1
|
Chen X, Jiang J, He B, Luo S, Tan Q, Yao Y, Wan R, Xu H, Liu S, Pan X, Chen X, Li J. Piezo1 aggravates ischemia/reperfusion-induced acute kidney injury by Ca 2+-dependent calpain/HIF-1α/Notch signaling. Ren Fail 2025; 47:2447801. [PMID: 39780511 PMCID: PMC11721879 DOI: 10.1080/0886022x.2024.2447801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Macrophages play a vital role in the inflammation and repair processes of ischemia/reperfusion-induced acute kidney injury (IR-AKI). The mechanosensitive ion channel Piezo1 is significant in these inflammatory processes. However, the exact role of macrophage Piezo1 in IR-AKI is unknown. The main purpose of this study was to determine the role of macrophage Piezo1 in the injury and repair process in IR-AKI. Genetically modified mice with targeted knockout of Piezo1 in myeloid cells were established, and acute kidney injury was induced by bilateral renal vascular clamping surgery. Additionally, hypoxia treatment was performed on bone marrow-derived macrophages in vitro. Our data indicate that Piezo1 is upregulated in renal macrophages in mice with IR-AKI. Myeloid Piezo1 knockout provided protective effects in mice with IR-AKI. Mechanistically, the regulatory effects of Piezo1 on macrophages are at least partially linked to calpain signaling. Piezo1 activates Ca2+-dependent calpain signaling, which critically upregulates HIF-1α signaling. This key pathway subsequently influences the Notch and CCL2/CCR2 pathways, driving the polarization of M1 macrophages. In conclusion, our findings elucidate the biological functions of Piezo1 in renal macrophages, underscoring its role as a crucial mediator of acute kidney injury. Consequently, the genetic or pharmacological inhibition of Piezo1 presents a promising strategy for treating IR-AKI.
Collapse
Affiliation(s)
- Xiaoting Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jintao Jiang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin He
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shangfei Luo
- Medical Research Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiaorui Tan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Youfen Yao
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rentao Wan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglin Xu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Silin Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianmei Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Chen
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Li
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- Medical Research Center, Shandong University of Traditional Chinese Medicine, Jinan, China
- Faculty of Biological Sciences, University of Leeds, UK
| |
Collapse
|
2
|
Li Z, Sun J, Jia T, Ji L, Li C. Respiratory modulation of beta corticomuscular coherence in isometric hand movements. Cogn Neurodyn 2025; 19:54. [PMID: 40129876 PMCID: PMC11929664 DOI: 10.1007/s11571-025-10245-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/15/2025] [Indexed: 03/26/2025] Open
Abstract
Respiration is a fundamental physiological function in humans, often synchronized with movement to enhance performance and efficiency. Recent studies have underscored the modulatory effects of respiratory rhythms on brain oscillations and various behavioral responses, including sensorimotor processes. In light of this connection, our study aimed to investigate the influence of different respiratory patterns on beta corticomuscular coherence (CMC) during isometric hand flexion and extension. Utilizing electroencephalogram (EEG) and surface electromyography (sEMG), we examined three breathing conditions: normal breathing, deep inspiration, and deep expiration. Two experimental protocols were employed: the first experiment required participants to simultaneously breathe and exert force, while the other involved maintaining a constant force while varying breathing patterns. The results revealed that deep inspiration significantly enhanced beta CMC during respiration-synchronized tasks, whereas normal breathing resulted in higher CMC compared to deep respiration during sustained force exertion. In the second experiment, beta CMC was cyclically modulated by respiratory phase across all breathing conditions. The difference in the outcomes from the two protocols demonstrated a task-specific modulation of respiration on motor control. Overall, these findings indicate the complex dynamics of respiration-related effects on corticomuscular neural communication and provide valuable insights into the mechanisms underpinning the coupling between respiration and motor function. Supplementary Information The online version contains supplementary material available at 10.1007/s11571-025-10245-x.
Collapse
Affiliation(s)
- Zhibin Li
- Lab of Intelligent and Bio-mimetic Machinery, Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Jingyao Sun
- Lab of Intelligent and Bio-mimetic Machinery, Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Tianyu Jia
- Lab of Intelligent and Bio-mimetic Machinery, Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Linhong Ji
- Lab of Intelligent and Bio-mimetic Machinery, Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Chong Li
- School of Clinical Medicine (BTCH), Tsinghua Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
3
|
Wang H, Xu X, Yang Z, Zhang T. Alterations of synaptic plasticity and brain oscillation are associated with autophagy induced synaptic pruning during adolescence. Cogn Neurodyn 2025; 19:2. [PMID: 39749102 PMCID: PMC11688264 DOI: 10.1007/s11571-024-10185-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Adolescent brain development is characterized by significant anatomical and physiological alterations, but little is known whether and how these alterations impact the neural network. Here we investigated the development of functional networks by measuring synaptic plasticity and neural synchrony of local filed potentials (LFPs), and further explored the underlying mechanisms. LFPs in the hippocampus were recorded in young (21 ~ 25 days), adolescent (1.5 months) and adult (3 months) rats. Long term potentiation (LTP) and neural synchrony were analyzed. The results showed that the LTP was the lowest in adolescent rats. During development, the theta coupling strength was increased progressively but there was no significant change of gamma coupling between young rats and adolescent rats. The density of dendrite spines was decreased progressively during development. The lowest levels of NR2A, NR2B and PSD95 were detected in adolescent rats. Importantly, it was found that the expression levels of autophagy markers were the highest during adolescent compared to that in other developmental stages. Moreover, there were more co-localization of autophagosome and PSD95 in adolescent rats. It suggests that autophagy is possibly involved in synaptic elimination during adolescence, and further impacts synaptic plasticity and neural synchrony.
Collapse
Affiliation(s)
- Hui Wang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 PR China
| | - Xiaxia Xu
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 PR China
| | - Zhuo Yang
- College of Medicine Science, Nankai University, Tianjin, 300071 PR China
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071 PR China
| |
Collapse
|
4
|
Chen L, Hu Y, Wang Z, Zhang L, Jian C, Cheng S, Ming D. Effects of transcutaneous auricular vagus nerve stimulation (taVNS) on motor planning: a multimodal signal study. Cogn Neurodyn 2025; 19:35. [PMID: 39866662 PMCID: PMC11759740 DOI: 10.1007/s11571-025-10220-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/28/2025] Open
Abstract
Motor planning plays a pivotal role in daily life. Transcutaneous auricular vagus nerve stimulation (taVNS) has been demonstrated to enhance decision-making efficiency, illustrating its potential use in cognitive modulation. However, current research primarily focuses on behavioral and single-modal electrophysiological signal, such as electroencephalography (EEG) and electrocardiography (ECG). To investigate the effect of taVNS on motor planning, a total of 21 subjects were recruited for this study and were divided into two groups: active group (n = 10) and sham group (n = 11). Each subject was required to be involved in a single-blind, sham-controlled, between-subject end-state comfort (ESC) experiment. The study compared behavioral indicators and electrophysiological features before and following taVNS. The results indicated a notable reduction in reaction time and an appreciable increase in the proportion of end-state comfort among the participants following taVNS, accompanied by notable alterations in motor-related cortical potential (MRCP) amplitude, low-frequency power of HRV (LF), and cortico-cardiac coherence, particularly in the parietal and occipital regions. These findings show that taVNS may impact the brain and heart, potentially enhancing their interaction, and improve participants' ability of motor planning.
Collapse
Affiliation(s)
- Long Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300392 China
| | - Yihao Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
| | - Zhongpeng Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300392 China
| | - Lei Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300392 China
| | - Chuxiang Jian
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
| | - Shengcui Cheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072 China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300392 China
| | - Dong Ming
- College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, 300072 China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300392 China
| |
Collapse
|
5
|
Agadagba SK, Yau SY, Liang Y, Dalton K, Thompson B. Bidirectional causality of physical exercise in retinal neuroprotection. Neural Regen Res 2025; 20:3400-3415. [PMID: 39688575 PMCID: PMC11974656 DOI: 10.4103/nrr.nrr-d-24-00942] [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: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Physical exercise is recognized as an effective intervention to improve mood, physical performance, and general well-being. It achieves these benefits through cellular and molecular mechanisms that promote the release of neuroprotective factors. Interestingly, reduced levels of physical exercise have been implicated in several central nervous system diseases, including ocular disorders. Emerging evidence has suggested that physical exercise levels are significantly lower in individuals with ocular diseases such as glaucoma, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy. Physical exercise may have a neuroprotective effect on the retina. Therefore, the association between reduced physical exercise and ocular diseases may involve a bidirectional causal relationship whereby visual impairment leads to reduced physical exercise and decreased exercise exacerbates the development of ocular disease. In this review, we summarize the evidence linking physical exercise to eye disease and identify potential mediators of physical exercise-induced retinal neuroprotection. Finally, we discuss future directions for preclinical and clinical research in exercise and eye health.
Collapse
Affiliation(s)
- Stephen K. Agadagba
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Suk-yu Yau
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Ying Liang
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Kristine Dalton
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Benjamin Thompson
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
6
|
Shentu W, Kong Q, Zhang Y, Li W, Chen Q, Yan S, Wang J, Lai Q, Xu Q, Qiao S. Functional abnormalities of the glymphatic system in cognitive disorders. Neural Regen Res 2025; 20:3430-3447. [PMID: 39820293 PMCID: PMC11974647 DOI: 10.4103/nrr.nrr-d-24-01049] [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: 09/06/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 01/19/2025] Open
Abstract
Various pathological mechanisms represent distinct therapeutic targets for cognitive disorders, but a balance between clearance and production is essential for maintaining the stability of the brain's internal environment. Thus, the glymphatic system may represent a common pathway by which to address cognitive disorders. Using the established model of the glymphatic system as our foundation, this review disentangles and analyzes the components of its clearance mechanism, including the initial inflow of cerebrospinal fluid, the mixing of cerebrospinal fluid with interstitial fluid, and the outflow of the mixed fluid and the clearance. Each section summarizes evidence from experimental animal models and human studies, highlighting the normal physiological properties of key structures alongside their pathological manifestations in cognitive disorders. The same pathologic manifestations of different cognitive disorders appearing in the glymphatic system and the same upstream influences are main points of interest of this review. We conclude this article by discussing new findings and outlining the limitations identified in current research progress.
Collapse
Affiliation(s)
- Wuyue Shentu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qi Kong
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Yier Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Wenyao Li
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qiulu Chen
- Department of Neurology, Zhejiang Medical & Health Group Hangzhou Hospital, Hangzhou, Zhejiang Province, China
| | - Sicheng Yan
- Department of Neurology, Liuzhou People’s Hospital, Liuzhou, Guangxi Zhuang Autonomous Region, China
| | - Junjun Wang
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Qilun Lai
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Qi Xu
- Department of Radiology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Song Qiao
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| |
Collapse
|
7
|
Liang Y, Du M, Li X, Gao J, Li Q, Li H, Li J, Gao X, Cong H, Huang Y, Li X, Wang L, Cui J, Gan Y, Tu H. Upregulation of Lactobacillus spp. in gut microbiota as a novel mechanism for environmental eustress-induced anti-pancreatic cancer effects. Gut Microbes 2025; 17:2470372. [PMID: 39988618 PMCID: PMC11853549 DOI: 10.1080/19490976.2025.2470372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with limited effective treatment options. Emerging evidence links enriched environment (EE)-induced eustress to PDAC inhibition. However, the underlying mechanisms remain unclear. In this study, we explored the role of gut microbiota in PDAC-suppressive effects of EE. We demonstrated that depletion of gut microbiota with antibiotics abolished EE-induced tumor suppression, while fecal microbiota transplantation (FMT) from EE mice significantly inhibited tumor growth in both subcutaneous and orthotopic PDAC models housed in standard environment. 16S rRNA sequencing revealed that EE enhanced gut microbiota diversity and selectively enriched probiotic Lactobacillus, particularly L. reuteri. Treatment with L. reuteri significantly suppressed PDAC tumor growth and increased natural killer (NK) cell infiltration into the tumor microenvironment. Depletion of NK cells alleviated the anti-tumor effects of L. reuteri, underscoring the essential role of NK cell-mediated immunity in anti-tumor response. Clinical analysis of PDAC patients showed that higher fecal Lactobacillus abundance correlated with improved progression-free and overall survival, further supporting the therapeutic potential of L. reuteri in PDAC. Overall, this study identifies gut microbiota as a systemic regulator of PDAC under psychological stress. Supplementation of psychobiotic Lactobacillus may offer a novel therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Yiyi Liang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huimin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Gao
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Hui Cong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimeng Huang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinran Li
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiujie Cui
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Gao S, Zhu R, Qin Y, Tang W, Zhou H. Sg-snn: a self-organizing spiking neural network based on temporal information. Cogn Neurodyn 2025; 19:14. [PMID: 39801909 PMCID: PMC11718035 DOI: 10.1007/s11571-024-10199-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Neurodynamic observations indicate that the cerebral cortex evolved by self-organizing into functional networks, These networks, or distributed clusters of regions, display various degrees of attention maps based on input. Traditionally, the study of network self-organization relies predominantly on static data, overlooking temporal information in dynamic neuromorphic data. This paper proposes Temporal Self-Organizing (TSO) method for neuromorphic data processing using a spiking neural network. The TSO method incorporates information from multiple time steps into the selection strategy of the Best Matching Unit (BMU) neurons. It enables the coupled BMUs to radiate the weight across the same layer of neurons, ultimately forming a hierarchical self-organizing topographic map of concern. Additionally, we simulate real neuronal dynamics, introduce a glial cell-mediated Glial-LIF (Leaky Integrate-and-fire) model, and adjust multiple levels of BMUs to optimize the attention topological map.Experiments demonstrate that the proposed Self-organizing Glial Spiking Neural Network (SG-SNN) can generate attention topographies for dynamic event data from coarse to fine. A heuristic method based on cognitive science effectively guides the network's distribution of excitatory regions. Furthermore, the SG-SNN shows improved accuracy on three standard neuromorphic datasets: DVS128-Gesture, CIFAR10-DVS, and N-Caltech 101, with accuracy improvements of 0.3%, 2.4%, and 0.54% respectively. Notably, the recognition accuracy on the DVS128-Gesture dataset reaches 99.3%, achieving state-of-the-art (SOTA) performance.
Collapse
Affiliation(s)
| | | | - Yu Qin
- Shanghai University, Shanghai, China
| | | | - Hao Zhou
- Shanghai University, Shanghai, China
| |
Collapse
|
9
|
Su CW, Yang F, Lai R, Li Y, Naeem H, Yao N, Zhang SP, Zhang H, Li Y, Huang ZG. Unraveling the functional complexity of the locus coeruleus-norepinephrine system: insights from molecular anatomy to neurodynamic modeling. Cogn Neurodyn 2025; 19:29. [PMID: 39866663 PMCID: PMC11757662 DOI: 10.1007/s11571-024-10208-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/08/2024] [Accepted: 09/29/2024] [Indexed: 01/28/2025] Open
Abstract
The locus coeruleus (LC), as the primary source of norepinephrine (NE) in the brain, is central to modulating cognitive and behavioral processes. This review synthesizes recent findings to provide a comprehensive understanding of the LC-NE system, highlighting its molecular diversity, neurophysiological properties, and role in various brain functions. We discuss the heterogeneity of LC neurons, their differential responses to sensory stimuli, and the impact of NE on cognitive processes such as attention and memory. Furthermore, we explore the system's involvement in stress responses and pain modulation, as well as its developmental changes and susceptibility to stressors. By integrating molecular, electrophysiological, and theoretical modeling approaches, we shed light on the LC-NE system's complex role in the brain's adaptability and its potential relevance to neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Chun-Wang Su
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Fan Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Runchen Lai
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Yanhai Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Hadia Naeem
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Nan Yao
- Department of Applied Physics, Xi’an University of Technology, 710054 Shaanxi, China
| | - Si-Ping Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Haiqing Zhang
- Xi’an Children’s Hospital, Xi’an, 710003 Shaanxi China
| | - Youjun Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Zi-Gang Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| |
Collapse
|
10
|
Wang J, Yang X. Dynamic modeling of astrocyte-neuron interactions under the influence of Aβ deposition. Cogn Neurodyn 2025; 19:60. [PMID: 40226235 PMCID: PMC11985881 DOI: 10.1007/s11571-025-10246-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/18/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
β-amyloid (Aβ) protein accumulation is recognized as a key factor in Alzheimer's disease (AD) pathogenesis. Its effects on astrocyte function appear primarily as disturbances to intracellular calcium signaling, which, in turn, affects neuronal excitability. We propose an innovative neuron-astrocyte interaction model to examine how Aβ accumulation influences astrocyte calcium oscillation and neuronal excitability, emphasizing its significance in AD pathogenesis. This comprehensive model describes not only the response of the astrocyte to presynaptic neuron stimulation but also the release of the downstream signaling glutamate and its consequential feedback on neurons. Our research concentrates on changes within two prominent pathways affected by Aβ: the creation of Aβ astrocyte membrane pores and the enhanced sensitivity of ryanodine receptors. By incorporating these adjustments into our astrocyte model, we can reproduce previous experimental findings regarding aberrant astrocyte calcium activity and neural behavior associated with Aβ from a neural computational viewpoint. Within a specified range of Aβ influence, our numerical analysis reveals that astrocyte cytoplasmic calcium rises, calcium oscillation frequency increases, and the time to the first calcium peak shortens, indicating the disrupted astrocyte calcium signaling. Simultaneously, the neuronal firing rate and cytosolic calcium concentration increase while the threshold current for initiating repetitive firing diminishes, implying heightened neuronal excitability. Given that increased neuronal excitability commonly occurs in early AD patients and correlates with cognitive decline, our findings may highlight the importance of Aβ accumulation in AD pathogenesis and provide a theoretical basis for identifying neuronal markers in the early stages of the disease.
Collapse
Affiliation(s)
- JiangNing Wang
- School of Mathematics and Statistics, Shaanxi Normal University, Xi’an, 710119 China
| | - XiaoLi Yang
- School of Mathematics and Statistics, Shaanxi Normal University, Xi’an, 710119 China
| |
Collapse
|
11
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
12
|
He X, Li Y, Xiao X, Li Y, Fang J, Zhou R. Multi-level cognitive state classification of learners using complex brain networks and interpretable machine learning. Cogn Neurodyn 2025; 19:5. [PMID: 39758356 PMCID: PMC11699182 DOI: 10.1007/s11571-024-10203-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/06/2024] [Accepted: 10/13/2024] [Indexed: 01/07/2025] Open
Abstract
Identifying the cognitive state can help educators understand the evolving thought processes of learners, and it is important in promoting the development of higher-order thinking skills (HOTS). Cognitive neuroscience research identifies cognitive states by designing experimental tasks and recording electroencephalography (EEG) signals during task performance. However, most of the previous studies primarily concentrated on extracting features from individual channels in single-type tasks, ignoring the interconnection across channels. In this study, three learning activities (i.e., video watching activity, keyword extracting activity, and essay creating activity) were designed based on a revised Bloom's taxonomy and the Interactive-Constructive-Active-Passive framework and used with 31 college students. The EEG signals were recorded when they were engaged in these activities. First, whole-brain network temporal dynamics were characterized by EEG microstate sequence analysis. Such dynamic changes rely on learning activity and corresponding functional brain systems. Subsequently, phase locking value was used to construct synchrony-based functional brain networks. The network characteristics were extracted to be inputted into different machine learning classifiers: Support Vector Machine, K-Nearest Neighbour, Random Forest, and eXtreme Gradient Boosting (XGBoost). XGBoost showed superior performance in the classification of cognitive states, with an accuracy of 88.07%. Furthermore, SHapley Additive exPlanations (SHAP) was adopted to reveal the connections between different brain regions that contributed to the classification of cognitive state. SHAP analysis reveals that the connections in the frontal, temporal, and central regions are most important for the high cognitive state. Collectively, this study may provide further evidence for educators to design cognitive-guided instructional activities to enhance learners' HOTS.
Collapse
Affiliation(s)
- Xiuling He
- National Engineering Research Center of Educational Big Data, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
- National Engineering Research Center for E-Learning, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
| | - Yue Li
- National Engineering Research Center of Educational Big Data, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
- National Engineering Research Center for E-Learning, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
| | - Xiong Xiao
- National Engineering Research Center of Educational Big Data, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
- National Engineering Research Center for E-Learning, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
| | - Yingting Li
- National Engineering Research Center of Educational Big Data, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
- National Engineering Research Center for E-Learning, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
| | - Jing Fang
- National Engineering Research Center of Educational Big Data, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
- National Engineering Research Center for E-Learning, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
| | - Ruijie Zhou
- National Engineering Research Center of Educational Big Data, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
- National Engineering Research Center for E-Learning, Central China Normal University, Luoyu Road, Wuhan, 430079 Hubei China
| |
Collapse
|
13
|
Yang W, Bian ZZ, Li Z, Zhang YT, Liu LB, Chang JT, Li D, Wang PG, An J, Wang W. An immunocompetent mouse model revealed that congenital Zika virus infection disrupted hippocampal function by activating autophagy. Emerg Microbes Infect 2025; 14:2465327. [PMID: 39945741 PMCID: PMC11873970 DOI: 10.1080/22221751.2025.2465327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 03/01/2025]
Abstract
Congenital Zika virus (ZIKV) infection significantly affects neurological development in infants and subsequently induces neurodevelopmental abnormality symptoms; however, the potential mechanism is still unknown. Therefore, in order to effectively intervene in neurodevelopmental abnormalities in infected infants, it is necessary to identify the main brain regions affected by congenital infection. In this study, we constructed a congenital ZIKV-infected murine model using immunocompetent human STAT2 knock-in mice, which presented long-term neurodevelopmental abnormalities with abnormal neurodevelopmental symptoms. We found that the hippocampus, which regulates cognitive behaviour and processes spatial information and navigation, was the main brain region affected by congenital infection and that hippocampal cells were more prone to autophagy during the growth period of these mice at the transcriptional and pathological levels. These findings highlighted that congenital ZIKV infection could interrupt hippocampal function by activating autophagy, thus providing a theoretical basis for the clinical treatment of congenital ZIKV-infected infants.
Collapse
Affiliation(s)
- Wei Yang
- National Center of Technology Innovation for animal model, National Human Diseases Animal Model Resource Center, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing, People’s Republic of China
| | - Zhan-Zhan Bian
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Zhe Li
- National Center of Technology Innovation for animal model, National Human Diseases Animal Model Resource Center, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing, People’s Republic of China
| | - Yi-Teng Zhang
- National Center of Technology Innovation for animal model, National Human Diseases Animal Model Resource Center, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing, People’s Republic of China
| | - Li-Bo Liu
- Department of Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Jia-Tong Chang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Dan Li
- National Center of Technology Innovation for animal model, National Human Diseases Animal Model Resource Center, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing, People’s Republic of China
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Wei Wang
- National Center of Technology Innovation for animal model, National Human Diseases Animal Model Resource Center, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing, People’s Republic of China
| |
Collapse
|
14
|
Ren X, Wang Y, Li X, Wang X, Liu Z, Yang J, Wang L, Zheng C. Attenuated heterogeneity of hippocampal neuron subsets in response to novelty induced by amyloid-β. Cogn Neurodyn 2025; 19:56. [PMID: 40161457 PMCID: PMC11947398 DOI: 10.1007/s11571-025-10237-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Alzheimer's disease (AD) patients exhibited episodic memory impairments including location-object recognition in a spatial environment, which was also presented in animal models with amyloid-β (Aβ) accumulation. A potential cellular mechanism was the unstable representation of spatial information and lack of discrimination ability of novel stimulus in the hippocampal place cells. However, how the firing characteristics of different hippocampal subsets responding to diverse spatial information were interrupted by Aβ accumulation remains unclear. In this study, we observed impaired novel object-location recognition in Aβ-treated Long-Evans rats, with larger receptive fields of place cells in hippocampal CA1, compared with those in the saline-treated group. We identified two subsets of place cells coding object information (ObjCell) and global environment (EnvCell) during the task, with firing heterogeneity in response to introduced novel information. ObjCells displayed a dynamic representation responding to the introduction of novel information, while EnvCells exhibited a stable representation to support the recognition of the familiar environment. However, the dynamic firing patterns of these two subsets of cells were disrupted to present attenuated heterogeneity under Aβ accumulation. The impaired spatial representation novelty information could be due to the disturbed gamma modulation of neural activities. Taken together, these findings provide new evidence for novelty recognition impairments of AD rats with spatial representation dysfunctions of hippocampal subsets.
Collapse
Affiliation(s)
- Xiaoxin Ren
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Yimeng Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xin Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xueling Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Zhaodi Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Jiajia Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Ling Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Chenguang Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Brain Science and Neuroengineering, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| |
Collapse
|
15
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [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: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
16
|
Wang Y, Li D, Xu K, Wang G, Zhang F. Copper homeostasis and neurodegenerative diseases. Neural Regen Res 2025; 20:3124-3143. [PMID: 39589160 PMCID: PMC11881714 DOI: 10.4103/nrr.nrr-d-24-00642] [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: 07/23/2024] [Revised: 07/27/2024] [Accepted: 10/14/2024] [Indexed: 11/27/2024] Open
Abstract
Copper, one of the most prolific transition metals in the body, is required for normal brain physiological activity and allows various functions to work normally through its range of concentrations. Copper homeostasis is meticulously maintained through a complex network of copper-dependent proteins, including copper transporters (CTR1 and CTR2), the two copper ion transporters the Cu -transporting ATPase 1 (ATP7A) and Cu-transporting beta (ATP7B), and the three copper chaperones ATOX1, CCS, and COX17. Disruptions in copper homeostasis can lead to either the deficiency or accumulation of copper in brain tissue. Emerging evidence suggests that abnormal copper metabolism or copper binding to various proteins, including ceruloplasmin and metallothionein, is involved in the pathogenesis of neurodegenerative disorders. However, the exact mechanisms underlying these processes are not known. Copper is a potent oxidant that increases reactive oxygen species production and promotes oxidative stress. Elevated reactive oxygen species levels may further compromise mitochondrial integrity and cause mitochondrial dysfunction. Reactive oxygen species serve as key signaling molecules in copper-induced neuroinflammation, with elevated levels activating several critical inflammatory pathways. Additionally, copper can bind aberrantly to several neuronal proteins, including alpha-synuclein, tau, superoxide dismutase 1, and huntingtin, thereby inducing neurotoxicity and ultimately cell death. This study focuses on the latest literature evaluating the role of copper in neurodegenerative diseases, with a particular focus on copper-containing metalloenzymes and copper-binding proteins in the regulation of copper homeostasis and their involvement in neurodegenerative disease pathogenesis. By synthesizing the current findings on the functions of copper in oxidative stress, neuroinflammation, mitochondrial dysfunction, and protein misfolding, we aim to elucidate the mechanisms by which copper contributes to a wide range of hereditary and neuronal disorders, such as Wilson's disease, Menkes' disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. Potential clinically significant therapeutic targets, including superoxide dismutase 1, D-penicillamine, and 5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline, along with their associated therapeutic agents, are further discussed. Ultimately, we collate evidence that copper homeostasis may function in the underlying etiology of several neurodegenerative diseases and offer novel insights into the potential prevention and treatment of these diseases based on copper homeostasis.
Collapse
Affiliation(s)
- Yuanyuan Wang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Daidi Li
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Kaifei Xu
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Guoqing Wang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Feng Zhang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
17
|
Song X, Li R, Chu X, Li Q, Li R, Li Q, Tong KY, Gu X, Ming D. Multilevel analysis of the central-peripheral-target organ pathway: contributing to recovery after peripheral nerve injury. Neural Regen Res 2025; 20:2807-2822. [PMID: 39435615 PMCID: PMC11826472 DOI: 10.4103/nrr.nrr-d-24-00641] [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: 06/11/2024] [Revised: 08/06/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Peripheral nerve injury is a common neurological condition that often leads to severe functional limitations and disabilities. Research on the pathogenesis of peripheral nerve injury has focused on pathological changes at individual injury sites, neglecting multilevel pathological analysis of the overall nervous system and target organs. This has led to restrictions on current therapeutic approaches. In this paper, we first summarize the potential mechanisms of peripheral nerve injury from a holistic perspective, covering the central nervous system, peripheral nervous system, and target organs. After peripheral nerve injury, the cortical plasticity of the brain is altered due to damage to and regeneration of peripheral nerves; changes such as neuronal apoptosis and axonal demyelination occur in the spinal cord. The nerve will undergo axonal regeneration, activation of Schwann cells, inflammatory response, and vascular system regeneration at the injury site. Corresponding damage to target organs can occur, including skeletal muscle atrophy and sensory receptor disruption. We then provide a brief review of the research advances in therapeutic approaches to peripheral nerve injury. The main current treatments are conducted passively and include physical factor rehabilitation, pharmacological treatments, cell-based therapies, and physical exercise. However, most treatments only partially address the problem and cannot complete the systematic recovery of the entire central nervous system-peripheral nervous system-target organ pathway. Therefore, we should further explore multilevel treatment options that produce effective, long-lasting results, perhaps requiring a combination of passive (traditional) and active (novel) treatment methods to stimulate rehabilitation at the central-peripheral-target organ levels to achieve better functional recovery.
Collapse
Affiliation(s)
- Xizi Song
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Ruixin Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Xiaolei Chu
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qi Li
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Ruihua Li
- Department of Hand Microsurgery, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qingwen Li
- School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Kai-Yu Tong
- Department of Biomedical Engineering, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiaosong Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Machine Interface and Human-Machine Fusion, Tianjin, China
| |
Collapse
|
18
|
Martinez B, Peplow PV. Autism spectrum disorder: difficulties in diagnosis and microRNA biomarkers. Neural Regen Res 2025; 20:2776-2786. [PMID: 39314171 PMCID: PMC11826456 DOI: 10.4103/nrr.nrr-d-24-00712] [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: 06/28/2024] [Revised: 08/17/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
We performed a PubMed search for microRNAs in autism spectrum disorder that could serve as diagnostic biomarkers in patients and selected 17 articles published from January 2008 to December 2023, of which 4 studies were performed with whole blood, 4 with blood plasma, 5 with blood serum, 1 with serum neural cell adhesion molecule L1-captured extracellular vesicles, 1 with blood cells, and 2 with peripheral blood mononuclear cells. Most of the studies involved children and the study cohorts were largely males. Many of the studies had performed microRNA sequencing or quantitative polymerase chain reaction assays to measure microRNA expression. Only five studies had used real-time polymerase chain reaction assay to validate microRNA expression in autism spectrum disorder subjects compared to controls. The microRNAs that were validated in these studies may be considered as potential candidate biomarkers for autism spectrum disorder and include miR-500a-5p, -197-5p, -424-5p, -664a-3p, -365a-3p, -619-5p, -664a-3p, -3135a, -328-3p, and -500a-5p in blood plasma and miR-151a-3p, -181b-5p, -320a, -328, -433, -489, -572, -663a, -101-3p, -106b-5p, -19b-3p, -195-5p, and -130a-3p in blood serum of children, and miR-15b-5p and -6126 in whole blood of adults. Several important limitations were identified in the studies reviewed, and need to be taken into account in future studies. Further studies are warranted with children and adults having different levels of autism spectrum disorder severity and consideration should be given to using animal models of autism spectrum disorder to investigate the effects of suppressing or overexpressing specific microRNAs as a novel therapy.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Pharmacology, University of Nevada-Reno, Reno, NV, USA
- Department of Medicine, University of Nevada-Reno, Reno, NV, USA
| | - Philip V. Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Li Y, Hao P, Duan H, Hao F, Zhao W, Gao Y, Yang Z, So KF, Li X. Activation of adult endogenous neurogenesis by a hyaluronic acid collagen gel containing basic fibroblast growth factor promotes remodeling and functional recovery of the injured cerebral cortex. Neural Regen Res 2025; 20:2923-2937. [PMID: 39610105 PMCID: PMC11826446 DOI: 10.4103/nrr.nrr-d-23-01706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 02/02/2024] [Accepted: 04/20/2024] [Indexed: 11/30/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00024/figure1/v/2024-11-26T163120Z/r/image-tiff The presence of endogenous neural stem/progenitor cells in the adult mammalian brain suggests that the central nervous system can be repaired and regenerated after injury. However, whether it is possible to stimulate neurogenesis and reconstruct cortical layers II to VI in non-neurogenic regions, such as the cortex, remains unknown. In this study, we implanted a hyaluronic acid collagen gel loaded with basic fibroblast growth factor into the motor cortex immediately following traumatic injury. Our findings reveal that this gel effectively stimulated the proliferation and migration of endogenous neural stem/progenitor cells, as well as their differentiation into mature and functionally integrated neurons. Importantly, these new neurons reconstructed the architecture of cortical layers II to VI, integrated into the existing neural circuitry, and ultimately led to improved brain function. These findings offer novel insight into potential clinical treatments for traumatic cerebral cortex injuries.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Hao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Kwok-Fai So
- Guangdong–HongKong–Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong Province, China
- Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administration Region, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong–HongKong–Macao Greater Bay Area, Guangzhou, Guangdong Province, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
20
|
Moreira P, Pocock R. Functions of nuclear factor Y in nervous system development, function and health. Neural Regen Res 2025; 20:2887-2894. [PMID: 39610092 PMCID: PMC11826454 DOI: 10.4103/nrr.nrr-d-24-00684] [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] [Revised: 09/04/2024] [Accepted: 09/27/2024] [Indexed: 11/30/2024] Open
Abstract
Nuclear factor Y is a ubiquitous heterotrimeric transcription factor complex conserved across eukaryotes that binds to CCAAT boxes, one of the most common motifs found in gene promoters and enhancers. Over the last 30 years, research has revealed that the nuclear factor Y complex controls many aspects of brain development, including differentiation, axon guidance, homeostasis, disease, and most recently regeneration. However, a complete understanding of transcriptional regulatory networks, including how the nuclear factor Y complex binds to specific CCAAT boxes to perform its function remains elusive. In this review, we explore the nuclear factor Y complex's role and mode of action during brain development, as well as how genomic technologies may expand understanding of this key regulator of gene expression.
Collapse
Affiliation(s)
- Pedro Moreira
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
21
|
Salasova A, Nykjær A. Emerging potential of progranulin-dependent SorCS2 signaling in healthy and diseased nervous systems. Neural Regen Res 2025; 20:2591-2593. [PMID: 39503427 PMCID: PMC11801297 DOI: 10.4103/nrr.nrr-d-24-00734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 02/08/2025] Open
Affiliation(s)
- Alena Salasova
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Centre of Excellence PROMEMO, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Nykjær
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Centre of Excellence PROMEMO, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
22
|
Qin Y, Zhu W, Guo T, Zhang Y, Xing T, Yin P, Li S, Li XJ, Yang S. Reduced mesencephalic astrocyte-derived neurotrophic factor expression by mutant androgen receptor contributes to neurodegeneration in a model of spinal and bulbar muscular atrophy pathology. Neural Regen Res 2025; 20:2655-2666. [PMID: 38934406 PMCID: PMC11801304 DOI: 10.4103/nrr.nrr-d-23-01666] [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: 10/07/2023] [Revised: 01/08/2024] [Accepted: 02/08/2024] [Indexed: 06/28/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00027/figure1/v/2024-11-05T132919Z/r/image-tiff Spinal and bulbar muscular atrophy is a neurodegenerative disease caused by extended CAG trinucleotide repeats in the androgen receptor gene, which encodes a ligand-dependent transcription factor. The mutant androgen receptor protein, characterized by polyglutamine expansion, is prone to misfolding and forms aggregates in both the nucleus and cytoplasm in the brain in spinal and bulbar muscular atrophy patients. These aggregates alter protein-protein interactions and compromise transcriptional activity. In this study, we reported that in both cultured N2a cells and mouse brain, mutant androgen receptor with polyglutamine expansion causes reduced expression of mesencephalic astrocyte-derived neurotrophic factor. Overexpression of mesencephalic astrocyte-derived neurotrophic factor ameliorated the neurotoxicity of mutant androgen receptor through the inhibition of mutant androgen receptor aggregation. Conversely, knocking down endogenous mesencephalic astrocyte-derived neurotrophic factor in the mouse brain exacerbated neuronal damage and mutant androgen receptor aggregation. Our findings suggest that inhibition of mesencephalic astrocyte-derived neurotrophic factor expression by mutant androgen receptor is a potential mechanism underlying neurodegeneration in spinal and bulbar muscular atrophy.
Collapse
Affiliation(s)
- Yiyang Qin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Wenzhen Zhu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Tingting Guo
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Yiran Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Tingting Xing
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Peng Yin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Su Yang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong–HongKong–Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
23
|
Su C, Huang T, Zhang M, Zhang Y, Zeng Y, Chen X. Glucocorticoid receptor signaling in the brain and its involvement in cognitive function. Neural Regen Res 2025; 20:2520-2537. [PMID: 39248182 PMCID: PMC11801288 DOI: 10.4103/nrr.nrr-d-24-00355] [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: 04/01/2024] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-adrenal axis regulates the secretion of glucocorticoids in response to environmental challenges. In the brain, a nuclear receptor transcription factor, the glucocorticoid receptor, is an important component of the hypothalamic-pituitary-adrenal axis's negative feedback loop and plays a key role in regulating cognitive equilibrium and neuroplasticity. The glucocorticoid receptor influences cognitive processes, including glutamate neurotransmission, calcium signaling, and the activation of brain-derived neurotrophic factor-mediated pathways, through a combination of genomic and non-genomic mechanisms. Protein interactions within the central nervous system can alter the expression and activity of the glucocorticoid receptor, thereby affecting the hypothalamic-pituitary-adrenal axis and stress-related cognitive functions. An appropriate level of glucocorticoid receptor expression can improve cognitive function, while excessive glucocorticoid receptors or long-term exposure to glucocorticoids may lead to cognitive impairment. Patients with cognitive impairment-associated diseases, such as Alzheimer's disease, aging, depression, Parkinson's disease, Huntington's disease, stroke, and addiction, often present with dysregulation of the hypothalamic-pituitary-adrenal axis and glucocorticoid receptor expression. This review provides a comprehensive overview of the functions of the glucocorticoid receptor in the hypothalamic-pituitary-adrenal axis and cognitive activities. It emphasizes that appropriate glucocorticoid receptor signaling facilitates learning and memory, while its dysregulation can lead to cognitive impairment. This provides clues about how glucocorticoid receptor signaling can be targeted to overcome cognitive disability-related disorders.
Collapse
Affiliation(s)
- Chonglin Su
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Taiqi Huang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Meiyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
24
|
Hayat M, Syed RA, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Albassam LAM, Kaleem I, Wang X, Wang R, Bhatti MS, Bashir S. Decoding molecular mechanisms: brain aging and Alzheimer's disease. Neural Regen Res 2025; 20:2279-2299. [PMID: 39104174 PMCID: PMC11759015 DOI: 10.4103/nrr.nrr-d-23-01403] [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: 08/20/2023] [Revised: 01/23/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
The complex morphological, anatomical, physiological, and chemical mechanisms within the aging brain have been the hot topic of research for centuries. The aging process alters the brain structure that affects functions and cognitions, but the worsening of such processes contributes to the pathogenesis of neurodegenerative disorders, such as Alzheimer's disease. Beyond these observable, mild morphological shifts, significant functional modifications in neurotransmission and neuronal activity critically influence the aging brain. Understanding these changes is important for maintaining cognitive health, especially given the increasing prevalence of age-related conditions that affect cognition. This review aims to explore the age-induced changes in brain plasticity and molecular processes, differentiating normal aging from the pathogenesis of Alzheimer's disease, thereby providing insights into predicting the risk of dementia, particularly Alzheimer's disease.
Collapse
Affiliation(s)
- Mahnoor Hayat
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rafay Ali Syed
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad (IIUI), Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Bioengineering, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | | | - Imdad Kaleem
- Department of Biosciences, Commission on Science and Technology for Sustainable Development in the South (COMSATS University), Islamabad, Pakistan
| | - Xueyi Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ran Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mehwish S. Bhatti
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
25
|
You X, Niu L, Fu J, Ge S, Shi J, Zhang Y, Zhuang P. Bidirectional regulation of the brain-gut-microbiota axis following traumatic brain injury. Neural Regen Res 2025; 20:2153-2168. [PMID: 39359076 PMCID: PMC11759007 DOI: 10.4103/nrr.nrr-d-24-00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/20/2024] [Accepted: 05/11/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00002/figure1/v/2024-09-30T120553Z/r/image-tiff Traumatic brain injury is a prevalent disorder of the central nervous system. In addition to primary brain parenchymal damage, the enduring biological consequences of traumatic brain injury pose long-term risks for patients with traumatic brain injury; however, the underlying pathogenesis remains unclear, and effective intervention methods are lacking. Intestinal dysfunction is a significant consequence of traumatic brain injury. Being the most densely innervated peripheral tissue in the body, the gut possesses multiple pathways for the establishment of a bidirectional "brain-gut axis" with the central nervous system. The gut harbors a vast microbial community, and alterations of the gut niche contribute to the progression of traumatic brain injury and its unfavorable prognosis through neuronal, hormonal, and immune pathways. A comprehensive understanding of microbiota-mediated peripheral neuroimmunomodulation mechanisms is needed to enhance treatment strategies for traumatic brain injury and its associated complications. We comprehensively reviewed alterations in the gut microecological environment following traumatic brain injury, with a specific focus on the complex biological processes of peripheral nerves, immunity, and microbes triggered by traumatic brain injury, encompassing autonomic dysfunction, neuroendocrine disturbances, peripheral immunosuppression, increased intestinal barrier permeability, compromised responses of sensory nerves to microorganisms, and potential effector nuclei in the central nervous system influenced by gut microbiota. Additionally, we reviewed the mechanisms underlying secondary biological injury and the dynamic pathological responses that occur following injury to enhance our current understanding of how peripheral pathways impact the outcome of patients with traumatic brain injury. This review aimed to propose a conceptual model for future risk assessment of central nervous system-related diseases while elucidating novel insights into the bidirectional effects of the "brain-gut-microbiota axis."
Collapse
Affiliation(s)
- Xinyu You
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Niu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiafeng Fu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shining Ge
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangwei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanjun Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Pengwei Zhuang
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
26
|
Fabregat-Safont D, Coadic L, Haro N, Montané-Garcia M, Canaguier J, Mallaret G, Davidovic L, Pozo ÓJ. Improving the detectability of low-abundance p-cresol in biological matrices by chemical derivatization and LC-MS/MS determination. Talanta 2025; 290:127770. [PMID: 40010114 DOI: 10.1016/j.talanta.2025.127770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Gut microbiota produces a wide range of microbial metabolites with potential neuroactive properties. Among these, p-cresol, a by-product of tyrosine breakdown, has gained significant attention in various neuropsychiatric disorders, including autism spectrum disorder. However, current methods fail to detect p-cresol at trace levels in both the systemic circulation and brain, limiting the study of its role in neuropsychiatric disorders. There, the precise and accurate determination of p-cresol at low picogram levels is an unmet analytical need. To address this gap, we developed a highly-sensitive, validated method for quantifying p-cresol at low picogram levels in urine, plasma, and brain using chemical derivatization and liquid chromatography-tandem mass spectrometry (LC-MS/MS). We found that derivatization with 1,2-dimethylimidazole-5-sulfonyl chloride (5-DMIS-Cl or 5-DMISC) increased up to 40-fold the sensitivity compared to traditional dansyl derivatization. Therefore, a method based on 5-DMISC derivatization and sum of transitions was selected for validation. The method was accurate (recoveries 91-100 %) and precise (RSD <15 %) in all tested matrices, enabling detection down to100 pg/mL for urine, 20 pg/mL for plasma, and 0.04 pg/mg for brain tissue. The method was applied to plasma and brain samples from control and p-cresol-treated mice, revealing significant increases in p-cresol levels in treated animals. For the first time, we successfully quantified p-cresol levels in the brain, demonstrating its ability to cross the blood-brain barrier. In summary, this validated method offers a powerful tool for exploring the role of p-cresol -and potentially other phenolic compounds-in the microbiota-gut-brain axis and neuropsychiatric disorders.
Collapse
Affiliation(s)
- David Fabregat-Safont
- Applied Metabolomics Research Group, Hospital del Mar Research Institute, Barcelona, Spain; Environmental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water (IUPA), Univ. Jaume I, Castelló, Spain
| | - Lena Coadic
- Applied Metabolomics Research Group, Hospital del Mar Research Institute, Barcelona, Spain
| | - Noemí Haro
- Applied Metabolomics Research Group, Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Juliette Canaguier
- Université Côte d'Azur, CNRS UMR7275, INSERM U1318, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Geoffroy Mallaret
- Université Côte d'Azur, CNRS UMR7275, INSERM U1318, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Laetitia Davidovic
- Université Côte d'Azur, CNRS UMR7275, INSERM U1318, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Óscar J Pozo
- Applied Metabolomics Research Group, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
27
|
Ganesan S, Misaki M, Zalesky A, Tsuchiyagaito A. Functional brain network dynamics of brooding in depression: Insights from real-time fMRI neurofeedback. J Affect Disord 2025; 380:191-202. [PMID: 40122254 DOI: 10.1016/j.jad.2025.03.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Brooding is a critical symptom and prognostic factor of major depressive disorder (MDD), which involves passively dwelling on self-referential dysphoria and related abstractions. The neurobiology of brooding remains under characterized. We aimed to elucidate neural dynamics underlying brooding, and explore their responses to neurofeedback intervention in MDD. METHODS We investigated functional MRI (fMRI) dynamic functional network connectivity (dFNC) in 36 MDD subjects and 26 healthy controls (HCs) during rest and brooding. Rest was measured before and after fMRI neurofeedback (MDD-active/sham: n = 18/18, HC-active/sham: n = 13/13). Baseline brooding severity was recorded using Ruminative Response Scale - Brooding subscale (RRS-B). RESULTS Four recurrent dFNC states were identified. Measures of time spent were not significantly different between MDD and HC for any of these states during brooding or rest. RRS-B scores in MDD showed significant negative correlation with measures of time spent in dFNC state 3 during brooding (r = -0.4, p = 0.002, FDR-significant). This state comprises strong connections spanning several brain systems involved in sensory, attentional and cognitive processing. Time spent in this anti-brooding dFNC state significantly increased following neurofeedback only in the MDD active group (z = -2.09, FWE-p = 0.034). LIMITATIONS The sample size was small and imbalanced between groups. Brooding condition was not examined post-neurofeedback. CONCLUSION We identified a densely connected anti-brooding dFNC brain state in MDD. MDD subjects spent significantly longer time in this state after active neurofeedback intervention, highlighting neurofeedback's potential for modulating dysfunctional brain dynamics to treat MDD.
Collapse
Affiliation(s)
- Saampras Ganesan
- Department of Psychiatry, Melbourne Medical School, Carlton, Victoria 3053, Australia; Department of Biomedical Engineering, The University of Melbourne, Carlton, Victoria 3053, Australia; Contemplative Studies Centre, Melbourne School of Psychological Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia.
| | - Masaya Misaki
- Laureate Institute for Brain Research, Tulsa, OK, USA; Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, USA
| | - Andrew Zalesky
- Department of Psychiatry, Melbourne Medical School, Carlton, Victoria 3053, Australia; Department of Biomedical Engineering, The University of Melbourne, Carlton, Victoria 3053, Australia
| | - Aki Tsuchiyagaito
- Laureate Institute for Brain Research, Tulsa, OK, USA; Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, USA; Research Center for Child Mental Development, Chiba University, Chiba, Japan
| |
Collapse
|
28
|
Frigon A, Lecomte CG. Stepping up after spinal cord injury: negotiating an obstacle during walking. Neural Regen Res 2025; 20:1919-1929. [PMID: 39254549 PMCID: PMC11691478 DOI: 10.4103/nrr.nrr-d-24-00369] [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: 04/04/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 09/11/2024] Open
Abstract
Every day walking consists of frequent voluntary modifications in the gait pattern to negotiate obstacles. After spinal cord injury, stepping over an obstacle becomes challenging. Stepping over an obstacle requires sensorimotor transformations in several structures of the brain, including the parietal cortex, premotor cortex, and motor cortex. Sensory information and planning are transformed into motor commands, which are sent from the motor cortex to spinal neuronal circuits to alter limb trajectory, coordinate the limbs, and maintain balance. After spinal cord injury, bidirectional communication between the brain and spinal cord is disrupted and animals, including humans, fail to voluntarily modify limb trajectory to step over an obstacle. Therefore, in this review, we discuss the neuromechanical control of stepping over an obstacle, why it fails after spinal cord injury, and how it recovers to a certain extent.
Collapse
Affiliation(s)
- Alain Frigon
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Charly G. Lecomte
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
29
|
Ignat LA, Tipa RO, Cehan AC, Bacârea VC. BK channels and alcohol tolerance: Insights from studies on Drosophila, nematodes, rodents and cell lines: A systematic review. MEDICINE INTERNATIONAL 2025; 5:33. [PMID: 40236633 PMCID: PMC11995379 DOI: 10.3892/mi.2025.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
Addictive disorders markedly affect the emotional, physical and financial wellbeing of individuals, placing a substantial burden on the healthcare system. With their widespread presence in the brain, large-conductance calcium and voltage-activated potassium (BK) channels play a crucial role in various aspects of neuronal function. They contribute to behavioral tolerance and are closely linked to neuronal activity and modulation through intracellular calcium levels. As such, BK channels serve as key models for investigating the mechanisms of the effects of alcohol. Investigating their role in alcohol tolerance provides a broader understanding of their physiological and pharmacological importance. The present systematic review examined the literature on the role of BK channels in alcohol tolerance and comprehensively explored the topic. For this purpose, two databases, Web of Science and PubMed, were searched, and studies published from 2000 until June, 2024 were included. After applying specific inclusion and exclusion criteria, 35 studies underwent analysis to present a chronological overview of BK channels and their relevance in alcohol tolerance development. The studies were categorized into four main groups, according to research conducted on: i) Fruit flies; ii) nematodes; iii) rodents; and iv) cell lines. Understanding the mechanisms through which alcohol interacts with these channels may help to elucidate the cellular and molecular mechanisms underlying alcohol tolerance. There is a growing interest in developing drugs that can precisely modulate BK channel activity to treat alcohol dependence and tolerance. However, additional studies are required to fully explain the complex mechanisms through which BK channels influence alcohol-related behaviors and to interpret these findings into clinical applications.
Collapse
Affiliation(s)
- Luciana Angela Ignat
- Doctoral School, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
- ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
| | - Raluca Oana Tipa
- ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
- Department of Psychiatry, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alina Cehan Cehan
- Plastic and Reconstructive Surgery, Emergency Clinical County Hospital of Targu Mures, 540136 Targu Mures, Romania
| | - Vladimir Constantin Bacârea
- Department of Scientific Research Methodology, ‘George Emil Palade’ University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| |
Collapse
|
30
|
Xue X, Gu X, Zhang Y, Wu X, Xia T, Lu R, Wang H, Hua Y. Corticospinal tract alterations after ankle sprain in adolescence: Insights from the mouse model. SPORTS MEDICINE AND HEALTH SCIENCE 2025; 7:292-298. [PMID: 40264833 PMCID: PMC12010401 DOI: 10.1016/j.smhs.2024.06.006] [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: 08/31/2023] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 04/24/2025] Open
Abstract
Purpose Lateral ankle sprains (LAS) are associated with corticospinal pathway deficits. Existing evidence is primarily based on cross-sectional investigations and noncausal speculations. This study aims to determine whether maladaptive corticospinal pathway alterations occur pre- and postligament transection in LAS mouse models. Additionally, this study explores whether the alterations are more pronounced in adolescent mice than adults. Methods Twenty-four 8-week-old adolescent and twenty-four 24-week-old adult mice were randomly assigned to lateral ankle ligament transection or sham surgery. Diffusion-weighted imaging of the corticospinal pathway was performed presurgery and 8 weeks postsurgery. Fractional anisotropy (FA) values, reflecting fiber integrity within the corticospinal subregions of the medulla, pons, midbrain, and cerebrum, were extracted. Results Overall, 41 mice completed repeated image acquisition. Before surgery, no significant group effects on FA within the four corticospinal subregions were detected in either adolescent or adult mice. Two months after surgery, the adolescent cohort displayed a significant reduction in FA in the medulla subregion following ankle ligament transection (β-baseline-adjusted = -0.083, 95% CI , -0.145 to -0.021, p-corrected = 0.048). Conversely, no significant effects of ankle ligament transection on corticospinal FA were observed in the adult cohort. Conclusion The maladaptive alterations in the corticospinal tract could be observed in the adolescent LAS mouse model, characterized by reduced fiber integrity in the medulla subregion. While these results are derived from an animal model, they provide a foundation for future investigations into the mechanisms underlying neurological deficits following musculoskeletal injuries.
Collapse
Affiliation(s)
- Xiao'ao Xue
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xicheng Gu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Xuejun Wu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Tian Xia
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Rong Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Yinghui Hua
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Chen X, Peng Y, Liu XS. DNA Methylation in Long-Term Memory. Physiology (Bethesda) 2025; 40:0. [PMID: 39907057 DOI: 10.1152/physiol.00032.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Understanding the neural mechanisms of memory has been one of the key questions in biology. Long-term memory, specifically, allows one to travel mentally without constraints of time and space. A long-term memory must have gone through a series of temporal processes: encoding, consolidation, storage, and retrieval. Decades of studies have revealed cellular and molecular mechanisms underlying each process. In this article, we first review the emerging concept of memory engrams and technologies of engram labeling, as these methods provide a new avenue to study the molecular mechanisms for memory. Then, we focus on DNA methylation and its role in long-term memory. Finally, we discuss some key remaining questions in this field and their implications in memory-related disease.
Collapse
Affiliation(s)
- Xinyue Chen
- Department of Neuroscience, Columbia University, New York, New York, United States
- Department of Pathology and Cell Biology, Columbia University Medical Center, Columbia University, New York, New York, United States
- Department of Neurology, Columbia University Medical Center, Columbia University, New York, New York, United States
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, Columbia University, New York, New York, United States
| | - Yueqing Peng
- Department of Pathology and Cell Biology, Columbia University Medical Center, Columbia University, New York, New York, United States
- Department of Neurology, Columbia University Medical Center, Columbia University, New York, New York, United States
| | - X Shawn Liu
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, Columbia University, New York, New York, United States
| |
Collapse
|
32
|
Kujur PP, Ellappan S, Mondal AC. Neuronal and therapeutic perspectives on empathic pain: A rational insight. Neuropharmacology 2025; 272:110414. [PMID: 40081793 DOI: 10.1016/j.neuropharm.2025.110414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/08/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Empathy is the capacity to experience and understand the feelings of others, thereby playing a key role in a person's mental well-being essentially by promoting kindness and a sense of belongingness to the group. However, too much empathy may result in psychological problems such as empathic distress, compassion fatigue, and burnout, collectively termed empathic pain. Several brain regions are implicated in processing empathic pain perception. Neuroimaging investigations bring in the context of brain structures involved in this emotional exchange, pointing toward the anterior insula (AI) and anterior cingulate cortex (ACC), indicating an overlap between the neural representation of direct and simulative pain. To discern such overlaps, therapeutic techniques for managing empathic pain require understanding different brain regions and their respective neural networks. At the moment, empathic pain is being treated using various methods, including pharmacological treatments such as antidepressants and psychological treatments such as mindfulness or meditation. For instance, researchers have been exploring the modulatory effects of neurotransmitters like serotonin, norepinephrine, and oxytocin on individuals' responses to empathic pain experience. Importantly, this review focuses on the specific brain parts and their unique roles in neurobiological pathways associated with emphatic pain and how shared neural networks play into available treatment options, suggesting possible future health benefits. Such an understanding of empathy can lead to more efficient management of types of care, focusing on enhancing social connections and mental well-being.
Collapse
Affiliation(s)
- Punit Prasanna Kujur
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences Jawaharlal Nehru University, New Delhi, 110067, India
| | - Surendar Ellappan
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
33
|
Bu S, Wang Q, Zhang G, Zhang Z, Dai J, Zhang Z. Inflammation molecular network alterations in a depressive-like primate model. J Affect Disord 2025; 379:410-420. [PMID: 40081592 DOI: 10.1016/j.jad.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
At present, there are no definitive biomarkers for major depressive disorder (MDD). Previous studies prompted that neuroimmunoinflammation is involved in the pathogenesis of depression and its factors become potential diagnostic biomarkers. Non-human primates exhibit depression-like behavior similar to humans in chronically stressed environments. Therefore, in the present study, after completing Whole transcriptome sequencing of peripheral blood, neurology-related and inflammatory molecules in plasma and cerebrospinal fluid were measured by Olink proximity extension assay technology simultaneously in 4 natural depressive-like (DL) cynomolgus monkeys and 4 normal controls to screen potential biological markers. Further, postmortem brain tissues and peripheral blood RNA sequencing data from MDD patients available in the Gene Expression Omnibus (GEO) database were used for cross-species validation. Compared to control monkeys, depressive-like monkeys exhibited elevated levels of neurocan (NCAN). RNA sequencing revealed Toll-like receptor 4 (TLR4) and the interacting S100 calcium-binding protein A family as key molecules in the inflammatory gene network. GEO brain tissue data showed up-regulation of S100A8 and S100A9 in the anterior cingulate cortex of MDD patients. These findings suggest that depressive-like monkeys are in a state of chronic low-grade inflammation and identify NCAN and TLR4 inflammatory network molecules as potential biomarkers of MDD.
Collapse
Affiliation(s)
- Siyuan Bu
- Department of Neurology in Affiliated ZhongDa Hospital and Jiangsu Provincial Medical Key Discipline, School of Medicine, Institution of Neuropsychiatry, Key Laboratory of Developmental Genes and Human Disease of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China
| | - Qingyun Wang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, 999077, Hong Kong
| | - Gaojia Zhang
- Department of Neurology in Affiliated ZhongDa Hospital and Jiangsu Provincial Medical Key Discipline, School of Medicine, Institution of Neuropsychiatry, Key Laboratory of Developmental Genes and Human Disease of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China; Department of Psychology and Sleep Medicine, the Second Hospital of Anhui Medical University, Hefei 230000, China
| | - Zhiting Zhang
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ji Dai
- Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zhijun Zhang
- Department of Neurology in Affiliated ZhongDa Hospital and Jiangsu Provincial Medical Key Discipline, School of Medicine, Institution of Neuropsychiatry, Key Laboratory of Developmental Genes and Human Disease of Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, China; Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences of Shenzhen University of Advanced Technology, The Brain Cognition and Brain Disease Institute of Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
34
|
Cao X, Zhang Y, Wu H, Da H, Xiao Q, Shi H. Decoding depression: How DLPFC and SMA mediate stress perception's role in mental health? J Affect Disord 2025; 379:323-331. [PMID: 40086480 DOI: 10.1016/j.jad.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Depression is a common mental disorder that significantly impacts global well-being. Although stress is a major contributor to depression, not all stress leads to depressive outcomes due to differences in stress perception. Understanding the neural mechanisms of stress perception may help identify biomarkers for targeted interventions to alleviate stress-related depression. METHODS This study included 113 participants. Each participant completed a Verbal Fluency Task (VFT) while undergoing functional near-infrared spectroscopy (fNIRS) to monitor brain activity. Oxyhemoglobin (Oxy-Hb) concentration data were analyzed using Matlab, and PROCESS v4.1 to examine neural mechanisms connecting stress perception and depression. RESULTS Correlation analysis showed a significant negative association between depression severity and Oxy-Hb concentration in several brain regions, including the bilateral dorsolateral prefrontal cortex (DLPFC), bilateral Broca's area (BA), right frontal pole (FP), and right orbitofrontal cortex (OFC). Mediation and moderation analyses revealed that the bilateral DLPFC serves as a key mediator in the relationship between stress perception and depression, with the supplementary motor area (SMA) acts as a moderator. Functional differentiation was observed, with the left DLPFC and left SMA influencing the effect of nervous on depression, and the right DLPFC and right SMA influencing the effect of uncontrolled on depression. CONCLUSION The bilateral DLPFC and SMA play critical roles in mediating and moderating stress perception's impact on depression, suggesting these regions as potential targets for interventions in stress-related depressive disorders.
Collapse
Affiliation(s)
- Xiaochen Cao
- School of Education, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yan Zhang
- School of Education, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Huifen Wu
- School of Education, Hubei Engineering University, Xiaogan 432000, China
| | - Hui Da
- School of Education, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qiang Xiao
- Department of Neurology, Hospital of Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hui Shi
- The Department of Cardio-Psychiatry Liaison Consultation, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
35
|
Oliveira A, Azevedo M, Seixas R, Silva S, Martinho R, Serrão P, Silva E, Moreira-Rodrigues M. Hippocampus muscarinic M4 receptor mRNA expression may influence central cholinergic activity, causing fear memory strengthening by peripheral adrenaline. Neuropharmacology 2025; 271:110382. [PMID: 39988278 DOI: 10.1016/j.neuropharm.2025.110382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Adrenaline (Ad) strengthens contextual fear memory by increasing blood glucose, possibly enhancing hippocampus acetylcholine synthesis. Nevertheless, it is unclear if peripheral Ad influences the cholinergic system, contributing to contextual fear memory strengthening. We aimed to evaluate whether peripheral Ad alters muscarinic receptor expression and if the cholinergic system is involved in peripheral Ad contextual fear memory strengthening effect. Wild-type (WT) and Ad-deficient male mice (129 × 1/SvJ) underwent a fear conditioning procedure followed by intraperitoneal pre-training and pre-context administration of Ad (0.1 mg/kg), atropine (10 mg/kg), methylatropine (0.5 mg/kg), Ad (0.1 mg/kg) plus atropine (10 mg/kg) or vehicle (NaCl, 0.9%). Shock responsiveness and freezing behaviour were accessed. Hippocampal M1, M2, and M4 mRNA expression were evaluated. Ad-deficient mice presented decreased hippocampal muscarinic M4 subtype receptor mRNA expression compared to WT mice. In Ad-administered Ad-deficient mice, hippocampal muscarinic M4 subtype receptor mRNA expression increased compared with vehicle-administered Ad-deficient mice. On the context day, atropine-administered WT mice presented decreased freezing behaviour compared to vehicle or methylatropine-administered WT mice. Moreover, Ad plus atropine-administered Ad-deficient mice led to decreased freezing behaviour compared to Ad-administered Ad-deficient mice. In conclusion, Ad-deficient mice's contextual fear memory impairment was associated with hippocampal muscarinic M4 subtype receptor down expression, which was reversed by Ad. This may be related to contextual fear memory consolidation or retrieval induced by peripheral Ad. Furthermore, the effect of Ad contextual fear memory might be due to increased hippocampus muscarinic subtype M4 expression, which may contribute to increased cholinergic activity in the central nervous system.
Collapse
Affiliation(s)
- Ana Oliveira
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Márcia Azevedo
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Rafaela Seixas
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Soraia Silva
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Raquel Martinho
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Paula Serrão
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
| | - Elisabete Silva
- Ageing and Stress Group, i3S- Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Faculty of Veterinary Medicine, Lusófona University, Lisbon, Portugal
| | - Mónica Moreira-Rodrigues
- Center for Drug Discovery and Innovative Medicines (MedInUP) and RISE-Health, Department of Immuno-physiology and Pharmacology, Laboratory of Physiology, School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal.
| |
Collapse
|
36
|
Pirri F, McCormick CM. Oxytocin receptors within the caudal lateral septum regulate social approach-avoidance, long-term social discrimination, and anxiety-like behaviors in adult male and female rats. Neuropharmacology 2025; 271:110409. [PMID: 40074168 DOI: 10.1016/j.neuropharm.2025.110409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
OTR signaling promotes social approach or facilitates social avoidance, depending on the brain region involved. The lateral septum plays a critical role in regulating social interactions and memory. We investigated the role of OTR signaling in the caudodorsal lateral septum (LSc.d) in modulating social approach-avoidance behavior, long-term social discrimination memory, and anxiety-like behaviors in adult rats. Local infusion of the selective OTR antagonist L-368,899 (1 μg/0.5 μl) into the LSc.d decreased social approach, increased social vigilance, and reduced long-term social discrimination memory in both sexes. Administration of the biased OTR/Gq agonist carbetocin (0.5 μg/0.5 μl) reduced social approach and long-term social discrimination memory in both sexes, and had anxiogenic effects (increased latency to consume palatable food in test arena) only in males. In contrast, the full OTR agonist TGOT (50 ng/0.5 μl) had no effect on social approach or long-term social discrimination memory, and decreased latency to consume palatable food (anxiolytic effect). The results indicate that the oxytocin system can both promote and inhibit social behaviors depending on the differential activation of G-protein subunits and β-arrestins, as well as the pivotal role of the LS in modulating social and anxiety-like behavior in rats.
Collapse
Affiliation(s)
- Fardad Pirri
- Biological Sciences Department, Brock University, Canada
| | - Cheryl M McCormick
- Biological Sciences Department, Brock University, Canada; Psychology Department, Brock University, Canada.
| |
Collapse
|
37
|
Sanie-Jahromi F, Sadeghi N, Moayedfard Z, Gharegezloo Z, Nejabat M, Nowroozzadeh MH. Effects of exosomes derived from activated corneal stromal keratocytes on the inflammation, proliferation, neuroprotection and epithelial-mesenchymal transition in retinal pigment epithelium cells. Life Sci 2025; 371:123592. [PMID: 40174671 DOI: 10.1016/j.lfs.2025.123592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
AIMS This study investigated the effects of activated keratocyte-derived exosomes (aKExo) on retinal pigment epithelial (RPE) cells in-vitro, focusing on cell viability, inflammatory cytokine expression, and neuroprotective properties. MATERIALS AND METHODS Keratocytes were cultured, and exosomes were extracted and characterized using transmission electron microscopy (TEM), scanning electron microscopy (SEM), flow cytometry, and dynamic light scattering (DLS). RPE cells, isolated from a human donor, were confirmed via RPE65 expression. aKExo effects on RPE cells were assessed using MTT assay at concentrations from 10-1 (35 μg/mL) to 10-5 (3.5 × 10-3 μg/mL). The optimal aKExo concentration (10-5) enhanced cell viability and exhibited the highest proliferative potential compared to the control group, making it the optimal dose for subsequent experiments including gene expression analysis, and ELISA. KEY FINDINGS aKExo downregulated IL-6 mRNA (0.70 ± 0.06, p = 0.0009) and marginally reduced TGF-β mRNA (0.75 ± 0.16, p = 0.0575). ELISA confirmed a reduction in IL-6 (31.33 ± 5.77 pg/mL vs. 50.22 ± 13.47 pg/mL, p = 0.0894) and TGF-β (8.91 ± 0.16 pg/mL vs. 11.39 ± 1.49 pg/mL, p = 0.0460). No significant changes were observed for IL-1β expression or other epithelial-mesenchymal transition (EMT)-related genes (α-SMA, ZEB-1, β-catenin). Neuroprotective genes NGF (4.34 ± 1.05, p = 0.0053) and CD90 (1.55 ± 0.25, p = 0.0184) were significantly upregulated, while VEGF-A was elevated (1.65 ± 0.15, p = 0.0018). SIGNIFICANCE These findings highlight aKExo's immunomodulatory, neuroprotective, and anti-EMT effects, suggesting potential therapeutic applications for retinal disorders, while noting that VEGF-A upregulation requires further investigation.
Collapse
Affiliation(s)
- Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Niloofar Sadeghi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Moayedfard
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Gharegezloo
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Nejabat
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - M Hossein Nowroozzadeh
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
38
|
Estupiñán HY, Baladi T, Roudi S, Munson MJ, Bost J, Gustafsson O, Velásquez-Ramírez D, Bhatt DK, Hagey D, Hekman D, Andersson S, Andaloussi SEL, Dahlén A. Design and screening of novel endosomal escape compounds that enhance functional delivery of oligonucleotides in vitro. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102522. [PMID: 40235852 PMCID: PMC11999280 DOI: 10.1016/j.omtn.2025.102522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/16/2025] [Indexed: 04/17/2025]
Abstract
Antisense oligonucleotides (ASOs), including splice-switching oligonucleotides (SSOs), are promising therapeutic approaches for targeting genetic defects. ASOs act in the nucleus and the cytosol to cleave mRNAs via the RNaseH1 mechanism (e.g., gapmers), while SSOs alter transcript splicing to restore or inhibit protein function. RNA interference (RNAi) is another approach to down-regulate gene expression via the RISC complex. However, a major challenge is the effective delivery of these nucleic acid-based therapeutics. Recent developments focus on enhancing cellular uptake and endosomal release, including the use of small-molecule endosomal escape enhancers (EEEs) such as chloroquine. Here, we disclose a next generation of EEEs, which efficiently enhance SSOs and gapmers in vitro activity. We identify proton sponge-mediated endosomal leakage as a mechanism of action and observe, by Gene Ontology analysis on bulk RNA sequencing, that EEE treatment increased gene expression of markers associated with vesicle organization. Additionally, using primary human hepatocytes, we demonstrate that EEEs enhance small interfering RNA (siRNA) activity. Unconjugated siRNA reached similar levels of mRNA knockdown to the observed GalNAc-conjugated siRNA. Substantial GalNAc conjugated siRNA enhancement was also observed when used together with EEE. Our results indicate that these EEEs constitute a promising strategy to enhance the activity of multimodal oligonucleotide therapeutics.
Collapse
Affiliation(s)
- H. Yesid Estupiñán
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Departamento de Ciencias Básicas, Universidad Industrial de Santander, Bucaramanga, Colombia
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Tom Baladi
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Samantha Roudi
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Michael J. Munson
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Jeremy Bost
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Oskar Gustafsson
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Daniel Velásquez-Ramírez
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Deepak Kumar Bhatt
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Hagey
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Dennis Hekman
- DMPK, Research and Early Development Cardiovascular Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Shalini Andersson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Huddinge, Sweden
- Karolinska ATMP Center, ANA Futura, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Huddinge, Sweden
| | - Anders Dahlén
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
39
|
Tang Z, Li R, Guo X, Wang Z, Wu J. Regulation of blood-brain barrier integrity by brain microvascular endothelial cells in ischemic stroke: A therapeutic opportunity. Eur J Pharmacol 2025; 996:177553. [PMID: 40147580 DOI: 10.1016/j.ejphar.2025.177553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Stroke is the second leading cause of death from cardiovascular diseases. Brain microvascular endothelial cells (BMECs) are crucial in the treatment of cerebral ischemic stroke, as their functional status directly affects the integrity of the blood-brain barrier (BBB). This review systematically discusses the central role of BMECs in ischemia. The mitochondrial dysfunction and activation of apoptosis/necrosis pathways in BMECs directly disrupt the integrity of the BBB and the degradation of junctional complexes (such as TJs and AJs) further exacerbates its permeability. In the neurovascular unit (NVU), astrocytes, microglia, and pericytes regulate the function of BMECs by secreting cytokines (such as TGF-β and VEGF), showing dual effects of promoting repair and damage. The dynamic changes of transporters, including those from the ATP-binding cassette and solute carrier families, as well as ion channels and exchangers, such as potassium and calcium channels, offer novel insights for the development of targeted drug delivery systems.
Collapse
Affiliation(s)
- Ziqi Tang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| | - Ruoxi Li
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| | - Xi Guo
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China.
| |
Collapse
|
40
|
He Q, Yang Q, Wu L, He Y, Zeng N, Wang Z. Neurotoxic effects of per- and polyfluoroalkyl substances (PFAS) mixture exposure in mice: Accumulations in brain and associated changes of behaviors, metabolome, and transcriptome. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137699. [PMID: 39987740 DOI: 10.1016/j.jhazmat.2025.137699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/22/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Humans are exposed to complex per- and polyfluoroalkyl substances (PFAS) mixtures, yet their neurotoxicity and mechanisms remains unclear. This study exposed male mice to 17 PFAS mixtures at low levels (0.2-20 µg/L) for 49 days via drinking water. Perfluoropentanoic acid (PFPeA), perfluoroheptanoic acid (PFHpA), 6:2 fluorotelomer sulfonic acid (6:2 FTS), and perfluorooctane Sulfonate (PFOS) accumulated in brain tissues, with brain/plasma ratios of 2.03-5.87, 2.94-12.88, 1.90-3.19, and 0.62-0.93, respectively. Electroencephalogram (EEG) results showed significant alterations, including a reduction in beta spectral edge (21.47-13.85 Hz) and an increase in gamma spectral edge (57.64-79.07 Hz). Histopathological analysis revealed necrosis in the hippocampus, contributing to the observed anxiety-like behaviors and memory impairments in exposed mice. Plasma metabolomics highlighted disrupted osmoprotectants, impaired glutamatergic synapse function, and tryptophan metabolism. Brain metabolomics demonstrated suppression of purine metabolism and activation of arachidonic acid metabolism, suggesting involvement in neurotoxic effects. Transcriptomic profiling further identified dysregulation in neuroactive ligand-receptor interactions, cholinergic and GABAergic synapses, and calcium signaling pathways, with oxytocin signaling highlighted as a critical mechanism. This study, for the first time, links PFAS mixture to neurotoxicity via neurotransmitter-related pathways, underscoring the need for public health policies and preventive strategies to mitigate PFAS exposure risks.
Collapse
Affiliation(s)
- Qiurong He
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qingkun Yang
- West China School of Public Health, Sichuan University, Chengdu 610041, PR China
| | - Lin Wu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yuhang He
- West China School of Public Health, Sichuan University, Chengdu 610041, PR China
| | - Ni Zeng
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhenglu Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
41
|
Shan S, Cheng D, Li H, Yao W, Kou R, Ji J, Liu N, Zeng T, Zhao X. Short-term PS-NP exposure in early adulthood induces neuronal damage in middle-aged mice via microglia-mediated neuroinflammation. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137615. [PMID: 39978191 DOI: 10.1016/j.jhazmat.2025.137615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Nanoplastics (NPs) are ubiquitous environmental pollutants that have garnered considerable attention for their potential adverse health effects. In this study, male C57BL/6 J mice were orally treated with a mixture of 50-nm and 200-nm polystyrene (PS)-NPs for one week followed by measurements of their neurobehavioral performance and neuronal damage 10 months later. Notably, PS-NPs were detected in the brains of the mice by transmission electron microscopy (TEM) and a nanoscale hyperspectral microscope imaging system 10 months after the PS-NP exposure. The mice exposed to short-term PS-NPs exhibited cognitive dysfunction and anxiety-like symptoms, neuronal damage and synapse loss, and an increase in the number of M1-polarized microglia and A1-reactive astrocytes. Interestingly, the inhibition of microglial activation by minocycline significantly mitigated the PS-NP-induced synapse loss and neuron damage. In vitro studies showed that PS-NPs could be readily internalized by three types of neurovascular unit (NVU) cells, including microglia, astrocytes, and brain microvascular endothelial cells, via multiple pathways. RNA-seq analysis confirmed that microglia-mediated neuronal injury was associated with disturbances in synapse and cell death signaling pathways. Collectively, these findings suggest that short-term PS-NP exposure-induced neuroinflammation in early adulthood may not be resolved naturally but may deteriorate under the interaction of microglia and astrocytes, leading to synapse loss, neuron degeneration, and cognitive dysfunction in middle age. The results of the present study provide important insights into the potential neurological impacts of NPs and suggest that targeting microglia to suppress inflammation might be a potential intervention strategy for neurodegeneration induced by NPs.
Collapse
Affiliation(s)
- Shan Shan
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Dong Cheng
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Hui Li
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Wenhuan Yao
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Ruirui Kou
- Experimental Center, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jing Ji
- Experimental Center, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Na Liu
- Experimental Center, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Xiulan Zhao
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
42
|
Socała K, Kowalczuk-Vasilev E, Komar M, Szalak R, Wyska E, Wlaź P. Effect of apigenin on seizure susceptibility, parvalbumin immunoreactivity, and GABA A receptor expression in the hippocampal neurons in mice. Eur J Pharmacol 2025; 996:177548. [PMID: 40157704 DOI: 10.1016/j.ejphar.2025.177548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Apigenin is a flavonoid compound found in many fruits, vegetables, and medicinal herbs. It possesses a wide range of biological activities including antioxidant, anti-inflammatory, anti-apoptotic, and neuroprotective effects. Some initial evidence also suggests that apigenin may have antiseizure properties. In this study, we aimed at providing more insight into the potential influence of apigenin on seizure susceptibility in mice and its influence on parvalbumin and GABAA receptor immunoreactivity (PV-IR and GABAAR-IR) in the hippocampus. We also assessed the pharmacokinetic profile of apigenin after intraperitoneal route of administration and its possible side effects. Acute administration of apigenin, at doses of 100 and 150 mg/kg, significantly increased the threshold for various types of seizures, i.e., the myoclonic twitch in the ivPTZ test, the 6 Hz-induced seizure, and hindlimb tonus in the maximal electroshock seizure test. A 14-day treatment with apigenin at a lower dose of 50 mg/kg also increased the threshold for maximal electroshock-induced seizures. Pharmacokinetic profiling revealed a very slow elimination of apigenin from serum, and even slower from the brain, after intraperitoneal administration. In consequence, a relatively high accumulation of this compound in the brain can be expected. In addition, apigenin given repeatedly increased the mean number of GABAAR-IR and PV-IR neurons in hippocampal fields suggesting its potential influence on neuronal Ca2+ metabolism. This study indicates that apigenin has a relatively minor effect on acute seizures in mice. However, modifying the quantity of GABAAR-IR and PV-IR hippocampal neurons provides further support for the neuroprotective effects of apigenin.
Collapse
Affiliation(s)
- Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland.
| | - Edyta Kowalczuk-Vasilev
- Institute of Animal Nutrition and Bromatology, Faculty of Animal Science and Bioeconomy, University of Life Sciences, Akademicka 13, PL 20-950, Lublin, Poland
| | - Małgorzata Komar
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, PL 20-950, Lublin, Poland
| | - Radosław Szalak
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, PL 20-950, Lublin, Poland
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, PL 30-688, Kraków, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| |
Collapse
|
43
|
Li Y, Xue J, Ma Y, Ye K, Zhao X, Ge F, Zheng F, Liu L, Gao X, Wang D, Xia Q. The complex roles of m 6 A modifications in neural stem cell proliferation, differentiation, and self-renewal and implications for memory and neurodegenerative diseases. Neural Regen Res 2025; 20:1582-1598. [PMID: 38845217 PMCID: PMC11688559 DOI: 10.4103/nrr.nrr-d-23-01872] [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: 11/13/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 08/07/2024] Open
Abstract
N6-methyladenosine (m 6 A), the most prevalent and conserved RNA modification in eukaryotic cells, profoundly influences virtually all aspects of mRNA metabolism. mRNA plays crucial roles in neural stem cell genesis and neural regeneration, where it is highly concentrated and actively involved in these processes. Changes in m 6 A modification levels and the expression levels of related enzymatic proteins can lead to neurological dysfunction and contribute to the development of neurological diseases. Furthermore, the proliferation and differentiation of neural stem cells, as well as nerve regeneration, are intimately linked to memory function and neurodegenerative diseases. This paper presents a comprehensive review of the roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, as well as its implications in memory and neurodegenerative diseases. m 6 A has demonstrated divergent effects on the proliferation and differentiation of neural stem cells. These observed contradictions may arise from the time-specific nature of m 6 A and its differential impact on neural stem cells across various stages of development. Similarly, the diverse effects of m 6 A on distinct types of memory could be attributed to the involvement of specific brain regions in memory formation and recall. Inconsistencies in m 6 A levels across different models of neurodegenerative disease, particularly Alzheimer's disease and Parkinson's disease, suggest that these disparities are linked to variations in the affected brain regions. Notably, the opposing changes in m 6 A levels observed in Parkinson's disease models exposed to manganese compared to normal Parkinson's disease models further underscore the complexity of m 6 A's role in neurodegenerative processes. The roles of m 6 A in neural stem cell proliferation, differentiation, and self-renewal, and its implications in memory and neurodegenerative diseases, appear contradictory. These inconsistencies may be attributed to the time-specific nature of m 6 A and its varying effects on distinct brain regions and in different environments.
Collapse
Affiliation(s)
- Yanxi Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuejia Ma
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ke Ye
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xue Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fangliang Ge
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feifei Zheng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Basic Medical Institute, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- College of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang Province, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
44
|
Riley E, Cicero N, Mabry SA, Swallow KM, Anderson AK, De Rosa E. Age-related differences in locus coeruleus intensity across a demographically diverse sample. Neurobiol Aging 2025; 150:122-131. [PMID: 40101307 PMCID: PMC11981832 DOI: 10.1016/j.neurobiolaging.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
Understanding the trajectory of in vivo locus coeruleus (LC) signal intensity across the adult lifespan and among various demographic groups, particularly during middle age, may be crucial for early detection of neurodegenerative diseases, which begin in the LC decades before symptom onset. Even though pathological changes in the LC are thought to begin in middle age, its characteristics across the adult lifespan, and its consistency and variation across demographic groups, remain not well understood. Using T1-weighted turbo spin echo magnetic resonance (MRI) scans to characterize the LC, we measured LC signal intensity in 134 participants aged 19-86 years, with an effort to recruit a more racially diverse sample (41 % non-White). LC signal intensity was lowest in early adulthood, peaked around age 60, and then decreased again in the oldest adults, particularly in the caudal portion of the LC, which exhibited the greatest overall signal intensity; education, income, and history of early trauma did not alter this general pattern. Rostral LC signal intensity was further heightened in women and Black participants. In higher-performing older adults, increased rostral LC signal intensity was positively associated with higher fluid cognition. The potential accumulation of LC signal intensity across the adult lifespan and its possible dissipation in later life as well as its modification by demographic factors, may be associated with differential susceptibility to neurocognitive aging.
Collapse
Affiliation(s)
- Elizabeth Riley
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA.
| | - Nicholas Cicero
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA
| | | | - Khena M Swallow
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA
| | - Adam K Anderson
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA
| | - Eve De Rosa
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
45
|
He S, Lu JJ, Wu JJ, Zheng MX, Ma J, Hua XY, Xu JG. Altered cerebellar activity and cognitive deficits in Type 2 diabetes: Insights from resting-state fMRI. Brain Res 2025; 1856:149586. [PMID: 40113193 DOI: 10.1016/j.brainres.2025.149586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/13/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE To investigate alterations in brain activity in patients with Type 2 Diabetes and explore the relationship between altered regions and neuropsychological performances. METHODS A total of 36 patients with Type 2 Diabetes and 40 age- and education-matched healthy controls were recruited for this case-control study. All participants underwent resting-state functional magnetic resonance imaging (Resting-state fMRI) and neuropsychological tests. The neuropsychological scales included the Auditory Verbal Learning Test (AVLT), Shape Trajectory Test B (STT-B), Hamilton Anxiety Scale (HAMA), Hamilton Depression Scale (HAMD), and Boston Naming Test (BNT), Symbol Digit Modality Test (SDMT), Regional homogeneity (ReHo) and the amplitude of low-frequency fluctuations (ALFF) were used to assess differences in spontaneous regional brain activity. For functional connectivity (FC) analyses, the differences identified among the groups were selected as seed regions. Then, the correlations between neuropsychological scale scores (AVLT, HAMA, HAMD, STT-B, BNT, and SDMT) and ALFF/ReHo values were specifically analyzed in the focal regions that exhibited significant alterations between the T2DM and control groups, as detailed in Tables 2 and 3. RESULTS Patients with Type 2 Diabetes exhibited significantly higher ALFF values in the superior lobe of the cerebellum, specifically in the left cerebellar crus I (Cerebellum_Crus I_L), left cerebellar lobule VI (Cerebellum_6_L), and left cerebellar lobule IV-V (Cerebellum_4_5_L). Additionally, they exhibited elevated ReHo values in the Cerebellum_Crus I_L and Cerebellum_6_L. The findings were statistically significant with a family-wise error-corrected, cluster-level p-value of less than 0.05. However, the FC analysis was not significant. AVLT scores were significantly lower in the diabetes group. The correlation analysis demonstrated a negative association between ALFF values of the Cerebellum_6_L and AVLT scores (R2 = 0.1375, P < 0.001). The ReHo values within the Cerebellum_6_L also exhibited a negative association with AVLT scores (R2 = 0.0937, P = 0.007). CONCLUSION Patients with Type 2 Diabetes showed abnormal neural activities in diverse cerebellar regions mainly related to cognitive functions. This provides supplementary information to deepen our insight into the neural mechanisms by which Type 2 Diabetes affects the functional activity of the brain's posterior circulation, as well as the potential association of these changes with cognitive impairment.
Collapse
Affiliation(s)
- Shuang He
- Department of Orthopaedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Juan-Juan Lu
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Jia Wu
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Mou-Xiong Zheng
- Department of Orthopaedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China; Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, China
| | - Jie Ma
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xu-Yun Hua
- Department of Orthopaedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China; Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, China.
| | - Jian-Guang Xu
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, China.
| |
Collapse
|
46
|
Zhao Q, Wang Y, Lu Y, Luan M, Gao S, Li X, Zhou C. Computational bases of domain-specific action anticipation superiority in experts: Kinematic invariants mapping. Cognition 2025; 259:106121. [PMID: 40086085 DOI: 10.1016/j.cognition.2025.106121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
While experts consistently demonstrate superior action anticipation within their domains, the computational mechanisms underlying this ability remain unclear. This study investigated how the processing of kinematic invariants contributes to expert performance by examining table tennis players, volleyball players, and novices across two table tennis serve anticipation tasks using normal and point-light displays. Employing the kinematic coding framework, we established encoding and readout models to predict both actual action outcomes and participants' responses. Results showed that table tennis players consistently outperformed other groups across both tasks. Analysis of the intersection between encoding and readout models revealed a distinct mechanism: while both athlete groups showed enhanced ability to identify informative kinematic features compared to novices, only table tennis players demonstrated superiority in correctly utilizing these features to make precise predictions. This advantage in invariants mapping showed a positive correlation with domain-specific training experience and remained consistent across display formats, suggesting the development of a robust internal model through sustained domain-specific experience. Our findings illuminate the computational bases of domain-specific action anticipation, highlighting the significance of kinematic invariants mapping superiority in experts.
Collapse
Affiliation(s)
- Qiwei Zhao
- School of Psychology, Shanghai University of Sport, Shanghai 200438, China; Department of Neurology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Yinyue Wang
- School of Psychology, Shanghai University of Sport, Shanghai 200438, China.
| | - Yingzhi Lu
- School of Psychology, Shanghai University of Sport, Shanghai 200438, China.
| | - Mengkai Luan
- School of Psychology, Shanghai University of Sport, Shanghai 200438, China.
| | - Siyu Gao
- School of Psychology, Shanghai University of Sport, Shanghai 200438, China.
| | - Xizhe Li
- School of Psychology, Shanghai University of Sport, Shanghai 200438, China.
| | - Chenglin Zhou
- School of Psychology, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
47
|
Li T, Wang N, Yi D, Xiao Y, Li X, Shao B, Wu Z, Bai J, Shi X, Wu C, Qiu T, Yang G, Sun X, Zhang R. ROS-mediated ferroptosis and pyroptosis in cardiomyocytes: An update. Life Sci 2025; 370:123565. [PMID: 40113077 DOI: 10.1016/j.lfs.2025.123565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
The cardiomyocyte is an essential component of the heart, communicating and coordinating with non-cardiomyocytes (endothelial cells, fibroblasts, and immune cells), and are critical for the regulation of structural deformation, electrical conduction, and contractile properties of healthy and remodeled myocardium. Reactive oxygen species (ROS) in cardiomyocytes are mainly produced by the mitochondrial oxidative respiratory chain, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), xanthine oxidoreductase (XOR), monoamine oxidase (MAO), and p66shc. Under physiological conditions, ROS are involved in the regulation of cardiac development and cardiomyocyte maturation, cardiac calcium handling, and excitation-contraction coupling. In contrast, dysregulation of ROS metabolism is involved in the development and progression of cardiovascular diseases (CVDs), including myocardial hypertrophy, hyperlipidemia, myocardial ischemia/reperfusion injury, arrhythmias and diabetic cardiomyopathy. Further oxidative stress induced by ROS dyshomeostasis was found to be the major reason for cardiomyocyte death in cardiac diseases, and in recent years, ferroptosis induced by oxidative stress have been considered to be fatal to cardiomyocytes. In addition, ROS is also a key trigger for the activation of pyroptosis, which induces and exacerbates the inflammatory response caused by various cardiac diseases and plays a critical role in CVDs. Therefore, in this review, the sources and destinations of ROS in cardiomyocytes will be systematically addressed, so as to reveal the molecular mechanisms by which ROS accumulation triggers cardiomyocyte ferroptosis and pyroptosis under pathological conditions, and provide a new concept for the research and treatment of heart-related diseases.
Collapse
Affiliation(s)
- Tao Li
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, PR China
| | - Ningning Wang
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China; Global Health Research Center, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Dongxin Yi
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Yuji Xiao
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China; Bishan Hospital of Chongqing Medical University, Chongqing 402760, PR China
| | - Xiao Li
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Bing Shao
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Ziyi Wu
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Jie Bai
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Xiaoxia Shi
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Chenbing Wu
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Tianming Qiu
- Global Health Research Center, Dalian Medical University, Dalian, Liaoning 116044, PR China; Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Guang Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Xiance Sun
- Global Health Research Center, Dalian Medical University, Dalian, Liaoning 116044, PR China; Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Rongfeng Zhang
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, PR China.
| |
Collapse
|
48
|
Ganse-Dumrath A, Chohan A, Samuel S, Bretherton P, Haenschel C, Fett AK. Systematic review and meta-analysis of early visual processing, social cognition, and functional outcomes in schizophrenia spectrum disorders. Schizophr Res Cogn 2025; 40:100351. [PMID: 40028174 PMCID: PMC11872129 DOI: 10.1016/j.scog.2025.100351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
Non-affective psychotic disorders are marked by cognitive and sensory processing abnormalities, including in early visual processing and social cognition. Understanding the relationships between these deficits and their impact on daily-life functional outcomes may help to improve outcomes in affected individuals. This systematic review and meta-analysis aimed to summarise the existing evidence on the relationships between early visual processing, social cognition, and functional outcomes, and to assess the evidence regarding the mediating role of social cognition in the association between early visual processing and functional outcomes in individuals with schizophrenia spectrum disorders. A comprehensive search across five databases identified 364 potentially eligible studies, with eight articles meeting all inclusion criteria. Meta-analytic techniques were employed to synthesise effect sizes and assess a meta-mediation model. Three random-effects meta-analyses revealed significant associations between all three domains of interest. Social cognition partially mediated the relationship between early visual processing and functional outcomes. The direct effect of early visual processing on functional outcomes remained significant, albeit with a reduced effect size. The findings suggest that interventions targeting both early visual processing and social cognition concurrently may improve functional outcomes more effectively than focusing on either domain alone.
Collapse
Affiliation(s)
- Akke Ganse-Dumrath
- Department of Psychology, School of Health and Medical Sciences, City St George's, University of London, UK
| | - Anya Chohan
- Department of Psychology, School of Health and Medical Sciences, City St George's, University of London, UK
| | - Steven Samuel
- Department of Psychology, School of Health and Medical Sciences, City St George's, University of London, UK
| | - Paul Bretherton
- Department of Psychology, School of Health and Medical Sciences, City St George's, University of London, UK
| | - Corinna Haenschel
- Department of Psychology, School of Health and Medical Sciences, City St George's, University of London, UK
| | - Anne-Kathrin Fett
- Department of Psychology, School of Health and Medical Sciences, City St George's, University of London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK
| |
Collapse
|
49
|
Chen Z, He J, Guo Y, Hao Y, Lv W, Chen Z, Wang J, Yang Y, Wang K, Liu Z, Ouyang Q, Su Z, Hu P, Xiao G. Adherent junctions: Physiology, role in hydrocephalus and potential therapeutic targets. IBRO Neurosci Rep 2025; 18:283-292. [PMID: 39995568 PMCID: PMC11849119 DOI: 10.1016/j.ibneur.2025.02.003] [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: 11/30/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
In all epithelial cells, the adherent junctions (AJs) with cadherin as the core play an important role in the maintenance of the connection and the formation of apical-basal polarity. The ependymal cells close to the ventricular system rely on AJs with N-cadherin at the core to maintain their normal morphology and function. Therefore, it has an important impact on the function and disease of the central nervous system. Hydrocephalus is a pathological phenomenon of excessive cerebrospinal fluid accumulating in the ventricular system accompanied by continuous ventricular dilatation, which can be divided into obstructive hydrocephalus and communicating hydrocephalus according to the pathogenesis. Obstructive hydrocephalus is often associated with excessive ependymal cells produced by differentiation of radial glial cells. The etiology of communicating hydrocephalus is mainly related to the dyskinesia of cerebrospinal fluid. In addition, the damage of the brain barrier can lead to brain edema and aggravate the symptoms. At present, the researches on the pathogenesis of hydrocephalus are mainly focused on the development of ependymal cells and cilia, while less attention has been paid to molecules such as AJs, which play an important role in maintaining the polarity of ependymal cells. This paper discusses the formation and function of AJs and their role in preventing hydrocephalus by preserving the polarity of ependymal cilia, regulating the number of ependymal cells, and upholding the brain barrier integrity to impede hydrocephalus exacerbation, which provides a new direction for the study of hydrocephalus.
Collapse
Affiliation(s)
- Zhiye Chen
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan 410008, PR China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jian He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yating Guo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yue Hao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wentao Lv
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zexin Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Junqiang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Kaiyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qian Ouyang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Department of Neurosurgery, Zhuzhou Hospital, Central South University Xiangya School of Medicine, Zhuzhou, Hunan 412000, PR China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB21 2QQ, UK
| | - Pingsheng Hu
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan 410008, PR China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
50
|
Wu Y, Wang C, Qian W, Wang L, Yu L, Zhang M, Yan M. Default mode network-basal ganglia network connectivity predicts the transition to postherpetic neuralgia. IBRO Neurosci Rep 2025; 18:135-141. [PMID: 39896717 PMCID: PMC11783054 DOI: 10.1016/j.ibneur.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Background Neuroimaging studies have revealed aberrant network functional connectivity in postherpetic neuralgia (PHN) patients. However, there is a lack of knowledge regarding the relationship between the brain network connectivity during the acute period and disease prognosis. Objective The purpose of this study was to detect characteristic network connectivity in the process of herpes zoster (HZ) pain chronification and to identify whether abnormal network connectivity in the acute period can predict the outcome of patients with HZ. Methods In this cross-sectional study, 31 patients with PHN, 33 with recuperation from herpes zoster (RHZ), and 28 with acute herpes zoster (AHZ) were recruited and underwent resting-state functional magnetic resonance imaging (fMRI). We investigated the differences in the connectivity of four resting-state networks (RSN) among the three groups. Receiver operating characteristic (ROC) curve analysis was performed to identify whether abnormal network connectivity in the acute period could predict the outcome of patients with HZ. Results First, we found within-basal ganglia network (BGN) and default mode network (DMN)-BGN connectivity differences, with PHN patients showing increased DMN-BGN connectivity compared to AHZ and RHZ patients, while RHZ patients showing increased within-BGN connectivity compared to AHZ and PHN patients. Moreover, DMN-BGN connectivity was associated with the ID pain score in patients with AHZ. Finally, the DMN-BGN connectivity of AHZ patients could predict the outcome of HZ patients with sensitivity and specificity of 77.8 % and 63.2 %, respectively. Conclusions Our results provide evidence that DMN-BGN connectivity during the acute period confers a risk for the development of chronic pain and can act as a neuroimaging biomarker to predict the outcome of patients with HZ.
Collapse
Affiliation(s)
- Ying Wu
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Chao Wang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Wei Qian
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Lieju Wang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Lina Yu
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Minming Zhang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Min Yan
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| |
Collapse
|