we outline potential treatment strategies for intracerebral hemorrhage and its secondary white matter injury. Our review highlights the critical role of the gut microbiota-brain axis and the NLRP3 inflammasome in white matter injury following intracerebral hemorrhage, paving the way for exploring potential therapeutic approaches.
Collapse
Affiliation(s)
- Xiaoxi Cai
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xinhong Cai
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Quanhua Xie
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xueqi Xiao
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Tong Li
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Tian Zhou
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong–Hong Kong–Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong–Hong Kong–Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
33
|
Arnold P, Zunke F. Base and translation of β-glucocerebrosidase and its transporter LIMP-2 in neuropathies. Neural Regen Res 2026; 21:314-315. [PMID: 39819831 PMCID: PMC12094559 DOI: 10.4103/nrr.nrr-d-24-01056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 01/19/2025] Open
Affiliation(s)
- Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
34
|
Rhymes ER, Sleigh JN. Shared mechanisms and pathological phenotypes underlying aminoacyl-tRNA synthetase-related neuropathies. Neural Regen Res 2026; 21:312-313. [PMID: 39851138 PMCID: PMC12094558 DOI: 10.4103/nrr.nrr-d-24-01080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 01/26/2025] Open
Affiliation(s)
- Elena R. Rhymes
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - James N. Sleigh
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
| |
Collapse
|
35
|
Jahan I, Harun-Ur-Rashid M, Islam MA, Sharmin F, Al Jaouni SK, Kaki AM, Selim S. Neuronal plasticity and its role in Alzheimer's disease and Parkinson's disease. Neural Regen Res 2026; 21:107-125. [PMID: 39688547 PMCID: PMC12094540 DOI: 10.4103/nrr.nrr-d-24-01019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/09/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Neuronal plasticity, the brain's ability to adapt structurally and functionally, is essential for learning, memory, and recovery from injuries. In neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, this plasticity is disrupted, leading to cognitive and motor deficits. This review explores the mechanisms of neuronal plasticity and its effect on Alzheimer's disease and Parkinson's disease. Alzheimer's disease features amyloid-beta plaques and tau tangles that impair synaptic function, while Parkinson's disease involves the loss of dopaminergic neurons affecting motor control. Enhancing neuronal plasticity offers therapeutic potential for these diseases. A systematic literature review was conducted using databases such as PubMed, Scopus, and Google Scholar, focusing on studies of neuronal plasticity in Alzheimer's disease and Parkinson's disease. Data synthesis identified key themes such as synaptic mechanisms, neurogenesis, and therapeutic strategies, linking molecular insights to clinical applications. Results highlight that targeting synaptic plasticity mechanisms, such as long-term potentiation and long-term depression, shows promise. Neurotrophic factors, advanced imaging techniques, and molecular tools (e.g., clustered regularly interspaced short palindromic repeats and optogenetics) are crucial in understanding and enhancing plasticity. Current therapies, including dopamine replacement, deep brain stimulation, and lifestyle interventions, demonstrate the potential to alleviate symptoms and improve outcomes. In conclusion, enhancing neuronal plasticity through targeted therapies holds significant promise for treating neurodegenerative diseases. Future research should integrate multidisciplinary approaches to fully harness the therapeutic potential of neuronal plasticity in Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Israt Jahan
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Mohammad Harun-Ur-Rashid
- Department of Chemistry, International University of Business Agriculture and Technology (IUBAT), Sector 10, Uttara Model Town, Dhaka, Bangladesh
| | - Md. Aminul Islam
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Farhana Sharmin
- Department of Anatomy, Shaheed Suhrawardy Medical College, Dhaka, Bangladesh
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah M. Kaki
- Department of Anesthesia and Pain Medicine, Director of Pain Clinic, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
36
|
Guo HH, Ou HN, Yu JS, Rosa JM, Formolo DA, Cheng T, Yau SY, Tsang HWH. Adiponectin as a potential mediator of the pro-cognitive effects of physical exercise on Alzheimer's disease. Neural Regen Res 2026; 21:96-106. [PMID: 39885660 PMCID: PMC12094572 DOI: 10.4103/nrr.nrr-d-23-00943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 01/11/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease is the primary cause of dementia and imposes a significant socioeconomic burden globally. Physical exercise, as an effective strategy for improving general health, has been largely reported for its effectiveness in slowing neurodegeneration and increasing brain functional plasticity, particularly in aging brains. However, the underlying mechanisms of exercise in cognitive aging remain largely unclear. Adiponectin, a cell-secreted protein hormone, has recently been found to regulate synaptic plasticity and mediate the antidepressant effects of physical exercise. Studies on the neuroprotective effects of adiponectin have revealed potential innovative treatments for Alzheimer's disease. Here, we reviewed the functions of adiponectin and its receptor in the brains of human and animal models of cognitive impairment. We summarized the role of adiponectin in Alzheimer's disease, focusing on its impact on energy metabolism, insulin resistance, and inflammation. We also discuss how exercise increases adiponectin secretion and its potential benefits for learning and memory. Finally, we highlight the latest research on chemical compounds that mimic exercise-enhanced secretion of adiponectin and its receptor in Alzheimer's disease.
Collapse
Affiliation(s)
- Hui-Hui Guo
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- Department of Rehabilitation Medicine, Shaoxing People’s Hospital, Shaoxing, Zhejiang Province, China
| | - Hai-Ning Ou
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- Department of Rehabilitation, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jia-Sui Yu
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Julia Macedo Rosa
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Douglas Affonso Formolo
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Tong Cheng
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Hector Wing Hong Tsang
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
37
|
Yu N, Zhao Y, Wang P, Zhang F, Wen C, Wang S. Changes in border-associated macrophages after stroke: Single-cell sequencing analysis. Neural Regen Res 2026; 21:346-356. [PMID: 39927762 PMCID: PMC12094533 DOI: 10.4103/nrr.nrr-d-24-01092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/09/2024] [Accepted: 12/27/2024] [Indexed: 02/11/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202601000-00038/figure1/v/2025-06-09T151831Z/r/image-tiff Border-associated macrophages are located at the interface between the brain and the periphery, including the perivascular spaces, choroid plexus, and meninges. Until recently, the functions of border-associated macrophages have been poorly understood and largely overlooked. However, a recent study reported that border-associated macrophages participate in stroke-induced inflammation, although many details and the underlying mechanisms remain unclear. In this study, we performed a comprehensive single-cell analysis of mouse border-associated macrophages using sequencing data obtained from the Gene Expression Omnibus (GEO) database (GSE174574 and GSE225948). Differentially expressed genes were identified, and enrichment analysis was performed to identify the transcription profile of border-associated macrophages. CellChat analysis was conducted to determine the cell communication network of border-associated macrophages. Transcription factors were predicted using the 'pySCENIC' tool. We found that, in response to hypoxia, border-associated macrophages underwent dynamic transcriptional changes and participated in the regulation of inflammatory-related pathways. Notably, the tumor necrosis factor pathway was activated by border-associated macrophages following ischemic stroke. The pySCENIC analysis indicated that the activity of signal transducer and activator of transcription 3 (Stat3) was obviously upregulated in stroke, suggesting that Stat3 inhibition may be a promising strategy for treating border-associated macrophages-induced neuroinflammation. Finally, we constructed an animal model to investigate the effects of border-associated macrophages depletion following a stroke. Treatment with liposomes containing clodronate significantly reduced infarct volume in the animals and improved neurological scores compared with untreated animals. Taken together, our results demonstrate comprehensive changes in border-associated macrophages following a stroke, providing a theoretical basis for targeting border-associated macrophages-induced neuroinflammation in stroke treatment.
Collapse
Affiliation(s)
- Ning Yu
- Department of Anesthesiology, Shandong Provincial Key Medical and Health Laboratory of Anesthesia and Brain Function (The Affiliated Hospital of Qingdao University), The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Yang Zhao
- Department of Anesthesiology, Shandong Provincial Key Medical and Health Laboratory of Anesthesia and Brain Function (The Affiliated Hospital of Qingdao University), The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Peng Wang
- Department of Anesthesiology, Shandong Provincial Key Medical and Health Laboratory of Anesthesia and Brain Function (The Affiliated Hospital of Qingdao University), The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Fuqiang Zhang
- Department of Anesthesiology, Shandong Provincial Key Medical and Health Laboratory of Anesthesia and Brain Function (The Affiliated Hospital of Qingdao University), The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Cuili Wen
- Department of Anesthesiology, Shandong Provincial Key Medical and Health Laboratory of Anesthesia and Brain Function (The Affiliated Hospital of Qingdao University), The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Shilei Wang
- Department of Anesthesiology, Shandong Provincial Key Medical and Health Laboratory of Anesthesia and Brain Function (The Affiliated Hospital of Qingdao University), The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
38
|
Negro S, Montecucco C, Rigoni M. Extra-pineal melatonin in perisynaptic Schwann cell-muscle fiber cross talk at the regenerating neuromuscular junction. Neural Regen Res 2026; 21:300-301. [PMID: 40489345 DOI: 10.4103/nrr.nrr-d-24-01136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/07/2024] [Indexed: 06/11/2025] Open
Affiliation(s)
- Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua, Italy (Negro S, Montecucco C, Rigoni M)
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy (Negro S, Montecucco C, Rigoni M)
- CNR Institute of Neuroscience, Padua, Italy (Montecucco C)
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua, Italy (Negro S, Montecucco C, Rigoni M)
| |
Collapse
|
39
|
She K, Yuan N, Huang M, Zhu W, Tang M, Ma Q, Chen J. Emerging role of microglia in the developing dopaminergic system: Perturbation by early life stress. Neural Regen Res 2026; 21:126-140. [PMID: 39589170 PMCID: PMC12094535 DOI: 10.4103/nrr.nrr-d-24-00742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Early life stress correlates with a higher prevalence of neurological disorders, including autism, attention-deficit/hyperactivity disorder, schizophrenia, depression, and Parkinson's disease. These conditions, primarily involving abnormal development and damage of the dopaminergic system, pose significant public health challenges. Microglia, as the primary immune cells in the brain, are crucial in regulating neuronal circuit development and survival. From the embryonic stage to adulthood, microglia exhibit stage-specific gene expression profiles, transcriptome characteristics, and functional phenotypes, enhancing the susceptibility to early life stress. However, the role of microglia in mediating dopaminergic system disorders under early life stress conditions remains poorly understood. This review presents an up-to-date overview of preclinical studies elucidating the impact of early life stress on microglia, leading to dopaminergic system disorders, along with the underlying mechanisms and therapeutic potential for neurodegenerative and neurodevelopmental conditions. Impaired microglial activity damages dopaminergic neurons by diminishing neurotrophic support (e.g., insulin-like growth factor-1) and hinders dopaminergic axon growth through defective phagocytosis and synaptic pruning. Furthermore, blunted microglial immunoreactivity suppresses striatal dopaminergic circuit development and reduces neuronal transmission. Furthermore, inflammation and oxidative stress induced by activated microglia can directly damage dopaminergic neurons, inhibiting dopamine synthesis, reuptake, and receptor activity. Enhanced microglial phagocytosis inhibits dopamine axon extension. These long-lasting effects of microglial perturbations may be driven by early life stress-induced epigenetic reprogramming of microglia. Indirectly, early life stress may influence microglial function through various pathways, such as astrocytic activation, the hypothalamic-pituitary-adrenal axis, the gut-brain axis, and maternal immune signaling. Finally, various therapeutic strategies and molecular mechanisms for targeting microglia to restore the dopaminergic system were summarized and discussed. These strategies include classical antidepressants and antipsychotics, antibiotics and anti-inflammatory agents, and herbal-derived medicine. Further investigations combining pharmacological interventions and genetic strategies are essential to elucidate the causal role of microglial phenotypic and functional perturbations in the dopaminergic system disrupted by early life stress.
Collapse
Affiliation(s)
- Kaijie She
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Naijun Yuan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- Shenzhen People’s Hospital, The 2 Clinical Medical College, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Shenzhen, Guangdong Province, China
| | - Minyi Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Wenjun Zhu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Manshi Tang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
40
|
Martinez-Salas E, Francisco-Velilla R. GEMIN5 and neurodevelopmental diseases: From functional insights to disease perception. Neural Regen Res 2026; 21:187-194. [PMID: 39819844 PMCID: PMC12094563 DOI: 10.4103/nrr.nrr-d-24-01010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/17/2024] [Accepted: 11/27/2024] [Indexed: 01/19/2025] Open
Abstract
GEMIN5 is a predominantly cytoplasmic multifunctional protein, known to be involved in recognizing snRNAs through its WD40 repeats domain placed at the N-terminus. A dimerization domain in the middle region acts as a hub for protein-protein interaction, while a non-canonical RNA-binding site is placed towards the C-terminus. The singular organization of structural domains present in GEMIN5 enables this protein to perform multiple functions through its ability to interact with distinct partners, both RNAs and proteins. This protein exerts a different role in translation regulation depending on its physiological state, such that while GEMIN5 down-regulates global RNA translation, the C-terminal half of the protein promotes translation of its mRNA. Additionally, GEMIN5 is responsible for the preferential partitioning of mRNAs into polysomes. Besides selective translation, GEMIN5 forms part of distinct ribonucleoprotein complexes, reflecting the dynamic organization of macromolecular complexes in response to internal and external signals. In accordance with its contribution to fundamental cellular processes, recent reports described clinical loss of function mutants suggesting that GEMIN5 deficiency is detrimental to cell growth and survival. Remarkably, patients carrying GEMIN5 biallelic variants suffer from neurodevelopmental delay, hypotonia, and cerebellar ataxia. Molecular analyses of individual variants, which are defective in protein dimerization, display decreased levels of ribosome association, reinforcing the involvement of the protein in translation regulation. Importantly, the number of clinical variants and the phenotypic spectrum associated with GEMIN5 disorders is increasing as the knowledge of the protein functions and the pathways linked to its activity augments. Here we discuss relevant advances concerning the functional and structural features of GEMIN5 and its separate domains in RNA-binding, protein interactome, and translation regulation, and how these data can help to understand the involvement of protein malfunction in clinical variants found in patients developing neurodevelopmental disorders.
Collapse
|
41
|
Liu Y, Ding X, Jia S, Gu X. Current understanding and prospects for targeting neurogenesis in the treatment of cognitive impairment. Neural Regen Res 2026; 21:141-155. [PMID: 39820472 PMCID: PMC12094536 DOI: 10.4103/nrr.nrr-d-24-00802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 07/24/2024] [Accepted: 10/31/2024] [Indexed: 01/19/2025] Open
Abstract
Adult hippocampal neurogenesis is linked to memory formation in the adult brain, with new neurons in the hippocampus exhibiting greater plasticity during their immature stages compared to mature neurons. Abnormal adult hippocampal neurogenesis is closely associated with cognitive impairment in central nervous system diseases. Targeting and regulating adult hippocampal neurogenesis have been shown to improve cognitive deficits. This review aims to expand the current understanding and prospects of targeting neurogenesis in the treatment of cognitive impairment. Recent research indicates the presence of abnormalities in AHN in several diseases associated with cognitive impairment, including cerebrovascular diseases, Alzheimer's disease, aging-related conditions, and issues related to anesthesia and surgery. The role of these abnormalities in the cognitive deficits caused by these diseases has been widely recognized, and targeting AHN is considered a promising approach for treating cognitive impairment. However, the underlying mechanisms of this role are not yet fully understood, and the effectiveness of targeting abnormal adult hippocampal neurogenesis for treatment remains limited, with a need for further development of treatment methods and detection techniques. By reviewing recent studies, we classify the potential mechanisms of adult hippocampal neurogenesis abnormalities into four categories: immunity, energy metabolism, aging, and pathological states. In immunity-related mechanisms, abnormalities in meningeal, brain, and peripheral immunity can disrupt normal adult hippocampal neurogenesis. Lipid metabolism and mitochondrial function disorders are significant energy metabolism factors that lead to abnormal adult hippocampal neurogenesis. During aging, the inflammatory state of the neurogenic niche and the expression of aging-related microRNAs contribute to reduced adult hippocampal neurogenesis and cognitive impairment in older adult patients. Pathological states of the body and emotional disorders may also result in abnormal adult hippocampal neurogenesis. Among the current strategies used to enhance this form of neurogenesis, physical therapies such as exercise, transcutaneous electrical nerve stimulation, and enriched environments have proven effective. Dietary interventions, including energy intake restriction and nutrient optimization, have shown efficacy in both basic research and clinical trials. However, drug treatments, such as antidepressants and stem cell therapy, are primarily reported in basic research, with limited clinical application. The relationship between abnormal adult hippocampal neurogenesis and cognitive impairment has garnered widespread attention, and targeting the former may be an important strategy for treating the latter. However, the mechanisms underlying abnormal adult hippocampal neurogenesis remain unclear, and treatments are lacking. This highlights the need for greater focus on translating research findings into clinical practice.
Collapse
Affiliation(s)
- Ye Liu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| | - Xibing Ding
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shushan Jia
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| | - Xiyao Gu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| |
Collapse
|
42
|
Riffo-Lepe N, González-Sanmiguel J, Armijo-Weingart L, Saavedra-Sieyes P, Hernandez D, Ramos G, San Martín LS, Aguayo LG. Synaptic and synchronic impairments in subcortical brain regions associated with early non-cognitive dysfunction in Alzheimer's disease. Neural Regen Res 2026; 21:248-264. [PMID: 39885666 PMCID: PMC12094569 DOI: 10.4103/nrr.nrr-d-24-01052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/16/2024] [Accepted: 12/21/2024] [Indexed: 02/01/2025] Open
Abstract
For many decades, Alzheimer's disease research has primarily focused on impairments within cortical and hippocampal regions, which are thought to be related to cognitive dysfunctions such as memory and language deficits. The exact cause of Alzheimer's disease is still under debate, making it challenging to establish an effective therapy or early diagnosis. It is widely accepted that the accumulation of amyloid-beta peptide in the brain parenchyma leads to synaptic dysfunction, a critical step in Alzheimer's disease development. The traditional amyloid cascade model is initiated by accumulating extracellular amyloid-beta in brain areas essential for memory and language. However, while it is possible to reduce the presence of amyloid-beta plaques in the brain with newer immunotherapies, cognitive symptoms do not necessarily improve. Interestingly, recent studies support the notion that early alterations in subcortical brain regions also contribute to brain damage and precognitive decline in Alzheimer's disease. A body of recent evidence suggests that early Alzheimer's disease is associated with alterations (e.g., motivation, anxiety, and motor impairment) in subcortical areas, such as the striatum and amygdala, in both human and animal models. Also, recent data indicate that intracellular amyloid-beta appears early in subcortical regions such as the nucleus accumbens, locus coeruleus, and raphe nucleus, even without extracellular amyloid plaques. The reported effects are mainly excitatory, increasing glutamatergic transmission and neuronal excitability. In agreement, data in Alzheimer's disease patients and animal models show an increase in neuronal synchronization that leads to electroencephalogram disturbances and epilepsy. The data indicate that early subcortical brain dysfunctions might be associated with non-cognitive symptoms such as anxiety, irritability, and motivation deficits, which precede memory loss and language alterations. Overall, the evidence reviewed suggests that subcortical brain regions could explain early dysfunctions and perhaps be targets for therapies to slow disease progression. Future research should focus on these non-traditional brain regions to reveal early pathological alterations and underlying mechanisms to advance our understanding of Alzheimer's disease beyond the traditionally studied hippocampal and cortical circuits.
Collapse
Affiliation(s)
- Nicolás Riffo-Lepe
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Juliana González-Sanmiguel
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Lorena Armijo-Weingart
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Concepción, Chile
| | - Paulina Saavedra-Sieyes
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - David Hernandez
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Gerson Ramos
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Loreto S. San Martín
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| | - Luis G. Aguayo
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| |
Collapse
|
43
|
Chocarro J, Lanciego JL. Adeno-associated viral vectors for modeling Parkinson's disease in non-human primates. Neural Regen Res 2026; 21:224-232. [PMID: 39885675 PMCID: PMC12094566 DOI: 10.4103/nrr.nrr-d-24-00896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/29/2024] [Accepted: 12/18/2024] [Indexed: 02/01/2025] Open
Abstract
The development of clinical candidates that modify the natural progression of sporadic Parkinson's disease and related synucleinopathies is a praiseworthy endeavor, but extremely challenging. Therapeutic candidates that were successful in preclinical Parkinson's disease animal models have repeatedly failed when tested in clinical trials. While these failures have many possible explanations, it is perhaps time to recognize that the problem lies with the animal models rather than the putative candidate. In other words, the lack of adequate animal models of Parkinson's disease currently represents the main barrier to preclinical identification of potential disease-modifying therapies likely to succeed in clinical trials. However, this barrier may be overcome by the recent introduction of novel generations of viral vectors coding for different forms of alpha-synuclein species and related genes. Although still facing several limitations, these models have managed to mimic the known neuropathological hallmarks of Parkinson's disease with unprecedented accuracy, delineating a more optimistic scenario for the near future.
Collapse
Affiliation(s)
- Julia Chocarro
- CNS Gene Therapy Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed-ISCIII), Madrid, Spain
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - José L. Lanciego
- CNS Gene Therapy Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed-ISCIII), Madrid, Spain
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
44
|
Geng R, Wang Y, Wang R, Wu J, Bao X. Enhanced neurogenesis after ischemic stroke: The interplay between endogenous and exogenous stem cells. Neural Regen Res 2026; 21:212-223. [PMID: 39820432 PMCID: PMC12094570 DOI: 10.4103/nrr.nrr-d-24-00879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/02/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025] Open
Abstract
Ischemic stroke is a significant global health crisis, frequently resulting in disability or death, with limited therapeutic interventions available. Although various intrinsic reparative processes are initiated within the ischemic brain, these mechanisms are often insufficient to restore neuronal functionality. This has led to intensive investigation into the use of exogenous stem cells as a potential therapeutic option. This comprehensive review outlines the ontogeny and mechanisms of activation of endogenous neural stem cells within the adult brain following ischemic events, with focus on the impact of stem cell-based therapies on neural stem cells. Exogenous stem cells have been shown to enhance the proliferation of endogenous neural stem cells via direct cell-to-cell contact and through the secretion of growth factors and exosomes. Additionally, implanted stem cells may recruit host stem cells from their niches to the infarct area by establishing so-called "biobridges." Furthermore, xenogeneic and allogeneic stem cells can modify the microenvironment of the infarcted brain tissue through immunomodulatory and angiogenic effects, thereby supporting endogenous neuroregeneration. Given the convergence of regulatory pathways between exogenous and endogenous stem cells and the necessity for a supportive microenvironment, we discuss three strategies to simultaneously enhance the therapeutic efficacy of both cell types. These approaches include: (1) co-administration of various growth factors and pharmacological agents alongside stem cell transplantation to reduce stem cell apoptosis; (2) synergistic administration of stem cells and their exosomes to amplify paracrine effects; and (3) integration of stem cells within hydrogels, which provide a protective scaffold for the implanted cells while facilitating the regeneration of neural tissue and the reconstitution of neural circuits. This comprehensive review highlights the interactions and shared regulatory mechanisms between endogenous neural stem cells and exogenously implanted stem cells and may offer new insights for improving the efficacy of stem cell-based therapies in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ruxu Geng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuhe Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Wu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| |
Collapse
|
45
|
Dukhinova MS, Guo J, Shen E, Liu W, Huang W, Shen Y, Wang L. Cerebellar microglia: On the edge between neuroinflammation and neuroregulation. Neural Regen Res 2026; 21:156-172. [PMID: 40489344 DOI: 10.4103/nrr.nrr-d-24-00550] [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: 06/17/2024] [Accepted: 09/14/2024] [Indexed: 06/11/2025] Open
Abstract
The cerebellum is receiving increasing attention for its cognitive, emotional, and social functions, as well as its unique metabolic profiles. Cerebellar microglia exhibit specialized and highly immunogenic phenotypes under both physiological and pathological conditions. These immune cells communicate with intrinsic and systemic factors and contribute to the structural and functional compartmentalization of the cerebellum. In this review, we discuss the roles of microglia in the cerebellar microenvironment, neuroinflammation, cerebellar adaptation, and neuronal activity, the associated molecular and cellular mechanisms, and potential therapeutic strategies targeting cerebellar microglia in the context of neuroinflammation. Future directions and unresolved questions in this field are further highlighted, particularly regarding therapeutic interventions targeting cerebellar microglia, functional mechanisms and activities of microglia in the cerebellar circuitry, neuronal connectivity, and neurofunctional outcomes of their activity. Cerebellar morphology and neuronal performance are influenced by both intrinsic and systemic factors that are actively monitored by microglia in both healthy and diseased states. Under pathological conditions, local subsets of microglia exhibit diverse responses to the altered microenvironment that contribute to the structural and functional compartmentalization of the cerebellum. Microglia in the cerebellum undergo early maturation during the embryonic stage and display specialized, highly immunogenic phenotypes. In summary, cerebellar microglia have the capacity to serve as regulatory tools that influence outcomes across a wide range of neurological and systemic conditions, including neurodevelopmental, neurodegenerative, metabolic, and stress-related disorders.
Collapse
Affiliation(s)
- Marina S Dukhinova
- Center for Brain Health, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, Zhejiang Province, China
| | - Jingwen Guo
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Enwei Shen
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Wanting Liu
- Center for Brain Health, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, Zhejiang Province, China
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Wanqi Huang
- Center for Brain Health, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, Zhejiang Province, China
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Shen
- Center for Brain Health, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, Zhejiang Province, China
- Department of Physiology and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Luxi Wang
- Center for Brain Health, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institute of Medicine, Zhejiang University, Yiwu, Zhejiang Province, China
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
46
|
Tang C, Wang P, Li Z, Zhong S, Yang L, Li G. Neural functional rehabilitation: Exploring neuromuscular reconstruction technology advancements and challenges. Neural Regen Res 2026; 21:173-186. [PMID: 39665789 PMCID: PMC12094537 DOI: 10.4103/nrr.nrr-d-24-00613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/18/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Neural machine interface technology is a pioneering approach that aims to address the complex challenges of neurological dysfunctions and disabilities resulting from conditions such as congenital disorders, traumatic injuries, and neurological diseases. Neural machine interface technology establishes direct connections with the brain or peripheral nervous system to restore impaired motor, sensory, and cognitive functions, significantly improving patients' quality of life. This review analyzes the chronological development and integration of various neural machine interface technologies, including regenerative peripheral nerve interfaces, targeted muscle and sensory reinnervation, agonist-antagonist myoneural interfaces, and brain-machine interfaces. Recent advancements in flexible electronics and bioengineering have led to the development of more biocompatible and high-resolution electrodes, which enhance the performance and longevity of neural machine interface technology. However, significant challenges remain, such as signal interference, fibrous tissue encapsulation, and the need for precise anatomical localization and reconstruction. The integration of advanced signal processing algorithms, particularly those utilizing artificial intelligence and machine learning, has the potential to improve the accuracy and reliability of neural signal interpretation, which will make neural machine interface technologies more intuitive and effective. These technologies have broad, impactful clinical applications, ranging from motor restoration and sensory feedback in prosthetics to neurological disorder treatment and neurorehabilitation. This review suggests that multidisciplinary collaboration will play a critical role in advancing neural machine interface technologies by combining insights from biomedical engineering, clinical surgery, and neuroengineering to develop more sophisticated and reliable interfaces. By addressing existing limitations and exploring new technological frontiers, neural machine interface technologies have the potential to revolutionize neuroprosthetics and neurorehabilitation, promising enhanced mobility, independence, and quality of life for individuals with neurological impairments. By leveraging detailed anatomical knowledge and integrating cutting-edge neuroengineering principles, researchers and clinicians can push the boundaries of what is possible and create increasingly sophisticated and long-lasting prosthetic devices that provide sustained benefits for users.
Collapse
Affiliation(s)
- Chunxiao Tang
- Department of Neural Engineering Center, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong Province, China
| | - Ping Wang
- Department of Neural Engineering Center, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong Province, China
| | - Zhonghua Li
- Department of Human Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shizhen Zhong
- Department of Human Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lin Yang
- Department of Neural Engineering Center, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong Province, China
| | - Guanglin Li
- Department of Neural Engineering Center, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong Province, China
| |
Collapse
|
47
|
Lian X, Liu Z, Gan Z, Yan Q, Tong L, Qiu L, Liu Y, Chen JF, Li Z. Targeting the glymphatic system to promote α-synuclein clearance: a novel therapeutic strategy for Parkinson's disease. Neural Regen Res 2026; 21:233-247. [PMID: 39819820 PMCID: PMC12094544 DOI: 10.4103/nrr.nrr-d-24-00764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/23/2024] [Accepted: 09/05/2024] [Indexed: 01/19/2025] Open
Abstract
The excessive buildup of neurotoxic α-synuclein plays a pivotal role in the pathogenesis of Parkinson's disease, highlighting the urgent need for innovative therapeutic strategies to promote α-synuclein clearance, particularly given the current lack of disease-modifying treatments. The glymphatic system, a recently identified perivascular fluid transport network, is crucial for clearing neurotoxic proteins. This review aims to synthesize current knowledge on the role of the glymphatic system in α-synuclein clearance and its implications for the pathology of Parkinson's disease while emphasizing potential therapeutic strategies and areas for future research. The review begins with an overview of the glymphatic system and details its anatomical structure and physiological functions that facilitate cerebrospinal fluid circulation and waste clearance. It summarizes emerging evidence from neuroimaging and experimental studies that highlight the close correlation between the glymphatic system and clinical symptom severity in patients with Parkinson's disease, as well as the effect of glymphatic dysfunction on α-synuclein accumulation in Parkinson's disease models. Subsequently, the review summarizes the mechanisms of glymphatic system impairment in Parkinson's disease, including sleep disturbances, aquaporin-4 impairment, and mitochondrial dysfunction, all of which diminish glymphatic system efficiency. This creates a vicious cycle that exacerbates α-synuclein accumulation and worsens Parkinson's disease. The therapeutic perspectives section outlines strategies for enhancing glymphatic activity, such as improving sleep quality and pharmacologically targeting aquaporin-4 or its subcellular localization. Promising interventions include deep brain stimulation, melatonin supplementation, γ-aminobutyric acid modulation, and non-invasive methods (such as exercise and bright-light therapy), multisensory γ stimulation, and ultrasound therapy. Moreover, identifying neuroimaging biomarkers to assess glymphatic flow as an indicator of α-synuclein burden could refine Parkinson's disease diagnosis and track disease progression. In conclusion, the review highlights the critical role of the glymphatic system in α-synuclein clearance and its potential as a therapeutic target in Parkinson's disease. It advocates for further research to elucidate the specific mechanisms by which the glymphatic system clears misfolded α-synuclein and the development of imaging biomarkers to monitor glymphatic activity in patients with Parkinson's disease. Findings from this review suggest that enhancing glymphatic clearance is a promising strategy for reducing α-synuclein deposits and mitigating the progression of Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoyue Lian
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhenghao Liu
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zuobin Gan
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qingshan Yan
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Luyao Tong
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Linan Qiu
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuntao Liu
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiang-fan Chen
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhihui Li
- Molecular Neuropharmacology Laboratory and Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
48
|
Chilosi M, Piciucchi S, Ravaglia C, Spagnolo P, Sverzellati N, Tomassetti S, Wuyts W, Poletti V. "Alveolar stem cell exhaustion, fibrosis and bronchiolar proliferation" related entities. A narrative review. Pulmonology 2025; 31:2416847. [PMID: 39277539 DOI: 10.1016/j.pulmoe.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 09/17/2024] Open
Affiliation(s)
- M Chilosi
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì I
| | - S Piciucchi
- Department of Radiology, Ospedale GB Morgagni, Forlì I
| | - C Ravaglia
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì (I); DIMEC, Bologna University, Forlì Campus, Forlì I, Department
| | - P Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - N Sverzellati
- Scienze Radiologiche, Department of Medicine and Surgery, University Hospital Parma, Parma, Italy
| | - S Tomassetti
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - W Wuyts
- Pulmonology Department, UZ Leuven, Leuven, Belgium
| | - V Poletti
- Department of Medical Specialities/Pulmonology Ospedale GB Morgagni, Forlì (I); DIMEC, Bologna University, Forlì Campus, Forlì I, Department
- Department of Respiratory Diseases & Allergy, Aarhus University, Aarhus, Denmark
| |
Collapse
|
49
|
Champigneulle B, Stauffer E, Robach P, Doutreleau S, Howe CA, Pina A, Salazar-Granara AA, Hancco I, Guergour D, Brugniaux JV, Connes P, Pichon A, Verges S. Early effects of acetazolamide on hemoglobin mass and plasma volume in chronic mountain sickness at 5100 m. Pulmonology 2025; 31:2416794. [PMID: 37263861 DOI: 10.1016/j.pulmoe.2023.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/19/2023] [Accepted: 05/10/2023] [Indexed: 06/03/2023] Open
Abstract
INTRODUCTION AND OBJECTIVES Chronic Mountain Sickness (CMS) syndrome, combining excessive erythrocytosis and clinical symptoms in highlanders, remains a public health concern in high-altitude areas, especially in the Andes, with limited therapeutic approaches. The objectives of this study were to assess in CMS-highlanders permanently living in La Rinconada (5100-5300 m, Peru, the highest city in the world), the early efficacy of acetazolamide (ACZ) and atorvastatin to reduce hematocrit (Hct), as well as the underlying mechanisms focusing on intravascular volumes. MATERIALS AND METHODS Forty-one males (46±8 years of age) permanently living in La Rinconada for 15 [10-20] years and suffering from CMS were randomized between ACZ (250 mg once-daily; N = 13), atorvastatin (20 mg once-daily; N = 14) or placebo (N = 14) uptake in a double-blinded parallel study. Hematocrit (primary endpoint) as well as arterial blood gasses, total hemoglobin mass (Hbmass) and intravascular volumes were assessed at baseline and after a mean (±SD) treatment duration of 19±2 days. RESULTS ACZ increased PaO2 by +13.4% (95% CI: 4.3 to 22.5%) and decreased Hct by -5.2% (95% CI: -8.3 to -2.2%), whereas Hct remained unchanged with placebo or atorvastatin. ACZ tended to decrease Hbmass (-2.6%, 95% CI: -5.7 to 0.5%), decreased total red blood cell volume (RBCV, -5.3%, 95% CI: -10.3 to -0.3%) and increased plasma volume (PV, +17.6%, 95% CI: 4.9 to 30.3%). Atorvastatin had no effect on intravascular volumes, while Hbmass and RBCV increased in the placebo group (+6.1%, 95% CI: 4.2 to 7.9% and +7.0%, 95%CI: 2.7 to 11.4%, respectively). CONCLUSIONS Short-term ACZ uptake was effective to reduce Hct in CMS-highlanders living at extreme altitude >5,000 m and was associated with both an increase in PV and a reduction in RBCV.
Collapse
Affiliation(s)
- B Champigneulle
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | - E Stauffer
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
- Exploration Fonctionnelle Respiratoire, Médecine du Sport et de l'Activité Physique, Hospices Civils de Lyon, Hôpital Croix Rousse, Lyon, France
| | - P Robach
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
- National School for Mountain Sports, Site of the National School for Skiing and Mountaineering (ENSA), Chamonix, France
| | - S Doutreleau
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | - C A Howe
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia - Okanagan, Kelowna, Canada
| | - A Pina
- Department of Cardiovascular, Neural and Metabolic Sciences, Istituto Auxologico Italiano, IRCCS, S. Luca Hospital, Milan, Italy
| | - A A Salazar-Granara
- Universidad de San Martin de Porres, School of Medicine, Research Centre in Altitude Medicine, Lima, Peru
| | - I Hancco
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | - D Guergour
- Unité Biochimie Immunoanalyse, Service de Biochimie Biologie Moléculaire et Toxicologie Environnementale, Institut de Biologie et Pathologie, CHU Grenoble Alpes, France
| | - J V Brugniaux
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | - P Connes
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, France
- Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - A Pichon
- Laboratoire Move EA 6314, Faculté des Sciences du Sport, Universit. De Poitiers, Poitiers, France
| | - S Verges
- Univ. Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| |
Collapse
|
50
|
Münz C, Campbell GR, Esclatine A, Faure M, Labonte P, Lussignol M, Orvedahl A, Altan-Bonnet N, Bartenschlager R, Beale R, Cirone M, Espert L, Jung J, Leib D, Reggiori F, Sanyal S, Spector SA, Thiel V, Viret C, Wei Y, Wileman T, Wodrich H. Autophagy machinery as exploited by viruses. AUTOPHAGY REPORTS 2025; 4:27694127.2025.2464986. [PMID: 40201908 PMCID: PMC11921968 DOI: 10.1080/27694127.2025.2464986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
Viruses adapt and modulate cellular pathways to allow their replication in host cells. The catabolic pathway of macroautophagy, for simplicity referred to as autophagy, is no exception. In this review, we discuss anti-viral functions of both autophagy and select components of the autophagy machinery, and how viruses have evaded them. Some viruses use the membrane remodeling ability of the autophagy machinery to build their replication compartments in the cytosol or efficiently egress from cells in a non-lytic fashion. Some of the autophagy machinery components and their remodeled membranes can even be found in viral particles as envelopes or single membranes around virus packages that protect them during spreading and transmission. Therefore, studies on autophagy regulation by viral infections can reveal functions of the autophagy machinery beyond lysosomal degradation of cytosolic constituents. Furthermore, they can also pinpoint molecular interactions with which the autophagy machinery can most efficiently be manipulated, and this may be relevant to develop effective disease treatments based on autophagy modulation.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich Switzerland
| | - Grant R Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of SD, Vermillion, SD, USA
| | - Audrey Esclatine
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Patrick Labonte
- eINRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada
| | - Marion Lussignol
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anthony Orvedahl
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Bartenschlager
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division Virus-Associated Carcinogenesis, Heidelberg, Germany
- German Centre for Infection Research, Heidelberg partner site, Heidelberg, Germany
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
- Division of Medicine, University College London, London, UK
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucile Espert
- University of Montpellier, Montpellier, France
- CNRS, Institut de Recherche enInfectiologie deMontpellier (IRIM), Montpellier, France
| | - Jae Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - David Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH, USA
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, Aarhus C, Denmark
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, UK
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital, San Diego, CA, USA
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland, and Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Yu Wei
- Institut Pasteur-Theravectys Joint Laboratory, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia
- Quadram Institute Bioscience, Norwich Research Park, Norfolk, UK
| | - Harald Wodrich
- sLaboratoire de Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR, Université de Bordeaux, Bordeaux, France
| |
Collapse
|