1
|
Dong J, Tong W, Liu M, Liu M, Liu J, Jin X, Chen J, Jia H, Gao M, Wei M, Duan Y, Zhong X. Endosomal traffic disorders: a driving force behind neurodegenerative diseases. Transl Neurodegener 2024; 13:66. [PMID: 39716330 DOI: 10.1186/s40035-024-00460-7] [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: 05/15/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024] Open
Abstract
Endosomes are crucial sites for intracellular material sorting and transportation. Endosomal transport is a critical process involved in the selective uptake, processing, and intracellular transport of substances. The equilibrium between endocytosis and circulation mediated by the endosome-centered transport pathway plays a significant role in cell homeostasis, signal transduction, and immune response. In recent years, there have been hints linking endosomal transport abnormalities to neurodegenerative diseases, including Alzheimer's disease. Nonetheless, the related mechanisms remain unclear. Here, we provide an overview of endosomal-centered transport pathways and highlight potential physiological processes regulated by these pathways, with a particular focus on the correlation of endosomal trafficking disorders with common pathological features of neurodegenerative diseases. Additionally, we summarize potential therapeutic agents targeting endosomal trafficking for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jianru Dong
- School of Pharmacy, China Medical University, Shenyang, 110122, China
- Weifang Hospital of Traditional Chinese Medicine, Weifang, 261000, China
| | - Weiwei Tong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, 110069, China
| | - Mingyan Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Mengyu Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Jinyue Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ju Chen
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Huachao Jia
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Menglin Gao
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, 110167, China.
| | - Ying Duan
- Liaoning Maternal and Child Health Hospital, Shenyang, 110005, China.
| | - Xin Zhong
- School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
2
|
Gautier MK, Kelley CM, Lee SH, Mufson EJ, Ginsberg SD. Maternal choline supplementation rescues early endosome pathology in basal forebrain cholinergic neurons in the Ts65Dn mouse model of Down syndrome and Alzheimer's disease. Neurobiol Aging 2024; 144:30-42. [PMID: 39265450 PMCID: PMC11490376 DOI: 10.1016/j.neurobiolaging.2024.09.002] [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: 01/17/2024] [Revised: 07/27/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
Individuals with DS develop Alzheimer's disease (AD) neuropathology, including endosomal-lysosomal system abnormalities and degeneration of basal forebrain cholinergic neurons (BFCNs). We investigated whether maternal choline supplementation (MCS) affects early endosome pathology within BFCNs using the Ts65Dn mouse model of DS/AD. Ts65Dn and disomic (2N) offspring from dams administered MCS were analyzed for endosomal pathology at 3-4 months or 10-12 months. Morphometric analysis of early endosome phenotype was performed on individual BFCNs using Imaris. The effects of MCS on the endosomal interactome were interrogated by relative co-expression (RCE) analysis. MCS effectively reduced age- and genotype-associated increases in early endosome number in Ts65Dn and 2N offspring, and prevented increases in early endosome size in Ts65Dn offspring. RCE revealed a loss of interactome cooperativity among endosome genes in Ts65Dn offspring that was restored by MCS. These findings demonstrate MCS rescues early endosome pathology, a driver of septohippocampal circuit dysfunction. The genotype-independent benefits of MCS on endosomal phenotype indicate translational applicability as an early-life therapy for DS as well as other neurodevelopmental/neurodegenerative disorders involving endosomal pathology.
Collapse
Affiliation(s)
- Megan K Gautier
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Pathobiology and Translational Medicine Program, New York University Grossman School of Medicine, New York, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Christy M Kelley
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, AZ, USA; Institute for Future Health, Scottsdale, AZ, USA
| | - Sang Han Lee
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Elliott J Mufson
- Departments of Translational Neuroscience and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Wang W, Rui M. Advances in understanding the roles of actin scaffolding and membrane trafficking in dendrite development. J Genet Genomics 2024; 51:1151-1161. [PMID: 38925347 DOI: 10.1016/j.jgg.2024.06.010] [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: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Dendritic morphology is typically highly branched, and the branching and synaptic abundance of dendrites can enhance the receptive range of neurons and the diversity of information received, thus providing the basis for information processing in the nervous system. Once dendritic development is aberrantly compromised or damaged, it may lead to abnormal connectivity of the neural network, affecting the function and stability of the nervous system and ultimately triggering a series of neurological disorders. Research on the regulation of dendritic developmental processes has flourished, and much progress is now being made in its regulatory mechanisms. Noteworthily, dendrites are characterized by an extremely complex dendritic arborization that cannot be attributed to individual protein functions alone, requiring a systematic analysis of the intrinsic and extrinsic signals and the coordinated roles among them. Actin cytoskeleton organization and membrane vesicle trafficking are required during dendrite development, with actin providing tracks for vesicles and vesicle trafficking in turn providing material for actin assembly. In this review, we focus on these two basic biological processes and discuss the molecular mechanisms and their synergistic effects underlying the morphogenesis of neuronal dendrites. We also offer insights and discuss strategies for the potential preventive and therapeutic treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wanting Wang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China
| | - Menglong Rui
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China.
| |
Collapse
|
4
|
Kim SM, Quagraine Y, Singh M, Kim JH. Rab11 suppresses neuronal stress signaling by localizing dual leucine zipper kinase to axon terminals for protein turnover. eLife 2024; 13:RP96592. [PMID: 39475475 PMCID: PMC11524585 DOI: 10.7554/elife.96592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Dual leucine zipper kinase (DLK) mediates multiple neuronal stress responses, and its expression levels are constantly suppressed to prevent excessive stress signaling. We found that Wallenda (Wnd), the Drosophila ortholog of DLK, is highly enriched in the axon terminals of Drosophila sensory neurons in vivo and that this subcellular localization is necessary for Highwire-mediated Wnd protein turnover under normal conditions. Our structure-function analysis found that Wnd palmitoylation is essential for its axon terminal localization. Palmitoylation-defective Wnd accumulated in neuronal cell bodies, exhibited dramatically increased protein expression levels, and triggered excessive neuronal stress responses. Defective intracellular transport is implicated in neurodegenerative conditions. Comprehensive dominant-negative Rab protein screening identified Rab11 as an essential factor for Wnd localization in axon terminals. Consequently, Rab11 loss-of-function increased the protein levels of Wnd and induced neuronal stress responses. Inhibiting Wnd activity significantly ameliorated neuronal loss and c-Jun N-terminal kinase signaling triggered by Rab11 loss-of-function. Taken together, these suggest that DLK proteins are constantly transported to axon terminals for protein turnover and a failure of such transport can lead to neuronal loss. Our study demonstrates how subcellular protein localization is coupled to protein turnover for neuronal stress signaling.
Collapse
Affiliation(s)
- Seung Mi Kim
- Department of Biology, University of Nevada RenoRenoUnited States
| | - Yaw Quagraine
- Department of Biology, University of Nevada RenoRenoUnited States
| | - Monika Singh
- Department of Biology, University of Nevada RenoRenoUnited States
| | - Jung Hwan Kim
- Department of Biology, University of Nevada RenoRenoUnited States
| |
Collapse
|
5
|
Dwivedi SD, Shukla R, Yadav K, Rathor LS, Singh D, Singh MR. Mechanistic insight on the role of iRhom2-TNF-α-BAFF signaling pathway in various autoimmune disorders. Adv Biol Regul 2024; 92:101011. [PMID: 38151421 DOI: 10.1016/j.jbior.2023.101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/23/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
iRhom2 is a crucial cofactor involved in upregulation of TNF receptors (TNFRs) and the pro-inflammatory cytokine tumor necrosis factor (TNF-) from the cell surface by ADAM17. Tumor necrosis factor- α converting enzyme (TACE) is another name given to ADAM17. Many membrane attached biologically active molecules are cleaved by this enzyme which includes TNFRs and the pro-inflammatory cytokine tumor necrosis factor- α. The TNF receptors are of two types TNFR1 and TNFR2. iRhom2 belongs to the pseudo-protease class of rhomboid family, its abundance is observed in the immune cells. Biological activity of ADAM17 is affected in multiple levels by the iRhom2. ADAM17 is trafficked into the Golgi apparatus by the action of iRhom2, where it gets matured proteolytically and is stimulated to perform its function on the cell surface. This process of activation of ADAM17 results in the protection of the organism from the cascade of inflammatory reactions, as this activation blocks the TNF- α mediated secretion responsible for inflammatory responses produced. Present paper illustrates about the iRhom2-TNF-α-BAFF signaling pathway and its correlation with several autoimmune disorders such as Rheumatoid Arthritis, Systemic Lupus Erythematosus, Hemophilia Arthropathy, Alzheimer's disease and Tylosis with esophageal cancer etc.
Collapse
Affiliation(s)
- Shradha Devi Dwivedi
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur (C.G), 492010, India
| | - Rashi Shukla
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur (C.G), 492010, India
| | - Krishna Yadav
- Raipur Institute of Pharmaceutical Educations and Research, Sarona, Raipur, Chhattisgarh, 492010, India
| | - Lokendra Singh Rathor
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur (C.G), 492010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur (C.G), 492010, India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur (C.G), 492010, India.
| |
Collapse
|
6
|
Yao H, Shen Y, Song Z, Han A, Chen X, Zhang Y, Hu B. Rab11 promotes single Mauthner cell axon regeneration in vivo through axon guidance molecule Ntng2b. Exp Neurol 2024; 374:114715. [PMID: 38325655 DOI: 10.1016/j.expneurol.2024.114715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/23/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
Effective axon regeneration within the central nervous system (CNS) is pivotal for achieving functional recovery following spinal cord injury (SCI). Numerous extrinsic and intrinsic factors exert influences on the axon regeneration. While prior studies have demonstrated crucial involvement of specific members the Rab protein family in axon regeneration in the peripheral nervous system (PNS), the precise function of Rab11 in CNS axon regeneration in vivo remains elusive. Thus, our study aimed to elucidate the impact of Rab11 on the axon regeneration of Mauthner cells (M-cells) in zebrafish larvae. Our findings demonstrated that overexpression of Rab11bb via single-cell electroporation significantly promoted axon regeneration in individual M-cells. Conversely, knockdown of Rab11bb inhibited the axon regeneration of M-cells. RNA-seq analysis revealed an upregulation of ntng2b following Rab11bb overexpression. As we hypothesized, overexpression of Ntng2b markedly enhanced axon regeneration, while Ntng2b knockdown in the context of Rab11bb pro-regeneration substantially hindered axon regrowth. In conclusion, our study demonstrated that Rab11 promotes axon regeneration of single M-cell in the CNS through the Rab11/axon guidance/Ntng2b pathway.
Collapse
Affiliation(s)
- Huaitong Yao
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Yueru Shen
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Zheng Song
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Along Han
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Xinghan Chen
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Yawen Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Bing Hu
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
7
|
Todd TW, Shao W, Zhang YJ, Petrucelli L. The endolysosomal pathway and ALS/FTD. Trends Neurosci 2023; 46:1025-1041. [PMID: 37827960 PMCID: PMC10841821 DOI: 10.1016/j.tins.2023.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/23/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are considered to be part of a disease spectrum that is associated with causative mutations and risk variants in a wide range of genes. Mounting evidence indicates that several of these genes are linked to the endolysosomal system, highlighting the importance of this pathway in ALS/FTD. Although many studies have focused on how disruption of this pathway impacts on autophagy, recent findings reveal that this may not be the whole picture: specifically, disrupting autophagy may not be sufficient to induce disease, whereas disrupting the endolysosomal system could represent a crucial pathogenic driver. In this review we discuss the connections between ALS/FTD and the endolysosomal system, including a breakdown of how disease-associated genes are implicated in this pathway. We also explore the potential downstream consequences of disrupting endolysosomal activity in the brain, outside of an effect on autophagy.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Wei Shao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
8
|
Bertin F, Jara-Wilde J, Auer B, Köhler-Solís A, González-Silva C, Thomas U, Sierralta J. Drosophila Atlastin regulates synaptic vesicle mobilization independent of bone morphogenetic protein signaling. Biol Res 2023; 56:49. [PMID: 37710314 PMCID: PMC10503011 DOI: 10.1186/s40659-023-00462-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND The endoplasmic reticulum (ER) contacts endosomes in all parts of a motor neuron, including the axon and presynaptic terminal, to move structural proteins, proteins that send signals, and lipids over long distances. Atlastin (Atl), a large GTPase, is required for membrane fusion and the structural dynamics of the ER tubules. Atl mutations are the second most common cause of Hereditary Spastic Paraplegia (HSP), which causes spasticity in both sexes' lower extremities. Through an unknown mechanism, Atl mutations stimulate the BMP (bone morphogenetic protein) pathway in vertebrates and Drosophila. Synaptic defects are caused by atl mutations, which affect the abundance and distribution of synaptic vesicles (SV) in the bouton. We hypothesize that BMP signaling, does not cause Atl-dependent SV abnormalities in Drosophila. RESULTS We show that atl knockdown in motor neurons (Atl-KD) increases synaptic and satellite boutons in the same way that constitutively activating the BMP-receptor Tkv (thick veins) (Tkv-CA) increases the bouton number. The SV proteins Cysteine string protein (CSP) and glutamate vesicular transporter are reduced in Atl-KD and Tkv-CA larvae. Reducing the activity of the BMP receptor Wishful thinking (wit) can rescue both phenotypes. Unlike Tkv-CA larvae, Atl-KD larvae display altered activity-dependent distributions of CSP staining. Furthermore, Atl-KD larvae display an increased FM 1-43 unload than Control and Tkv-CA larvae. As decreasing wit function does not reduce the phenotype, our hypothesis that BMP signaling is not involved is supported. We also found that Rab11/CSP colocalization increased in Atl-KD larvae, which supports the concept that late recycling endosomes regulate SV movements. CONCLUSIONS Our findings reveal that Atl modulates neurotransmitter release in motor neurons via SV distribution independently of BMP signaling, which could explain the observed SV accumulation and synaptic dysfunction. Our data suggest that Atl is involved in membrane traffic as well as formation and/or recycling of the late endosome.
Collapse
Affiliation(s)
- Francisca Bertin
- Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jorge Jara-Wilde
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Computational Sciences, Faculty of Physical and Mathematical Sciences, Universidad de Chile, Santiago, Chile
| | - Benedikt Auer
- Laboratory of Neuronal and Synaptic Signals, Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Andrés Köhler-Solís
- Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Carolina González-Silva
- Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ulrich Thomas
- Functional Genetics of the Synapse, Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Jimena Sierralta
- Biomedical Neuroscience Institute (BNI), Santiago, Chile.
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
9
|
Meng T, Chen X, He Z, Huang H, Lin S, Liu K, Bai G, Liu H, Xu M, Zhuang H, Zhang Y, Waqas A, Liu Q, Zhang C, Sun XD, Huang H, Umair M, Yan Y, Feng D. ATP9A deficiency causes ADHD and aberrant endosomal recycling via modulating RAB5 and RAB11 activity. Mol Psychiatry 2023; 28:1219-1231. [PMID: 36604604 PMCID: PMC9816018 DOI: 10.1038/s41380-022-01940-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 12/10/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023]
Abstract
ATP9A, a lipid flippase of the class II P4-ATPases, is involved in cellular vesicle trafficking. Its homozygous variants are linked to neurodevelopmental disorders in humans. However, its physiological function, the underlying mechanism as well as its pathophysiological relevance in humans and animals are still largely unknown. Here, we report two independent families in which the nonsense mutations c.433C>T/c.658C>T/c.983G>A (p. Arg145*/p. Arg220*/p. Trp328*) in ATP9A (NM_006045.3) cause autosomal recessive hypotonia, intellectual disability (ID) and attention deficit hyperactivity disorder (ADHD). Atp9a null mice show decreased muscle strength, memory deficits and hyperkinetic movement disorder, recapitulating the symptoms observed in patients. Abnormal neurite morphology and impaired synaptic transmission are found in the primary motor cortex and hippocampus of the Atp9a null mice. ATP9A is also required for maintaining neuronal neurite morphology and the viability of neural cells in vitro. It mainly localizes to endosomes and plays a pivotal role in endosomal recycling pathway by modulating small GTPase RAB5 and RAB11 activation. However, ATP9A pathogenic mutants have aberrant subcellular localization and cause abnormal endosomal recycling. These findings provide strong evidence that ATP9A deficiency leads to neurodevelopmental disorders and synaptic dysfunctions in both humans and mice, and establishes novel regulatory roles for ATP9A in RAB5 and RAB11 activity-dependent endosomal recycling pathway and neurological diseases.
Collapse
Affiliation(s)
- Tian Meng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 511436, Guangzhou, China
| | - Xiaoting Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Zhengjie He
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, China
| | - Haofeng Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Shiyin Lin
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Kunru Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Guo Bai
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Hao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 511436, Guangzhou, China.,Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511500, China
| | - Mindong Xu
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haixia Zhuang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yunlong Zhang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ahmed Waqas
- Department of Zoology, Division of Science and Technology, University of Education, Lahore, 54000, Pakistan
| | - Qian Liu
- Department of Cerebrovascular Disease Center, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Chuan Zhang
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital; Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, 730050, China
| | - Xiang-Dong Sun
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Huansen Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGH), Riyadh, 11481, Saudi Arabia. .,Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, 22209, Pakistan.
| | - Yousheng Yan
- Prenatal Diagnostic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China.
| | - Du Feng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China. .,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, China. .,State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 511436, Guangzhou, China.
| |
Collapse
|
10
|
Magrì A, Lipari CLR, Risiglione P, Zimbone S, Guarino F, Caccamo A, Messina A. ERK1/2-dependent TSPO overactivation associates with the loss of mitophagy and mitochondrial respiration in ALS. Cell Death Dis 2023; 14:122. [PMID: 36792609 PMCID: PMC9931716 DOI: 10.1038/s41419-023-05643-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/17/2023]
Abstract
Mitochondrial dysfunction and the loss of mitophagy, aimed at recycling irreversibly damaged organelles, contribute to the onset of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease affecting spinal cord motor neurons. In this work, we showed that the reduction of mitochondrial respiration, exactly oxygen flows linked to ATP production and maximal capacity, correlates with the appearance of the most common ALS motor symptoms in a transgenic mouse model expressing SOD1 G93A mutant. This is the result of the equal inhibition in the respiration linked to complex I and II of the electron transport chain, but not their protein levels. Since the overall mitochondrial mass was unvaried, we investigated the expression of the Translocator Protein (TSPO), a small mitochondrial protein whose overexpression was recently linked to the loss of mitophagy in a model of Parkinson's disease. Here we clearly showed that levels of TSPO are significantly increased in ALS mice. Mechanistically, this increase is linked to the overactivation of ERK1/2 pathway and correlates with a decrease in the expression of the mitophagy-related marker Atg12, indicating the occurrence of impairments in the activation of mitophagy. Overall, our work sets out TSPO as a key regulator of mitochondrial homeostasis in ALS.
Collapse
Affiliation(s)
- Andrea Magrì
- grid.8158.40000 0004 1757 1969Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy ,we.MitoBiotech S.R.L., C.so Italia 172, Catania, Italy
| | - Cristiana Lucia Rita Lipari
- grid.8158.40000 0004 1757 1969Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Pierpaolo Risiglione
- grid.8158.40000 0004 1757 1969Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stefania Zimbone
- grid.5326.20000 0001 1940 4177Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Section of Catania, Catania, Italy
| | - Francesca Guarino
- we.MitoBiotech S.R.L., C.so Italia 172, Catania, Italy ,grid.8158.40000 0004 1757 1969Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonella Caccamo
- grid.8158.40000 0004 1757 1969Department of Drug and Health Sciences, University of Catania, Catania, Italy ,grid.10438.3e0000 0001 2178 8421Department of Chemical, Biological, Pharmaceutical Sciences, University of Messina, Messina, Italy
| | - Angela Messina
- Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy. .,we.MitoBiotech S.R.L., C.so Italia 172, Catania, Italy.
| |
Collapse
|
11
|
Abstract
Vesicles mediate the trafficking of membranes/proteins in the endocytic and secretory pathways. These pathways are regulated by small GTPases of the Rab family. Rab proteins belong to the Ras superfamily of GTPases, which are significantly involved in various intracellular trafficking and signaling processes in the nervous system. Rab11 is known to play a key role especially in recycling many proteins, including receptors important for signal transduction and preservation of functional activities of nerve cells. Rab11 activity is controlled by GEFs (guanine exchange factors) and GAPs (GTPase activating proteins), which regulate its function through modulating GTP/GDP exchange and the intrinsic GTPase activity, respectively. Rab11 is involved in the transport of several growth factor molecules important for the development and repair of neurons. Overexpression of Rab11 has been shown to significantly enhance vesicle trafficking. On the other hand, a reduced expression of Rab11 was observed in several neurodegenerative diseases. Current evidence appears to support the notion that Rab11 and its cognate proteins may be potential targets for therapeutic intervention. In this review, we briefly discuss the function of Rab11 and its related interaction partners in intracellular pathways that may be involved in neurodegenerative processes.
Collapse
Affiliation(s)
| | - Jiri Novotny
- Jiri Novotny, Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
12
|
Swaroop RS, Pradhan SS, Darshan VMD, Phalguna KS, Sivaramakrishnan V. Integrated network pharmacology approach shows a potential role of Ginseng catechins and ginsenosides in modulating protein aggregation in Amyotrophic Lateral Sclerosis. 3 Biotech 2022; 12:333. [PMID: 36330377 PMCID: PMC9622974 DOI: 10.1007/s13205-022-03401-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/12/2022] [Indexed: 11/29/2022] Open
Abstract
Amyotrophic lateral Sclerosis is an incurable, progressive neurodegenerative motor neuron disease. The disease is characterized by protein aggregates. The symptoms include weakness, denervation of muscles, atrophy and progressive paralysis of bulbar and respiratory muscles and dysphagia. Various secondary metabolites are evaluated for their ability to improve symptoms in ALS. Ginseng has been traditionally used for treating several neurodegenerative diseases. Several studies using model systems have shown a potential role of Ginseng catechins and Ginsenosides in clearing protein aggregation associated with ALS. We focus on Network pharmacology approach to understand the effect of Ginseng catechins or ginsenosides on protein aggregation associated with ALS. A catechin/ginsenoside-protein interaction network was generated and the pathways obtained were compared with those obtained from transcriptomic datasets of ALS from GEO database. Knock out of MAPK14, AKT and GSK from Catechin and BACE 1 from ginsenoside modulated pathways inhibited protein aggregation. Catechins and ginsenosides have potential as therapeutic agents in the management of ALS. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03401-1.
Collapse
Affiliation(s)
- R. Sai Swaroop
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - V. M. Datta Darshan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - Kanikaram Sai Phalguna
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| |
Collapse
|
13
|
The Rab11-regulated endocytic pathway and BDNF/TrkB signaling: Roles in plasticity changes and neurodegenerative diseases. Neurobiol Dis 2022; 171:105796. [PMID: 35728773 DOI: 10.1016/j.nbd.2022.105796] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/10/2022] [Accepted: 06/14/2022] [Indexed: 02/08/2023] Open
Abstract
Neurons are highly polarized cells that rely on the intracellular transport of organelles. This process is regulated by molecular motors such as dynein and kinesins and the Rab family of monomeric GTPases that together help move cargo along microtubules in dendrites, somas, and axons. Rab5-Rab11 GTPases regulate receptor trafficking along early-recycling endosomes, which is a process that determines the intracellular signaling output of different signaling pathways, including those triggered by BDNF binding to its tyrosine kinase receptor TrkB. BDNF is a well-recognized neurotrophic factor that regulates experience-dependent plasticity in different circuits in the brain. The internalization of the BDNF/TrkB complex results in signaling endosomes that allow local signaling in dendrites and presynaptic terminals, nuclear signaling in somas and dynein-mediated long-distance signaling from axons to cell bodies. In this review, we briefly discuss the organization of the endocytic pathway and how Rab11-recycling endosomes interact with other endomembrane systems. We further expand upon the roles of the Rab11-recycling pathway in neuronal plasticity. Then, we discuss the BDNF/TrkB signaling pathways and their functional relationships with the postendocytic trafficking of BDNF, including axonal transport, emphasizing the role of BDNF signaling endosomes, particularly Rab5-Rab11 endosomes, in neuronal plasticity. Finally, we discuss the evidence indicating that the dysfunction of the early-recycling pathway impairs BDNF signaling, contributing to several neurodegenerative diseases.
Collapse
|
14
|
Identification of let-7f and miR-338 as plasma-based biomarkers for sporadic amyotrophic lateral sclerosis using meta-analysis and empirical validation. Sci Rep 2022; 12:1373. [PMID: 35082326 PMCID: PMC8791978 DOI: 10.1038/s41598-022-05067-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022] Open
Abstract
AbstractAmyotrophic lateral sclerosis (ALS) is a lethal neurodegenerative disease that in most cases occurs sporadic (sALS). The disease is not curable, and its pathogenesis mechanisms are not well understood yet. Given the intricacy of underlying molecular interactions and heterogeneity of ALS, the discovery of molecules contributing to disease onset and progression will open a new avenue for advancement in early diagnosis and therapeutic intervention. Here we conducted a meta-analysis of 12 circulating miRNA profiling studies using the robust rank aggregation (RRA) method, followed by enrichment analysis and experimental verification. We identified miR-451a and let-7f-5p as meta-signature miRNAs whose targets are involved in critical pathogenic pathways underlying ALS, including ‘FoxO signaling pathway’, ‘MAPK signaling pathway’, and ‘apoptosis’. A systematic review of 7 circulating gene profiling studies elucidated that 241 genes up-regulated in sALS circulation with concomitant being targets of the meta-signature miRNAs. Protein–protein interaction (PPI) network analysis of the candidate targets using MCODE algorithm revealed the main subcluster is involved in multiple cascades eventually leads apoptosis, including ‘positive regulation of neuron apoptosis. Besides, we validated the meta-analysis results using RT-qPCR. Indeed, relative expression analysis verified let-7f-5p and miR-338-3p as significantly down-regulated and up-regulated biomarkers in the plasma of sALS patients, respectively. Receiver operating characteristic (ROC) analysis also highlighted the let-7f-5p and miR-338-3p potential as robustness plasma biomarkers for diagnosis and potential therapeutic targets of sALS disease.
Collapse
|
15
|
Schiel KA. A beneficial role for elevated extracellular glutamate in Amyotrophic Lateral Sclerosis and cerebral ischemia. Bioessays 2021; 43:e2100127. [PMID: 34585427 DOI: 10.1002/bies.202100127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/06/2022]
Abstract
This hypothesis proposes that increased extracellular glutamate in Amyotrophic Lateral Sclerosis (ALS) and cerebral ischemia, currently viewed as a trigger for excitotoxicity, is actually beneficial as it stimulates the utilization of glutamate as metabolic fuel. Renewed appreciation of glutamate oxidation by ischemic neurons has raised questions regarding the role of extracellular glutamate in ischemia. Is it detrimental, as suggested by excitotoxicity in early in vitro studies, or beneficial, as suggested by its oxidation in later in vivo studies? The answer may depend on the activity of N-methyl-D-aspartate (NMDA) glutamate receptors. Early in vitro procedures co-activated NMDA receptors (NMDARs) containing 2A (GluN2A) and 2B (GluN2B) subunits, an event now believed to trigger excitotoxicity; however, during in vivo ischemia D-serine and zinc molecules are released and these ensure only GluN2B receptors are stimulated. This not only prevents excitotoxicity but also initiates signaling cascades that allow ischemic neurons to import and oxidize glutamate.
Collapse
|
16
|
Cong C, Liang W, Zhang C, Wang Y, Yang Y, Wang X, Wang S, Huo D, Wang H, Wang D, Feng H. PAK4 suppresses motor neuron degeneration in hSOD1 G93A -linked amyotrophic lateral sclerosis cell and rat models. Cell Prolif 2021; 54:e13003. [PMID: 33615605 PMCID: PMC8016643 DOI: 10.1111/cpr.13003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/24/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons (MN). CREB pathway-mediated inhibition of apoptosis contributes to neuron protection, and PAK4 activates CREB signalling in diverse cell types. This study aimed to investigate PAK4's effect and mechanism of action in ALS. METHODS We analysed RNA levels by qRT-PCR, protein levels by immunofluorescence and Western blotting, and apoptosis by flow cytometry and TUNEL staining. Cell transfection was performed for in vitro experiment. Mice were injected intraspinally to evaluate PAK4 function in vivo experiment. Rotarod test was performed to measure motor function. RESULTS The expression and activation of PAK4 significantly decreased in the cell and mouse models of ALS as the disease progressed, which was caused by the negative regulation of miR-9-5p. Silencing of PAK4 increased the apoptosis of MN by inhibiting CREB-mediated neuroprotection, whereas overexpression of PAK4 protected MN from hSOD1G93A -induced degeneration by activating CREB signalling. The neuroprotective effect of PAK4 was markedly inhibited by CREB inhibitor. In ALS models, the PAK4/CREB pathway was inhibited, and cell apoptosis increased. In vivo experiments revealed that PAK4 overexpression in the spinal neurons of hSOD1G93A mice suppressed MN degeneration, prolonged survival and promoted the CREB pathway. CONCLUSIONS PAK4 protects MN from degeneration by activating the anti-apoptotic effects of CREB signalling, suggesting it may be a therapeutic target in ALS.
Collapse
Affiliation(s)
- Chaohua Cong
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Weiwei Liang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Chunting Zhang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Ying Wang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Yueqing Yang
- Department of Neurology, The Second Clinical College of Harbin Medical University, Harbin, China
| | - Xudong Wang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Shuyu Wang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Di Huo
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Hongyong Wang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Di Wang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| | - Honglin Feng
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Gautier MK, Ginsberg SD. A method for quantification of vesicular compartments within cells using 3D reconstructed confocal z-stacks: Comparison of ImageJ and Imaris to count early endosomes within basal forebrain cholinergic neurons. J Neurosci Methods 2021; 350:109038. [PMID: 33338543 PMCID: PMC8026492 DOI: 10.1016/j.jneumeth.2020.109038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Phenotypic changes in vesicular compartments are an early pathological hallmark of many peripheral and central diseases. For example, accurate assessment of early endosome pathology is crucial to the study of Down syndrome (DS) and Alzheimer's disease (AD), as well as other neurological disorders with endosomal-lysosomal pathology. NEW METHOD We describe a method for quantification of immunolabeled early endosomes within transmitter-identified basal forebrain cholinergic neurons (BFCNs) using 3-dimensional (3D) reconstructed confocal z-stacks employing Imaris software. RESULTS Quantification of 3D reconstructed z-stacks was performed using two different image analysis programs: ImageJ and Imaris. We found ImageJ consistently overcounted the number of early endosomes present within individual BFCNs. Difficulty separating densely packed early endosomes within defined BFCNs was observed in ImageJ compared to Imaris. COMPARISON WITH EXISTING METHODS Previous methods quantifying endosomal-lysosomal pathology relied on confocal microscopy images taken in a single plane of focus. Since early endosomes are distributed throughout the soma and neuronal processes of BFCNs, critical insight into the abnormal early endosome phenotype may be lost as a result of analyzing only a single image of the perikaryon. Rather than relying on a representative sampling, this protocol enables precise, direct quantification of all immunolabeled vesicles within a defined cell of interest. CONCLUSIONS Imaris is an ideal program for accurately counting punctate vesicles in the context of dual label confocal microscopy. Superior image resolution and detailed algorithms offered by Imaris make precise and rigorous quantification of individual early endosomes dispersed throughout a BFCN in 3D space readily achievable.
Collapse
Affiliation(s)
- Megan K Gautier
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Program of Pathobiology and Translational Medicine, Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York, NY, USA; NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA; NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
18
|
Zhang J, Su G, Wu Q, Liu J, Tian Y, Liu X, Zhou J, Gao J, Chen W, Chen D, Zhang Z. Rab11-mediated recycling endosome role in nervous system development and neurodegenerative diseases. Int J Neurosci 2020; 131:1012-1018. [PMID: 32329391 DOI: 10.1080/00207454.2020.1761354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
STUDY Membrane trafficking process is significant for the complex and precise regulatory of the nervous system. Rab11, as a small GTPase of the Rab superfamily, controls endocytic vesicular trafficking to the cell surface after sorting in recycling endosome (RE), coordinating with its receptors to maintain neurological function. MATERIALS AND METHODS This article reviewed the literature of Rab11 in nervous system. RESULTS Rab11-positive vesicles targeted transport growth-related molecules, such as integrins, protrudin, tropomyosin receptor kinase (Trk) A/B receptor and AMPA receptor (AMPAR) to membrane surface to promote the regeneration capacity of axon and dendrites and maintain synaptic plasticity. In addition, many studies have shown that the expression of Rab11 is decreased in multiple neurodegenerative diseases, which is highly correlated with the process of production, transport and clearance of disease-related pathological proteins. And overexpression or increased activity of Rab11 can greatly improve the defect of membrane trafficking and slow down the disease process. CONCLUSION With increasing research efforts on Rab11-dependent membrane trafficking mechanisms, a potential target for nerve regeneration and neurodegenerative diseases will be provided.
Collapse
Affiliation(s)
- Jiajia Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Qionghui Wu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jifei Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Ye Tian
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiaoyan Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Juanping Zhou
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Deyi Chen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zhenchang Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
19
|
Romano N, Catalani A, Lattante S, Belardo A, Proietti S, Bertini L, Silvestri F, Catalani E, Cervia D, Zolla L, Sabatelli M, Welshhans K, Ceci M. ALS skin fibroblasts reveal oxidative stress and ERK1/2-mediated cytoplasmic localization of TDP-43. Cell Signal 2020; 70:109591. [PMID: 32126264 DOI: 10.1016/j.cellsig.2020.109591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/14/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
The main hallmark of many forms of familiar and sporadic amyotrophic lateral sclerosis (ALS) is a reduction in nuclear TDP-43 protein and its inclusion in cytoplasmic aggregates in motor neurons. In order to understand which cellular and molecular mechanisms underlie the mislocalization of TDP-43, we examined human skin fibroblasts from two individuals with familial ALS, both with mutations in TDP-43, and two individuals with sporadic ALS, both without TDP-43 mutations or mutations in other ALS related genes. We found that all ALS fibroblasts had a partially cytoplasmic localization of TDP-43 and had reduced cell metabolism as compared to fibroblasts from apparently healthy individuals. ALS fibroblasts showed an increase in global protein synthesis and an increase in 4E-BP1 and rpS6 phosphorylation, which is indicative of mTORC1 activity. We also observed a decrease in glutathione (GSH), which suggests that oxidative stress is elevated in ALS. ERK1/2 activity regulated the extent of oxidative stress and the localization of TDP-43 in the cytoplasm in all ALS fibroblasts. Lastly, ALS fibroblasts showed reduced stress granule formation in response to H2O2 stress. In conclusion, these findings identify specific cellular and molecular defects in ALS fibroblasts, thus providing insight into potential mechanisms that may also occur in degenerating motor neurons.
Collapse
Affiliation(s)
- Nicla Romano
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Alessia Catalani
- Department of Molecular Sciences, University of Urbino "Carlo Bo", Via Santa Chiara, 27 61029 Urbino, PU, Italy
| | - Serena Lattante
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Unità Operativa Complessa di Genetica Medica, 00168 Roma, Italy; Università Cattolica del Sacro Cuore, Istituto di Medicina Genomica, 00168 Roma, Italy
| | - Antonio Belardo
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Silvia Proietti
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Laura Bertini
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Federica Silvestri
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| | - Lello Zolla
- Department of Science and Technology for Agriculture, Forestry, Nature and Energy, University of Tuscia (DAFNE), 01100 Viterbo, Italy
| | - Mario Sabatelli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Neurologia, Dipartimento Scienze dell'invecchiamento, neurologiche, ortopediche e della testa-collo, 00168 Roma, Italy; Università Cattolica del Sacro Cuore, Istituto di Neurologia, Centro Clinico NEMO, 00168 Roma, Italy
| | - Kristy Welshhans
- Department of Biological Sciences, School of Biomedical Sciences and Brain Health Research Institute, Kent State University, Kent, OH 44236, USA
| | - Marcello Ceci
- Department of Ecological and Biological Science (DEB), University of Tuscia, Largo dell'Università, 01100 Viterbo, Italy.
| |
Collapse
|