1
|
Jia F, Fu L. Roles of Ubiquitin Ligases and Deubiquitylases in Alzheimer's Disease. Mol Neurobiol 2025; 62:7747-7761. [PMID: 39932514 DOI: 10.1007/s12035-025-04739-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 02/02/2025] [Indexed: 05/15/2025]
Abstract
The mechanisms responsible for the accumulation of Aβ plaques and neurofibrillary tangles, composed of phosphorylated Tau protein, in Alzheimer's disease (AD) remain a mystery. Dysfunction of the ubiquitin-proteasome system (UPS) largely contributes to abnormal protein aggregation. A cascade of ubiquitinating enzymes promotes protein ubiquitination, while deubiquitylases (DUBs) regulate its reversal. Disruptions in ubiquitination and deubiquitination processes result in abnormal protein aggregation and the formation of inclusion bodies, ultimately leading to neuronal damage. Recent studies have highlighted the significant role of protein ubiquitination and deubiquitination in the pathogenesis of AD. E3 ubiquitin ligases, which facilitate protein ubiquitination, are beneficial for Aβ clearance, synaptic function, gap junction maintenance, mitophagy, and neuroinflammation. Conversely, DUBs, responsible for removing ubiquitin from substrate proteins, inhibit Aβ and Tau degradation while promoting neuroinflammation in neurons. This review provides a thorough overview of the involvement of E3 ubiquitin ligases and DUBs in AD, highlighting their diverse roles in aspects of pathophysiological processes.
Collapse
Affiliation(s)
- Fengju Jia
- School of Nursing, Qingdao University, No.308 Ningxia Road, Qingdao, 266071, China.
| | - Lin Fu
- School of Basic Medicine, Qingdao University, No.308 Ningxia Road, Qingdao, 266072266071, China
| |
Collapse
|
2
|
BU C, ZHU X, ZHANG Q, SHAO W. [Recent advances on the role of exosomes in neurodegenerative diseases]. Se Pu 2025; 43:487-497. [PMID: 40331612 PMCID: PMC12059994 DOI: 10.3724/sp.j.1123.2024.10035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Indexed: 05/08/2025] Open
Abstract
Exosomes are nano-sized, lipid bilayer vesicles secreted by cells. They carry essential bioactive molecules, such as proteins, nucleic acids, and lipids, and are widely present in bodily fluids including blood and cerebrospinal fluid. Exosomes transfer bioactive molecules to target cells through various mechanisms, including endocytosis, ligand-receptor interactions, or direct membrane fusion, and play crucial roles in intercellular communication, including facilitating intercellular information exchange, maintaining nerve-cell function, participating in immune responses, and providing nutritional support. Exosomes significantly promote signal transmission and intercellular communication in the central nervous system and are involved in the pathogenesis and development of diseases by participating in the spread of pathological proteins, regulating neuroinflammation, and the deposition of pathological proteins. Therefore, exosomes play key roles in the occurrence and development of neurodegenerative diseases, and their contents, especially proteins and miRNAs, are specific for given pathological and physiological states and are relatively stable during extraction and analysis. Hence, exosomes are ideal tools for diagnosing diseases, staging their courses, and assisting prognosis. This article further explores exosomes derived from blood, saliva, urine, and cerebrospinal fluid as potential diagnostic biomarkers for neurodegenerative diseases. As natural drug-delivery systems, exosomes have the advantages of biocompatibility, ability to cross biological barriers, target specificity, stability, and containing natural therapeutic molecules, which can effectively improve the precision and efficacy of drug delivery and reduce side effects, making them an ideal carrier for delivering drugs to the central nervous system. Therefore, exosomes hold great potential in the diagnosis and treatment of central nervous system diseases. This article systematically reviews the latest advances in exosome research directed toward specific neurodegenerative diseases, focusing on their roles played in disease pathogenesis, progression, diagnosis, and treatment, with the aim of providing theoretical support and a reference for the early diagnosis and treatment of these diseases.
Collapse
|
3
|
Randhawa S, Saini TC, Bathla M, Bhardwaj R, Dhiman R, Acharya A. Nanomaterials in targeting amyloid-β oligomers: current advances and future directions for Alzheimer's disease diagnosis and therapy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:561-580. [PMID: 40297247 PMCID: PMC12035877 DOI: 10.3762/bjnano.16.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
The amyloid cascade hypothesis posits that amyloid-β oligomers (AβOs) are the most neurotoxic species in Alzheimer's disease (AD). These oligomers, characterized by their high β-sheet content, have been shown to significantly disrupt cell membranes, induce local inflammation, and impair autophagy processes, which collectively contribute to neuronal loss. As such, targeting AβOs specifically, rather than solely focusing on amyloid-β fibrils (AβFs), may offer a more effective therapeutic approach for AD. Recent advances in detection and diagnosis have emphasized the importance of accurately identifying AβOs in patient samples, enhancing the potential for timely intervention. In recent years, nanomaterials (NMs) have emerged as promising agents for addressing AβOs regarding their multivalent interactions, which can more effectively detect and inhibit AβO formation. This review provides an in-depth analysis of various nanochaperones developed to target AβOs, detailing their mechanisms of action and therapeutic potential via focusing on two main strategies, namely, disruption of AβOs through direct interaction and the inhibition of AβO nucleation by binding to intermediates of the oligomerization process. Evidence from in vivo studies indicate that NMs hold promise for ameliorating AD symptoms. Additionally, the review explores the different interaction mechanisms through which nanoparticles exhibit their inhibitory effects on AβOs, providing insights into their potential for clinical application. This comprehensive overview highlights the current advancements in NM-based therapies for AD and outlines future research directions aimed at optimizing these innovative treatments.
Collapse
Affiliation(s)
- Shiwani Randhawa
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Trilok Chand Saini
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Manik Bathla
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rahul Bhardwaj
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rubina Dhiman
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
| | - Amitabha Acharya
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
4
|
Jung YH, Jo HY, Kim DH, Oh YJ, Kim M, Na S, Song HY, Lee HJ. Exosome-Mediated Mitochondrial Regulation: A Promising Therapeutic Tool for Alzheimer's Disease and Parkinson's Disease. Int J Nanomedicine 2025; 20:4903-4917. [PMID: 40259919 PMCID: PMC12011032 DOI: 10.2147/ijn.s513816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are representative neurodegenerative diseases with abnormal energy metabolism and altered distribution and deformation of mitochondria within neurons, particularly in brain regions such as the hippocampus and substantia nigra. Neurons have high energy demands; thus, maintaining a healthy mitochondrial population is important for their biological function. Recently, exosomes have been reported to have mitochondrial regulatory potential and antineurodegenerative properties. This review presents the mitochondrial abnormalities in brain cells associated with AD and PD and the potential of exosomes to treat these diseases. Specifically, it recapitulates research on the molecular mechanisms whereby exosomes regulate mitochondrial biogenesis, fusion/fission dynamics, mitochondrial transport, and mitophagy. Furthermore, this review discusses exosome-triggered signaling pathways that regulate nuclear factor (erythroid-derived 2)-like 2-dependent mitochondrial antioxidation and hypoxia inducible factor 1α-dependent metabolic reprogramming. In summary, this review aims to provide a profound understanding of the regulatory effect of exosomes on mitochondrial function in neurons and to propose exosome-mediated mitochondrial regulation as a promising strategy for AD and PD.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Hyo Youn Jo
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Dae Hyun Kim
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Yeon Ju Oh
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Minsoo Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Seunghyun Na
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of Korea
- Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Republic of Korea
| |
Collapse
|
5
|
Hewitt K, Huang XF. The Role of Microglial Exosomes in Clozapine Treatment: Effect on Cognition in Schizophrenia. J Neuroimmune Pharmacol 2025; 20:42. [PMID: 40238023 PMCID: PMC12003456 DOI: 10.1007/s11481-024-10160-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/22/2024] [Indexed: 04/18/2025]
Abstract
Schizophrenia is a complex neuropsychiatric disorder characterized by a spectrum of symptoms including cognitive impairments and psychotic episodes. Clozapine, an atypical antipsychotic drug, is a widely recognised treatment option for patients with drug-resistant schizophrenia, due to it having the highest efficacy out of all the antipsychotic drugs. Despite its efficacy, clozapine's impact on cognition and brain structure in schizophrenia patients remains a subject of ongoing research and debate, with accumulating evidence indicating negative impacts on cognitive performance and changes in brain volume. Changes in the immune system are linked to variations in cognitive functioning in schizophrenia. Previous research has indicated that microglia, the primary innate immune cells of the brain, have been associated with decreased cognitive performance when dysfunctional. Evidence suggests that brain structure may mediate the observed relationship between microglia and cognition. Microglial exosomes, integral to neuroinflammation and cellular communication, could provide insight into the neurobiological mechanisms underpinning the effects of clozapine treatment. This review focuses on the proposition that alterations in microglial exosome composition, particularly miRNAs, are involved in mediating clozapine's diverse effects on cognition by influencing brain macrostructure. This review aims to highlight new directions for future research that could lead to more effective and targeted therapeutic approaches in the management of schizophrenia.
Collapse
Affiliation(s)
- Kyle Hewitt
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, 2522, Australia
| | - Xu-Feng Huang
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, 2522, Australia.
| |
Collapse
|
6
|
Zhao M, Wang J, Zhu S, Zhang S, Han C, Tan C, Huang Y, Sun Z, Wang L, Liu J. Human neural stem cell-derived exosomes activate PINK1/Parkin pathway to protect against oxidative stress-induced neuronal injury in ischemic stroke. J Transl Med 2025; 23:402. [PMID: 40188077 PMCID: PMC11971779 DOI: 10.1186/s12967-025-06283-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/20/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Mitochondria play a critical role in oxidative stress (OS)-induced neuronal injury during ischemic stroke (IS), making them promising therapeutic targets. Mounting evidence underscores the extraordinary therapeutic promise of exosomes derived from human neural stem cells (hNSCs) in the management of central nervous system (CNS) diseases. Nonetheless, the precise mechanisms by which these exosomes target mitochondria to ameliorate the effects of IS remain only partially elucidated. This study investigates the protective effects of hNSC derived exosomes (hNSC-Exos) on neuronal damage. METHODS Using a rat model of middle cerebral artery occlusion (MCAO) in vivo and OS-induced HT22 cells in vitro. Firstly, our research group independently isolated human neural stem cells (hNSCs) and subsequently prepared hNSC-Exos. In vivo, MCAO rats were restored to blood flow perfusion to simulate ischemia-reperfusion injury, and hNSC-Exos were injected through stereotaxic injection into the brain. Subsequently, the protective effects of hNSC-Exos on MCAO rats were evaluated, including histological studies, behavioral assessments. In vivo, H2O2 was used in HT22 cells to simulate the OS environment in MCAO, and then its protective effects on HT22 were evaluated by co-culturing with hNSC-Exos, including immunofluorescence staining, western blotting (WB), quantitative real time PCR (qRT-PCR). In the process of exploring specific mechanisms, we utilized RNA sequencing (RNA-seq) to detect the potential induction of mitophagy in OS-induced HT22 cells. Afterwards, we employed a series of mitochondrial function assessments and autophagy related detection techniques, including measuring mitochondrial membrane potential, reactive oxygen species (ROS) levels, transmission electron microscopy (TEM) imaging, monodansylcadaverine (MDC) staining, and mCherry-GFP-LC3B staining. In addition, we further investigated the regulatory pathway of hNSC-Exos by using autophagy inhibitor mdivi-1 and knocking out PTEN induced kinase 1 (PINK1) in HT22 cells. RESULTS Administration of hNSC-Exos significantly ameliorated brain tissue damage and enhanced behavioral outcomes in MCAO rats. This treatment led to a reduction in brain tissue apoptosis and facilitated the normalization of impaired neurogenesis and neuroplasticity. Notably, the application of hNSC-Exos in vitro resulted in an upregulation of mitophagy in HT22 cells, thereby remedying mitochondrial dysfunction. We demonstrate that hNSC-Exos activate mitophagy via the PINK1/Parkin pathway, improving mitochondrial function and reducing neuronal apoptosis. CONCLUSIONS These findings suggest that hNSC-Exos alleviate OS-induced neuronal damage by regulating the PINK1/Parkin pathway. These reveals a novel role of stem cell-derived mitochondrial therapy in promoting neuroprotection and suggest their potential as a therapeutic approach for OS-associated CNS diseases, including IS.
Collapse
Affiliation(s)
- Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Shuaiyu Zhu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Shensen Zhang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Chao Han
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Chengcheng Tan
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Yubing Huang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Zhaokai Sun
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China.
| |
Collapse
|
7
|
Huang M, Di J, He L, Li N, Tian Z, Xiao L, Zhu R, He T, Pang M, Liu B, Rong L. Double-target magnetic stimulation attenuates oligodendrocyte apoptosis and oxidative stress impairment after spinal cord injury via GAP43. Spine J 2025; 25:820-842. [PMID: 39701305 DOI: 10.1016/j.spinee.2024.12.025] [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: 07/16/2024] [Revised: 11/29/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND CONTEXT Spinal cord injury (SCI) causes neural circuit interruption and permanent functional damage. Magnetic stimulation in humans with SCI aims to engage residual neural networks to improve neurological functional, but the detailed mechanism remains unknown. PURPOSE This study evaluates functional recovery and neural circuitry improvements in rodent with double-target (brain and spinal cord) magnetic stimulation (DTMS) treatment and explores the effect of DTMS on the modulation of glial cells in vivo and in vitro. STUDY DESIGN In vivo animal study. METHODS SCI model rats at T10 level were induced via a weight-drop method and underwent long-time DTMS treatment. A series of behavioral assessments and tissue staining were used to evaluate neurological function and neural circuitry improvements. More importantly, single-cell RNA sequencing was conducted to identify the most significant glial cells after DTMS treatment. Furthermore, transmission electron microscopy, western blotting, immunofluorescence staining, TUNEL staining, Annexin V-FITC apoptosis kit and Lipid ROS kit were used to explore the mechanism underlying the observed changes. Study funding sources: National Natural Science Foundation of China (Grant number: U22A20297; Dollar amount: 62500); Key Research and Development Program of Guangzhou (Grant number: 202206060003; Dollar amount: 63750). There are no conflicts of interest or disclosures to report. RESULTS DTMS promoted the improvements of motor and sensory neural circuitry by modulating remyelination and neuronal survival, while silencing growth-associated protein 43 (GAP43) in oligodendrocytes suppressed these effects of DTMS in vivo. Mechanically, GAP43 played a crucial part to promote the branching and mature of oligodendrocytes and axonal regeneration via anti-apoptotic and antioxidative stress effects. Furthermore, oligodendrocytes subjected to magnetic stimulation exerted neuroprotective effects on neurons by secreting exosomes containing GAP43. CONCLUSIONS Our study revealed the neuroprotection of DTMS on SCI. The GAP43 in oligodendrocytes were associated with this relationship between magnetic stimulation and myelin and neuronal regeneration after SCI. CLINICAL SIGNIFICANCE The current study demonstrated the beneficial effects of DTMS on SCI based on functional, electrophysiological, cellular and histological evidence. According to these findings, we expect DTMS to make a positive and significant difference for SCI therapeutic screening.
Collapse
Affiliation(s)
- Mudan Huang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Jiawei Di
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Lei He
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Na Li
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Longyou Xiao
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Ruijue Zhu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Tianwei He
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
8
|
Yang Y, Chen H, Huang S, Chen H, Verkhratsky A, Niu J, Qu Y, Yi C. BOK-engaged mitophagy alleviates neuropathology in Alzheimer's disease. Brain 2025; 148:432-447. [PMID: 39054908 DOI: 10.1093/brain/awae241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 06/15/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Mitochondrial malfunction associated with impaired mitochondrial quality control and self-renewal machinery, known as mitophagy, is an under-appreciated mechanism precipitating synaptic loss and cognitive impairments in Alzheimer's disease. Promoting mitophagy has been shown to improve cognitive function in Alzheimer's disease animals. However, the regulatory mechanism was unclear, which formed the aim of this study. Here, we found that a neuron-specific loss of Bcl-2 family member BOK in patients with Alzheimer's disease and APPswe/PS1dE9 (APP/PS1) mice is closely associated with mitochondrial damage and mitophagy defects. We further revealed that BOK is the key to the Parkin-mediated mitophagy through competitive binding to the MCL1/Parkin complex, resulting in Parkin release and translocation to damaged mitochondria to initiate mitophagy. Furthermore, overexpressing bok in hippocampal neurons of APP/PS1 mice alleviated mitophagy and mitochondrial malfunction, resulting in improved cognitive function. Conversely, the knockdown of bok worsened the aforementioned Alzheimer's disease-related changes. Our findings uncover a novel mechanism of BOK signalling through regulating Parkin-mediated mitophagy to mitigate amyloid pathology, mitochondrial and synaptic malfunctions, and cognitive decline in Alzheimer's disease, thus representing a promising therapeutic target.
Collapse
Affiliation(s)
- Yang Yang
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Shuwen Huang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Hao Chen
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Bilbao 48011, Spain
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius 01102, Lithuania
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang 110122, China
| | - Jianqin Niu
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
- Chongqing Key Laboratory of Neurobiology, Chongqing 400038, China
| | - Yibo Qu
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou 50630, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen 518107, China
| |
Collapse
|
9
|
Chen L, Yan X. LncRNA NORAD sponging to miR-26b-5p represses the progression of Alzheimer's disease in vitro by upregulating MME expression. Cytotechnology 2025; 77:41. [PMID: 39803415 PMCID: PMC11723864 DOI: 10.1007/s10616-024-00691-6] [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: 11/22/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological condition that causes brain shrinkage and cell death. This study aimed to identify the role of the NORAD/miR-26b-5p axis in AD. StarBase was used to examine the binding sequences of miR-26b-5p to LncRNA NORAD or its target genes, which were verified by a double luciferase reporter assay. PC12 cells were processed with Aβ1-42 to construct an AD model in vitro, and LncRNA NORAD and miR-26b-5p levels in PC12 cells were identified by RT-qPCR. Cell viability and apoptosis were measured using the MTT assay and flow cytometry, respectively. LDH release and oxidative stress-related indicators (MDA, SOD, and CAT) were detected using the corresponding kits, and the levels of Bcl-2 and Bax were assessed by western blotting and RT-qPCR. Aβ1-42 distinctly decreased LncRNA NORAD and membrane metalloendopeptidase (MME) levels in PC12 cells, while miR-26b-5p was generally increased. The LncRNA NORAD can adsorb miR-26b-5p, and the target gene of miR-26b-5p is neprilysin (MME). In the Aβ1-42 induced AD model, PC12 cell activity decreased, LDH release and apoptosis increased, oxidative stress level increased, Bax expression increased, and Bcl-2 expression decreased. LncRNA NORAD plays a protective role in AD cell models by abrogating miR-26b-5p levels. Inhibition of MME expression eliminated the protective effects of the miR-26b-5p inhibitor in AD cell models. LncRNA NORAD inhibits AD progression in vitro by modulating the miR-26b-5p-MME signaling axis. The LncRNA NORAD/miR-26b-5p is expected to be a prospective therapeutic candidate for AD. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00691-6.
Collapse
Affiliation(s)
- Lizhu Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Traditional and Western Medicine, Jianghan District, No. 11 Lingjiaohu Road, Wuhan, 430015 China
| | - Xiaoqiong Yan
- Department of Neurology, Hubei Provincial Hospital of Integrated Traditional and Western Medicine, Jianghan District, No. 11 Lingjiaohu Road, Wuhan, 430015 China
| |
Collapse
|
10
|
Guo Y, Zhao J, Liu X, Lu P, Liang F, Wang X, Wu J, Hai Y. Ghrelin Induces Ferroptosis Resistance and M2 Polarization of Microglia to Alleviate Neuroinflammation and Cognitive Impairment in Alzheimer's Disease. J Neuroimmune Pharmacol 2025; 20:6. [PMID: 39797928 DOI: 10.1007/s11481-024-10165-3] [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: 07/26/2024] [Accepted: 11/29/2024] [Indexed: 01/13/2025]
Abstract
Microglial polarization and ferroptosis are important pathological features in Alzheimer's disease (AD). Ghrelin, a brain-gut hormone, has potential neuroprotective effects in AD. This study aimed to explore the potential mechanisms by which ghrelin regulates the progression of AD, as well as the crosstalk between microglial polarization and ferroptosis. Mouse BV2 microglial cells and male mice were treated with beta-amyloid (Aβ) (1-42) to simulate the AD environment. Microglia ferroptosis was measured by detecting levels of ferroptosis-related proteins (SLC7A11, GPX4, FTL1, and FTH1), metabolic markers (ROS, MDA, GSH, SOD), and observing mitochondrial morphological changes. Microglial polarization was evaluated by measuring levels of inflammatory markers and surface markers. The impact of ghrelin on Aβ1-42-exposed microglia was assessed by coupling with the ferroptosis activator Erastin. Cognitive impairment in AD mice was evaluated through behavioral tests. Tissue staining was applied to determine neuronal damage. In Aβ1-42-exposed microglia, ghrelin upregulated the protein expression of SLC7A11, GPX4, FTL1 and FTH1, reduced ROS and MDA levels, and elevated GSH and SOD levels through the BMP6/SMAD1 pathway. Ghrelin alleviated mitochondrial structural damage. Additionally, ghrelin reduced levels of pro-inflammatory factors and CD86, while increasing levels of anti-inflammatory factors and CD206. Erastin reversed the effects of ghrelin on ferroptosis and phenotypic polarization in Aβ1-42-exposed microglia. In AD mice, ghrelin ameliorated abnormal behavior, neuroinflammation, and plaque deposition. Ghrelin attenuated iNOS/IBA1-positive expression and enhanced Arg-1/IBA1-positive expression in the hippocampus. Ghrelin induces microglial M2 polarization by inhibiting microglia ferroptosis, thereby alleviating neuroinflammation. Our results indicate that ghrelin may serve as a promising potential agent for treating cognitive impairment in AD.
Collapse
Affiliation(s)
- Yaoxue Guo
- Department of Clinical Pharmacy, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Junli Zhao
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Xing Liu
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Pu Lu
- Oncology Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Furu Liang
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Xueyan Wang
- Oncology Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Jing Wu
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Yan Hai
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China.
- Baotou Central Hospital, 61 Huancheng Road, Donghe District, Baotou, 014040, Inner Mongolia, China.
| |
Collapse
|
11
|
Wang L, Zhang X, Yang Z, Wang B, Gong H, Zhang K, Lin Y, Sun M. Extracellular vesicles: biological mechanisms and emerging therapeutic opportunities in neurodegenerative diseases. Transl Neurodegener 2024; 13:60. [PMID: 39643909 PMCID: PMC11622582 DOI: 10.1186/s40035-024-00453-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles originating from different cells within the brain. The pathophysiological role of EVs in neurodegenerative diseases is progressively acknowledged. This field has advanced from basic biological research to essential clinical significance. The capacity to selectively enrich specific subsets of EVs from biofluids via distinctive surface markers has opened new avenues for molecular understandings across various tissues and organs, notably in the brain. In recent years, brain-derived EVs have been extensively investigated as biomarkers, therapeutic targets, and drug-delivery vehicles for neurodegenerative diseases. This review provides a brief overview of the characteristics and physiological functions of the various classes of EVs, focusing on the biological mechanisms by which various types of brain-derived EVs mediate the occurrence and development of neurodegenerative diseases. Concurrently, novel therapeutic approaches and challenges for the use of EVs as delivery vehicles are delineated.
Collapse
Affiliation(s)
- Ling Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaoyan Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ziyi Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Binquan Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hongyang Gong
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ke Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yi Lin
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mingkuan Sun
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
12
|
Wang HD, Lv CL, Feng L, Guo JX, Zhao SY, Jiang P. The role of autophagy in brain health and disease: Insights into exosome and autophagy interactions. Heliyon 2024; 10:e38959. [PMID: 39524893 PMCID: PMC11546156 DOI: 10.1016/j.heliyon.2024.e38959] [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: 09/21/2023] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Effective management of cellular components is essential for maintaining brain health, and studies have identified several crucial biological processes in the brain. Among these, autophagy and the role of exosomes in cellular communication are critical for brain health and disease. The interaction between autophagy and exosomes in the nervous system, as well as their contributions to brain damage, have garnered significant attention. This review summarizes that exosomes and their cargoes have been implicated in the autophagy process in the pathophysiology of nervous system diseases. Furthermore, the onset and progression of neurological disorders may be affected by autophagy regulation of the secretion and release of exosomes. These findings may provide new insights into the potential mechanism by which autophagy mediates different exosome secretion and release, as well as the valuable biomedical applications of exosomes in the prevention and treatment of various brain diseases by targeting autophagy.
Collapse
Affiliation(s)
- Hai-Dong Wang
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/Nanjing Medical University Kangda College First Affiliated Hospital/The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Chao-Liang Lv
- Department of Spine Surgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Lei Feng
- Department of Neurosurgery, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Jin-Xiu Guo
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Shi-Yuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| |
Collapse
|
13
|
Ghosh M, Pearse DD. The Yin and Yang of Microglia-Derived Extracellular Vesicles in CNS Injury and Diseases. Cells 2024; 13:1834. [PMID: 39594583 PMCID: PMC11592485 DOI: 10.3390/cells13221834] [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: 09/26/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in maintaining neural homeostasis but can also contribute to disease and injury when this state is disrupted or conversely play a pivotal role in neurorepair. One way that microglia exert their effects is through the secretion of small vesicles, microglia-derived exosomes (MGEVs). Exosomes facilitate intercellular communication through transported cargoes of proteins, lipids, RNA, and other bioactive molecules that can alter the behavior of the cells that internalize them. Under normal physiological conditions, MGEVs are essential to homeostasis, whereas the dysregulation of their production and/or alterations in their cargoes have been implicated in the pathogenesis of numerous neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), spinal cord injury (SCI), and traumatic brain injury (TBI). In contrast, MGEVs may also offer therapeutic potential by reversing inflammation or being amenable to engineering for the delivery of beneficial biologics or drugs. The effects of MGEVs are determined by the phenotypic state of the parent microglia. Exosomes from anti-inflammatory or pro-regenerative microglia support neurorepair and cell survival by delivering neurotrophic factors, anti-inflammatory mediators, and molecular chaperones. Further, MGEVs can also deliver components like mitochondrial DNA (mtDNA) and proteins to damaged neurons to enhance cellular metabolism and resilience. MGEVs derived from pro-inflammatory microglia can have detrimental effects on neural health. Their cargo often contains pro-inflammatory cytokines, molecules involved in oxidative stress, and neurotoxic proteins, which can exacerbate neuroinflammation, contribute to neuronal damage, and impair synaptic function, hindering neurorepair processes. The role of MGEVs in neurodegeneration and injury-whether beneficial or harmful-largely depends on how they modulate inflammation through the pro- and anti-inflammatory factors in their cargo, including cytokines and microRNAs. In addition, through the propagation of pathological proteins, such as amyloid-beta and alpha-synuclein, MGEVs can also contribute to disease progression in disorders such as AD and PD, or by the transfer of apoptotic or necrotic factors, they can induce neuron toxicity or trigger glial scarring during neurological injury. In this review, we have provided a comprehensive and up-to-date understanding of the molecular mechanisms underlying the multifaceted role of MGEVs in neurological injury and disease. In particular, the role that specific exosome cargoes play in various pathological conditions, either in disease progression or recovery, will be discussed. The therapeutic potential of MGEVs has been highlighted including potential engineering methodologies that have been employed to alter their cargoes or cell-selective targeting. Understanding the factors that influence the balance between beneficial and detrimental exosome signaling in the CNS is crucial for developing new therapeutic strategies for neurodegenerative diseases and neurotrauma.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
14
|
Feng L, Li B, Yong SS, Wen X, Tian Z. The emerging role of exercise in Alzheimer's disease: Focus on mitochondrial function. Ageing Res Rev 2024; 101:102486. [PMID: 39243893 DOI: 10.1016/j.arr.2024.102486] [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: 07/18/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory impairment and cognitive dysfunction, which eventually leads to the disability and mortality of older adults. Although the precise mechanisms by which age promotes the development of AD remains poorly understood, mitochondrial dysfunction plays a central role in the development of AD. Currently, there is no effective treatment for this debilitating disease. It is well accepted that exercise exerts neuroprotective effects by ameliorating mitochondrial dysfunction in the neurons of AD, which involves multiple mechanisms, including mitochondrial dynamics, biogenesis, mitophagy, transport, and signal transduction. In addition, exercise promotes mitochondria communication with other organelles in AD neurons, which should receive more attentions in the future.
Collapse
Affiliation(s)
- Lili Feng
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Bowen Li
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Su Sean Yong
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Xu Wen
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
15
|
Zhu L, Ma L, Du X, Jiang Y, Gao J, Fan Z, Zheng H, Zhu J, Zhang G. M2 Microglia-Derived Exosomes Protect Against Glutamate-Induced HT22 Cell Injury via Exosomal miR-124-3p. Mol Neurobiol 2024; 61:7845-7861. [PMID: 38433165 DOI: 10.1007/s12035-024-04075-x] [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: 11/08/2023] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
As one of the most serious complications of sepsis, sepsis-associated encephalopathy has not been effectively treated or prevented. Exosomes, as a new therapeutic method, play a protective role in neurodegenerative diseases, stroke and traumatic brain injury in recent years. The purpose of this study was to investigate the role of exosomes in glutamate (Glu)-induced neuronal injury, and to explore its mechanism, providing new ideas for the treatment of sepsis-associated encephalopathy. The neuron damage model induced by Glu was established, and its metabolomics was analyzed and identified. BV2 cells were induced to differentiate into M1 and M2 subtypes. After the exosomes from both M1-BV2 cells and M2-BV2 cells were collected, exosome morphological identification was performed by transmission electron microscopy and exosome-specific markers were also detected. These exosomes were then cocultured with HT22 cells. CCK-8 method and LDH kit were used to detect cell viability and toxicity. Cell apoptosis, mitochondrial membrane potential and ROS content were respectively detected by flow cytometry, JC-1 assay and DCFH-DA assay. MiR-124-3p expression level was detected by qRT-PCR and Western blot. Bioinformatics analysis and luciferase reporter assay predicted and verified the relationship between miR-124-3p and ROCK1 or ROCK2. Through metabolomics, 81 different metabolites were found, including fructose, GABA, 2, 4-diaminobutyric acid, etc. The enrichment analysis of differential metabolites showed that they were mainly enriched in glutathione metabolism, glycine and serine metabolism, and urea cycle. M2 microglia-derived exosomes could reduce the apoptosis, decrease the accumulation of ROS, restore the mitochondrial membrane potential and the anti-oxidative stress ability in HT22 cells induced by Glu. It was also found that the protective effect of miR-124-3p mimic on neurons was comparable to that of M2-EXOs. Additionally, M2-EXOs might carry miR-124-3p to target ROCK1 and ROCK2 in neurons, affecting ROCK/PTEN/AKT/mTOR signaling pathway, and then reducing Glu-induced neuronal apoptosis. M2 microglia-derived exosomes may protect HT22 cells against Glu-induced injury by transferring miR-124-3p into HT22 cells, with ROCK being a target gene for miR-124-3p.
Collapse
Affiliation(s)
- Lan Zhu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Limei Ma
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Xin Du
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Yuhao Jiang
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Jiake Gao
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Zihao Fan
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Hengheng Zheng
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Jianjun Zhu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China.
| | - Gaofeng Zhang
- Department of Critical Care Medicine, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No.6 Huanghe Road, Changshu, Jiangsu, 215500, People's Republic of China.
| |
Collapse
|
16
|
Fang X, Zhou D, Wang X, Ma Y, Zhong G, Jing S, Huang S, Wang Q. Exosomes: A Cellular Communication Medium That Has Multiple Effects On Brain Diseases. Mol Neurobiol 2024; 61:6864-6892. [PMID: 38356095 DOI: 10.1007/s12035-024-03957-4] [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: 09/18/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Exosomes, as membranous vesicles generated by multiple cell types and secreted to extracellular space, play a crucial role in a range of brain injury-related brain disorders by transporting diverse proteins, RNA, DNA fragments, and other functional substances. The nervous system's pathogenic mechanisms are complicated, involving pathological processes like as inflammation, apoptosis, oxidative stress, and autophagy, all of which result in blood-brain barrier damage, cognitive impairment, and even loss of normal motor function. Exosomes have been linked to the incidence and progression of brain disorders in recent research. As a result, a thorough knowledge of the interaction between exosomes and brain diseases may lead to the development of more effective therapeutic techniques that may be implemented in the clinic. The potential role of exosomes in brain diseases and the crosstalk between exosomes and other pathogenic processes were discussed in this paper. Simultaneously, we noted the delicate events in which exosomes as a media allow the brain to communicate with other tissues and organs in physiology and disease, and compiled a list of natural compounds that modulate exosomes, in order to further improve our understanding of exosomes and propose new ideas for treating brain disorders.
Collapse
Affiliation(s)
- Xiaoling Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Dishu Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Xinyue Wang
- Department of Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510405, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 510405, Guangzhou, China
| | - Yujie Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Shangwen Jing
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Shuiqing Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China.
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China.
| |
Collapse
|
17
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
18
|
Kong E, Geng X, Wu F, Yue W, Sun Y, Feng X. Microglial exosome miR-124-3p in hippocampus alleviates cognitive impairment induced by postoperative pain in elderly mice. J Cell Mol Med 2024; 28:e18090. [PMID: 38140846 PMCID: PMC10844686 DOI: 10.1111/jcmm.18090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/14/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Cognitive impairment induced by postoperative pain severely deteriorates the rehabilitation outcomes in elderly patients. The present study focused on the relationship between microglial exosome miR-124-3p in hippocampus and cognitive impairment induced by postoperative pain. Cognitive impairment model induced by postoperative pain was constructed by intramedullary nail fixation after tibial fracture. Morphine intraperitoneally was carried out for postoperative analgesia. Morris water maze tests were carried out to evaluate the cognitive impairment, while mRNA levels of neurotrophic factors (BDNF, NG) and neurodegenerative biomarker (VILIP-1) in hippocampus were tested by q-PCR. Transmission electron microscope was used to observe the axon degeneration in hippocampus. The levels of pro-inflammatory factors (TNF-α, IL-1β, IL-6), the levels of anti-inflammatory factors (Ym, Arg-1, IL-10) and microglia proliferation marker cyclin D1 in hippocampus were measured to evaluate microglia polarization. Bioinformatics analysis was conducted to identify key exosomes while BV-2 microglia overexpressing exosome miR-124-3p was constructed to observe microglia polarization in vitro experiments. Exogenous miR-124-3p-loaded exosomes were injected into hippocampus in vivo. Postoperative pain induced by intramedullary fixation after tibial fracture was confirmed by decreased mechanical and thermal pain thresholds. Postoperative pain induced cognitive impairment, promoted axon demyelination, decreased BDNF, NG and increased VILIP-1 expressions in hippocampus. Postoperative pain also increased pro-inflammatory factors, cyclin D1 and decreased anti-inflammatory factors in hippocampus. However, these changes were all reversed by morphine analgesia. Bioinformatics analysis identified the critical role of exosome miR-124-3p in cognitive impairment, which was confirmed to be down-regulated in hippocampus of postoperative pain mice. BV-2 microglia overexpressing exosome miR-124-3p showed decreased pro-inflammatory factors, cyclin D1 and increased anti-inflammatory factors. In vivo, stereotactic injection of exogenous miR-124-3p into hippocampus decreased pro-inflammatory factors, cyclin D1 and increased anti-inflammatory factors. The cognitive impairment, axon demyelination, decreased BDNF, NG and increased VILIP-1 expressions in hippocampus were all alleviated by exogenous exosome miR-124-3p. Microglial exosome miR-124-3p in hippocampus alleviates cognitive impairment induced by postoperative pain through microglia polarization in elderly mice.
Collapse
Affiliation(s)
- Erliang Kong
- Department of AnesthesiologyThe 988th Hospital of Joint Logistic Support Force of Chinese People's Liberation ArmyZhengzhouChina
| | - Xuqiang Geng
- Department of Rheumatology and Immunology, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Feixiang Wu
- Department of Intensive Care Unit, Shanghai Eastern Hepatobiliary Surgery HospitalThird Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Wei Yue
- Department of AnesthesiologyThe 988th Hospital of Joint Logistic Support Force of Chinese People's Liberation ArmyZhengzhouChina
| | - Yuming Sun
- Department of Anesthesiology, Shanghai Eastern Hepatobiliary Surgery HospitalThird Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Xudong Feng
- Department of AnesthesiologyThe 988th Hospital of Joint Logistic Support Force of Chinese People's Liberation ArmyZhengzhouChina
| |
Collapse
|
19
|
Wang Y, Yang Y, Song Y. Cardioprotective Effects of Exercise: The Role of Irisin and Exosome. Curr Vasc Pharmacol 2024; 22:316-334. [PMID: 38808716 DOI: 10.2174/0115701611285736240516101803] [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: 11/03/2023] [Revised: 01/14/2024] [Accepted: 04/16/2024] [Indexed: 05/30/2024]
Abstract
Exercise is an effective measure for preventing and treating cardiovascular diseases, although the exact molecular mechanism remains unknown. Previous studies have shown that both irisin and exosomes can improve the course of cardiovascular disease independently. Therefore, it is speculated that the cardiovascular protective effect of exercise is also related to its ability to regulate the concentrations of irisin and exosomes in the circulatory system. In this review, the potential synergistic interactions between irisin and exosomes are examined, as well as the underlying mechanisms including the AMPK/PI3K/AKT pathway, the TGFβ1/Smad2/3 pathway, the PI3K/AKT/VEGF pathway, and the PTEN/PINK1/Parkin pathway are examined. This paper provides evidence to propose that exercise promotes the release of exosomes enriched with irisin, miR-486-5p and miR-342-5p from skeletal muscles, which results in the activation protective networks in the cardiovascular system. Moreover, the potential synergistic effect in exosomal cargo can provide new ideas for clinical research of exercise mimics.
Collapse
Affiliation(s)
- Yuehuan Wang
- Graduate School, Wuhan Sports University, Wuhan, 430079, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, 430079, China
| | - Yi Yang
- Fitness Monitoring and Chronic Disease Intervention research center, Wuhan Sports University, Wuhan, 430079, China
- Hubei Key Laboratory of Exercise Training and Monitoring, Wuhan Sports University, Wuhan, 430079, China
| | - Yanjuan Song
- Graduate School, Wuhan Sports University, Wuhan, 430079, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, 430079, China
| |
Collapse
|
20
|
Wang D, Zheng J, Sun X, Xie L, Yang Y. Study on the Pharmacological Mechanism of Icariin for the Treatment of Alzheimer's Disease Based on Network Pharmacology and Molecular Docking Techniques. Metabolites 2023; 14:1. [PMID: 38276291 PMCID: PMC10820555 DOI: 10.3390/metabo14010001] [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: 11/23/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
The purpose of this study is to explore the pharmacological mechanism of icariin (ICA) in the treatment of Alzheimer's disease (AD) based on network pharmacology and network molecular docking technology. In order to investigate the regulatory effect of ICA on the expression level of AD pathological phosphorylation regulatory proteins, this study further explored the possible molecular mechanism of ICA regulating AD autophagy through network pharmacology. Macromolecular docking network was verified by Autodock Vina 1.1.2 software. The main active ingredients of ICA, the physicochemical properties, and pharmacokinetic information of ICA were predicted using online databases and relevant information. The results showed that the targets of MAPK3, AKT1, HSP90AA1, ESR1, and HSP90AA1 were more critical in the treatment of AD. Autophagy, apoptosis, senescence factors, phosphatidylinositide 3-kinase/protein kinase B (P13K/AKT) signaling pathway, MAKP, mTOR, and other pathways were significantly associated with AD. Docking of ICA with HIF-1, BNIP3, PINK1, and Parkin pathway molecules showed that the key targets of the signaling pathway were more stably bound to ICA, which may provide a better pathway for ICA to regulate autophagy by providing a better pathway. ICA can improve AD, and its mechanism may be related to the P13K/AKT, MAKP, and mTOR signaling pathways, thereby regulating autophagy-related proteins.
Collapse
Affiliation(s)
- Dongwei Wang
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Jilong Zheng
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Xingsheng Sun
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Liuwei Xie
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| | - Yang Yang
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| |
Collapse
|
21
|
Guo Y, Zhang L. Ghrelin inhibits NLRP3 inflammasome activation by upregulating autophagy to improve Alzheimer's disease. In Vitro Cell Dev Biol Anim 2023; 59:665-673. [PMID: 37989934 DOI: 10.1007/s11626-023-00818-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/21/2023] [Indexed: 11/23/2023]
Abstract
Nod-like receptor protein 3 (NLRP3) inflammasome, autophagy, and the aggregation of β-amyloid (Aβ) are key factors in Alzheimer's disease (AD) development. Ghrelin has shown promise in providing neuroprotection for AD. However, the mechanism underlying ghrelin's ability to improve AD by modulating autophagy and the NLRP3 inflammasome requires further clarification. Primary hippocampus neurons and mice were stimulated with Aβ1-42 to create an in vitro and in vivo AD model, followed by ghrelin administration for intervention. Additionally, we subjected the cells to 3-methyladenine (3-MA) treatment. Neuron morphology, microtubule-associated protein 2 (MAP-2) expression, apoptosis, cytokine levels, and protein expression were measured using various techniques. The escape latency of mice was assessed using the Morris water maze (MWM) test, and histopathology of the hippocampus was determined using hematoxylin-eosin staining. At 1-100 nM, ghrelin increased neuron/synapse numbers and MAP-2 expression dose-dependently while blocking apoptosis in Aβ1-42-treated cells. Moreover, ghrelin reduced the expression of Aβ1-42, p-Tau/Tau, p62, NLRP3, ASC, and cleaved Caspase-1, while increasing the expression of LC3II/LC3I and Beclin1 in AD cells. Furthermore, ghrelin treatment also decreased the levels of Aβ1-42, IL-1β, and IL-18 in the cells. The effects of ghrelin were reversed by 3-MA. Our in vivo experiments provided further confirmation of the above effect of ghrelin on AD. Additionally, the injection of Aβ1-42 induced increased escape latency in mice and histopathological changes in hippocampal neurons. All of these abnormalities were significantly improved following administration of ghrelin. Ghrelin mitigated Aβ1-42-induced neurotoxicity and relieved neuronal damage by upregulating autophagy to inactivate NLRP3, thus showing promising potential in treating AD.
Collapse
Affiliation(s)
- Yaoxue Guo
- Department of Clinical Pharmacy, Donghe District, Baotou Central Hospital, 61 Huancheng Road, Baotou, 014040, Inner Mongolia, China
| | - Lixiang Zhang
- Department of Clinical Pharmacy, Donghe District, Baotou Central Hospital, 61 Huancheng Road, Baotou, 014040, Inner Mongolia, China.
| |
Collapse
|
22
|
Wang L, Wei X. Exosome-based crosstalk in glaucoma pathogenesis: a focus on oxidative stress and neuroinflammation. Front Immunol 2023; 14:1202704. [PMID: 37529047 PMCID: PMC10388248 DOI: 10.3389/fimmu.2023.1202704] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/03/2023] [Indexed: 08/03/2023] Open
Abstract
Exosomes are membrane-bound tiny particles that are released by all live cells that contain multiple signal molecules and extensively participate in numerous normal physical activities and pathologies. In glaucoma, the crucial role of exosome-based crosstalk has been primarily revealed in animal models and ex vivo cell studies in the recent decade. In the aqueous drainage system, exosomes derived from non-pigment ciliary epithelium act in an endocrine manner and specifically regulate the function of the trabecular meshwork to cope with persistent oxidative stress challenges. In the retina, a more complicated regulatory network among microglia, retinal neurons, retinal ganglial cells, retinal pigment epithelium, and other immune effector cells by exosomes are responsible for the elaborate modulation of tissue homeostasis under physical state and the widespread propagation of neuroinflammation and its consequent neurodegeneration in glaucoma pathogenesis. Accumulating evidence indicates that exosome-based crosstalk depends on numerous factors, including the specific cargos they carried (particularly micro RNA), concentration, size, and ionization potentials, which largely remain elusive. In this narrative review, we summarize the latest research focus of exosome-based crosstalk in glaucoma pathogenesis, the current research progress of exosome-based therapy for glaucoma and provide in-depth perspectives on its current research gap.
Collapse
Affiliation(s)
- Lixiang Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Department of Ophthalmology, ShangjinNanfu Hospital, Chengdu, China
| |
Collapse
|
23
|
He A, Wang M, Li X, Chen H, Lim K, Lu L, Zhang C. Role of Exosomes in the Pathogenesis and Theranostic of Alzheimer's Disease and Parkinson's Disease. Int J Mol Sci 2023; 24:11054. [PMID: 37446231 DOI: 10.3390/ijms241311054] [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: 05/22/2023] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative diseases (NDDs) threatening the lives of millions of people worldwide, including especially elderly people. Currently, due to the lack of a timely diagnosis and proper intervention strategy, AD and PD largely remain incurable. Innovative diagnosis and therapy are highly desired. Exosomes are small vesicles that are present in various bodily fluids, which contain proteins, nucleic acids, and active biomolecules, and which play a crucial role especially in intercellular communication. In recent years, the role of exosomes in the pathogenesis, early diagnosis, and treatment of diseases has attracted ascending attention. However, the exact role of exosomes in the pathogenesis and theragnostic of AD and PD has not been fully illustrated. In the present review, we first introduce the biogenesis, components, uptake, and function of exosomes. Then we elaborate on the involvement of exosomes in the pathogenesis of AD and PD. Moreover, the application of exosomes in the diagnosis and therapeutics of AD and PD is also summarized and discussed. Additionally, exosomes serving as drug carriers to deliver medications to the central nervous system are specifically addressed. The potential role of exosomes in AD and PD is explored, discussing their applications in diagnosis and treatment, as well as their current limitations. Given the limitation in the application of exosomes, we also propose future perspectives for better utilizing exosomes in NDDs. Hopefully, it would pave ways for expanding the biological applications of exosomes in fundamental research as well as theranostics of NDDs.
Collapse
Affiliation(s)
- Aojie He
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Meiling Wang
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Xiaowan Li
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Hong Chen
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Kahleong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore
| | - Li Lu
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, 56 Xinjiannan Road, Taiyuan 030001, China
| |
Collapse
|
24
|
Elsherbini A, Zhu Z, Quadri Z, Crivelli SM, Ren X, Vekaria HJ, Tripathi P, Zhang L, Zhi W, Bieberich E. Novel Isolation Method Reveals Sex-Specific Composition and Neurotoxicity of Small Extracellular Vesicles in a Mouse Model of Alzheimer's Disease. Cells 2023; 12:1623. [PMID: 37371093 PMCID: PMC10297289 DOI: 10.3390/cells12121623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
We developed a new method to isolate small extracellular vesicles (sEVs) from male and female wild-type and 5xFAD mouse brains to investigate the sex-specific functions of sEVs in Alzheimer's disease (AD). A mass spectrometric analysis revealed that sEVs contained proteins critical for EV formation and Aβ. ExoView analysis showed that female mice contained more GFAP and Aβ-labeled sEVs, suggesting that a larger proportion of sEVs from the female brain is derived from astrocytes and/or more likely to bind to Aβ. Moreover, sEVs from female brains had more acid sphingomyelinase (ASM) and ceramide, an enzyme and its sphingolipid product important for EV formation and Aβ binding to EVs, respectively. We confirmed the function of ASM in EV formation and Aβ binding using co-labeling and proximity ligation assays, showing that ASM inhibitors prevented complex formation between Aβ and ceramide in primary cultured astrocytes. Finally, our study demonstrated that sEVs from female 5xFAD mice were more neurotoxic than those from males, as determined by impaired mitochondrial function (Seahorse assays) and LDH cytotoxicity assays. Our study suggests that sex-specific sEVs are functionally distinct markers for AD and that ASM is a potential target for AD therapy.
Collapse
Affiliation(s)
- Ahmed Elsherbini
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Zhihui Zhu
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Zainuddin Quadri
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Simone M. Crivelli
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Xiaojia Ren
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY 40536, USA;
- Veterans Affairs Medical Center, Lexington, KY 40502, USA
| | - Priyanka Tripathi
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Liping Zhang
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
| | - Wenbo Zhi
- Department of Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, USA;
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA; (A.E.); (Z.Z.); (Z.Q.); (S.M.C.); (X.R.); (P.T.); (L.Z.)
- Veterans Affairs Medical Center, Lexington, KY 40502, USA
| |
Collapse
|
25
|
Zhang Y, Miao Y, Xiong X, Tan J, Han Z, Chen F, Lei P, Zhang Q. Microglial exosomes alleviate intermittent hypoxia-induced cognitive deficits by suppressing NLRP3 inflammasome. Biol Direct 2023; 18:29. [PMID: 37312196 DOI: 10.1186/s13062-023-00387-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
Intermittent hypoxia is the best predictor of developing cognitive decline and Alzheimer's disease progression in patients with obstructive sleep apnea. The nucleotide-binding oligomerization domain-like receptor 3 (NLRP3) inflammasome has been poorly studied as a regulator of neuroinflammation in cognitive impairment caused by intermittent hypoxia. As critical inflammatory cells, exosomes secreted by microglia have been found to affect the spread of pathologic proteins and neuropathology in neurodegenerative diseases. However, the effects of microglial exosomes on neuroinflammation and cognitive outcomes after intermittent hypoxia remain unclear. In this study, the role of miRNAs in microglial exosomes in improving cognitive deficits in mice exposed to intermittent hypoxia was investigated. We demonstrated that miR-146a-5p fluctuated over time in microglial exosomes of mice exposed to intermittent hypoxia for different periods of time, which could regulate neuronal NLRP3 inflammasome and neuroinflammation. In primary neurons, we found that miR-146a-5p regulated mitochondrial reactive oxygen species by targeting HIF1α, thus affecting the NLRP3 inflammasome and secretion of inflammatory factors. Similarly, further studies showed that inhibition of NLRP3 by administering overexpressed miR-146a-5p in microglial exosomes and MCC950 has improved neuroinflammation and cognitive dysfunction in mice after intermittent hypoxia. In conclusion, NLRP3 inflammasome may be a regulatory target for ameliorating cognitive impairment caused by intermittent hypoxia, and microglial exosomal miR-146a-5p may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yaodan Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yuyang Miao
- Tianjin Medical University, Tianjin, 300052, China
| | - Xiangyang Xiong
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
| | - Zhaoli Han
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China.
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, China.
| |
Collapse
|
26
|
Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther 2023; 8:239. [PMID: 37291105 PMCID: PMC10248351 DOI: 10.1038/s41392-023-01502-8] [Citation(s) in RCA: 429] [Impact Index Per Article: 214.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/26/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Aging is characterized by systemic chronic inflammation, which is accompanied by cellular senescence, immunosenescence, organ dysfunction, and age-related diseases. Given the multidimensional complexity of aging, there is an urgent need for a systematic organization of inflammaging through dimensionality reduction. Factors secreted by senescent cells, known as the senescence-associated secretory phenotype (SASP), promote chronic inflammation and can induce senescence in normal cells. At the same time, chronic inflammation accelerates the senescence of immune cells, resulting in weakened immune function and an inability to clear senescent cells and inflammatory factors, which creates a vicious cycle of inflammation and senescence. Persistently elevated inflammation levels in organs such as the bone marrow, liver, and lungs cannot be eliminated in time, leading to organ damage and aging-related diseases. Therefore, inflammation has been recognized as an endogenous factor in aging, and the elimination of inflammation could be a potential strategy for anti-aging. Here we discuss inflammaging at the molecular, cellular, organ, and disease levels, and review current aging models, the implications of cutting-edge single cell technologies, as well as anti-aging strategies. Since preventing and alleviating aging-related diseases and improving the overall quality of life are the ultimate goals of aging research, our review highlights the critical features and potential mechanisms of inflammation and aging, along with the latest developments and future directions in aging research, providing a theoretical foundation for novel and practical anti-aging strategies.
Collapse
Affiliation(s)
- Xia Li
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China
| | - Chentao Li
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Wanying Zhang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Yanan Wang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Pengxu Qian
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China.
| |
Collapse
|
27
|
Si Q, Wu L, Pang D, Jiang P. Exosomes in brain diseases: Pathogenesis and therapeutic targets. MedComm (Beijing) 2023; 4:e287. [PMID: 37313330 PMCID: PMC10258444 DOI: 10.1002/mco2.287] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 06/15/2023] Open
Abstract
Exosomes are extracellular vesicles with diameters of about 100 nm that are naturally secreted by cells into body fluids. They are derived from endosomes and are wrapped in lipid membranes. Exosomes are involved in intracellular metabolism and intercellular communication. They contain nucleic acids, proteins, lipids, and metabolites from the cell microenvironment and cytoplasm. The contents of exosomes can reflect their cells' origin and allow the observation of tissue changes and cell states under disease conditions. Naturally derived exosomes have specific biomolecules that act as the "fingerprint" of the parent cells, and the contents changed under pathological conditions can be used as biomarkers for disease diagnosis. Exosomes have low immunogenicity, are small in size, and can cross the blood-brain barrier. These characteristics make exosomes unique as engineering carriers. They can incorporate therapeutic drugs and achieve targeted drug delivery. Exosomes as carriers for targeted disease therapy are still in their infancy, but exosome engineering provides a new perspective for cell-free disease therapy. This review discussed exosomes and their relationship with the occurrence and treatment of some neuropsychiatric diseases. In addition, future applications of exosomes in the diagnosis and treatment of neuropsychiatric disorders were evaluated in this review.
Collapse
Affiliation(s)
- Qingying Si
- Department of EndocrinologyTengzhou Central People's HospitalTengzhouChina
| | - Linlin Wu
- Department of OncologyTengzhou Central People's HospitalTengzhouChina
| | - Deshui Pang
- Department of EndocrinologyTengzhou Central People's HospitalTengzhouChina
| | - Pei Jiang
- Translational Pharmaceutical LaboratoryJining First People's HospitalShandong First Medical UniversityJiningChina
- Institute of Translational PharmacyJining Medical Research AcademyJiningChina
| |
Collapse
|
28
|
Wang C, Zou Q, Pu Y, Cai Z, Tang Y. Berberine Rescues D-Ribose-Induced Alzheimer's Pathology via Promoting Mitophagy. Int J Mol Sci 2023; 24:ijms24065896. [PMID: 36982968 PMCID: PMC10055824 DOI: 10.3390/ijms24065896] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Mitochondrial dysfunction is considered an early event of Alzheimer disease (AD). D-ribose is a natural monosaccharide that exists in cells, especially in mitochondria, and can lead to cognitive dysfunction. However, the reason for this is unclear. Berberine (BBR) is an isoquinoline alkaloid that can target mitochondria and has great prospect in the treatment of AD. The methylation of PINK1 reinforces the burden of Alzheimer's pathology. This study explores the role of BBR and D-ribose in the mitophagy and cognitive function of AD related to DNA methylation. APP/PS1 mice and N2a cells were treated with D-ribose, BBR, and mitophagy inhibitor Mdivi-1 to observe their effects on mitochondrial morphology, mitophagy, neuron histology, AD pathology, animal behavior, and PINK1 methylation. The results showed that D-ribose induced mitochondrial dysfunction, mitophagy damage, and cognitive impairment. However, BBR inhibition of PINK1 promoter methylation can reverse the above effects caused by D-ribose, improve mitochondrial function, and restore mitophagy through the PINK1-Parkin pathway, thus reducing cognitive deficits and the burden of AD pathology. This experiment puts a new light on the mechanism of action of D-ribose in cognitive impairment and reveals new insights in the use of BBR for AD treatment.
Collapse
Affiliation(s)
- Chuanling Wang
- Department of Histology and Embryology, School of Basic Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing 400016, China
| | - Qian Zou
- Chongqing Key Laboratory of Neurodegenerative Diseases, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China
- Department of Neurology, Chongqing General Hospital, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China
| | - Yinshuang Pu
- Chongqing Key Laboratory of Neurodegenerative Diseases, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China
- Department of Neurology, Chongqing General Hospital, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China
| | - Zhiyou Cai
- Chongqing Key Laboratory of Neurodegenerative Diseases, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China
- Department of Neurology, Chongqing General Hospital, No. 118 Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing 400016, China
| |
Collapse
|
29
|
Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, Li J. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther 2022; 7:391. [PMID: 36522308 PMCID: PMC9755275 DOI: 10.1038/s41392-022-01251-0] [Citation(s) in RCA: 566] [Impact Index Per Article: 188.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 12/23/2022] Open
Abstract
Aging is a gradual and irreversible pathophysiological process. It presents with declines in tissue and cell functions and significant increases in the risks of various aging-related diseases, including neurodegenerative diseases, cardiovascular diseases, metabolic diseases, musculoskeletal diseases, and immune system diseases. Although the development of modern medicine has promoted human health and greatly extended life expectancy, with the aging of society, a variety of chronic diseases have gradually become the most important causes of disability and death in elderly individuals. Current research on aging focuses on elucidating how various endogenous and exogenous stresses (such as genomic instability, telomere dysfunction, epigenetic alterations, loss of proteostasis, compromise of autophagy, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, deregulated nutrient sensing) participate in the regulation of aging. Furthermore, thorough research on the pathogenesis of aging to identify interventions that promote health and longevity (such as caloric restriction, microbiota transplantation, and nutritional intervention) and clinical treatment methods for aging-related diseases (depletion of senescent cells, stem cell therapy, antioxidative and anti-inflammatory treatments, and hormone replacement therapy) could decrease the incidence and development of aging-related diseases and in turn promote healthy aging and longevity.
Collapse
Affiliation(s)
- Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Lin Dou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mingjing Yan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| |
Collapse
|
30
|
Grimaldo L, Sandoval A, Duran P, Gómez Flores-Ramos L, Felix R. The ubiquitin E3 ligase Parkin regulates neuronal Ca V1.3 channel functional expression. J Neurophysiol 2022; 128:1555-1564. [PMID: 36350063 DOI: 10.1152/jn.00287.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Neuronal L-type Ca2+ channels of the CaV1.3 subclass are transmembrane protein complexes that contribute to the pacemaker activity in the adult substantia nigra dopaminergic neurons. The altered function of these channels may play a role in the development and progress of neurodegenerative mechanisms implicated in Parkinson's disease (PD). Although L-type channel expression is precisely regulated, an increased functional expression has been observed in PD. Previously, we showed that Parkin, an E3 enzyme of the ubiquitin-proteasome system (UPS) interacts with neuronal CaV2.2 channels promoting their ubiquitin-mediated degradation. In addition, previous studies show an increase in CaV1.3 channel activity in dopaminergic neurons of the SNc and that Parkin expression is reduced in PD. These findings suggest that the decrease in Parkin may affect the proteasomal degradation of CaV1.3, which helps explain the increase in channel activity. Therefore, the present report aims to gain insight into the degradation mechanisms of the neuronal CaV1.3 channel by the UPS. Immunoprecipitation assays showed the interaction between Parkin and the CaV1.3 channels expressed in HEK-293 cells and neural tissues. Likewise, Parkin overexpression reduced the total and membrane channel levels and decreased the current density. Consistent with this, patch-clamp recordings in the presence of an inhibitor of the UPS, MG132, prevented the effects of Parkin, suggesting enhanced channel proteasomal degradation. In addition, the half-life of the pore-forming CaV1.3α1 protein was significantly reduced by Parkin overexpression. Finally, electrophysiological recordings using a PRKN knockout HEK-293 cell line generated by CRISPR/Cas9 showed increased current density. These results suggest that Parkin promotes the proteasomal degradation of CaV1.3, which may be a relevant aspect for the pathophysiology of PD.NEW & NOTEWORTHY The increased expression of CaV1.3 calcium channels is a crucial feature of Parkinson's disease (PD) pathophysiology. However, the mechanisms that determine this increase are not yet defined. Parkin, an enzyme of the ubiquitin-proteasome system, is known to interact with neuronal channels promoting their ubiquitin-mediated degradation. Interestingly, Parkin mutations also play a role in PD. Here, the degradation mechanisms of CaV1.3 channels and their relationship with the pathophysiology of PD are studied in detail.
Collapse
Affiliation(s)
- Lizbeth Grimaldo
- Conacyt, Population Health Research Center, National Institute of Public Health, Cuernavaca, Mexico
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of Mexico (UNAM), Tlalnepantla, Mexico
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
| | | | - Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico
| |
Collapse
|