1
|
Deri E, Kumar Ojha S, Kartawy M, Khaliulin I, Amal H. Multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in autism spectrum disorder. Sci Rep 2025; 15:10878. [PMID: 40158064 PMCID: PMC11954894 DOI: 10.1038/s41598-025-95860-8] [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: 10/25/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Our multi-omics study investigated the molecular mechanisms underlying autism spectrum disorder (ASD) using Shank3Δ4-22 and Cntnap2-/- mouse models. Through global- and phospho- proteomics of the mouse cortex, we focused on shared molecular changes and found that autophagy was particularly affected in both models. Global proteomics identified a small number of differentially expressed proteins that significantly impact postsynaptic components and synaptic function, including key pathways such as mTOR signaling. Phosphoproteomics revealed unique phosphorylation sites in autophagy-related proteins such as ULK2, RB1CC1, ATG16L1, and ATG9, suggesting that altered phosphorylation patterns contribute to impaired autophagic flux in ASD. SH-SY5Y cells with SHANK3 gene deletion showed elevated LC3-II and p62 levels, indicating autophagosome accumulation and autophagy initiation, while the reduced level of the lysosomal activity marker LAMP1 suggested impaired autophagosome-lysosome fusion. The study highlights the involvement of reactive nitrogen species and nitric oxide (NO) on autophagy disruption. Importantly, inhibition of neuronal NO synthase (nNOS) by 7-NI normalized autophagy markers levels in the SH-SY5Y cells and primary cultured neurons. We have previously shown that nNOS inhibition improved synaptic and behavioral phenotypes in Shank3Δ4-22 and Cntnap2-/- mouse models. Our multi-omics study reveals differential expression and phosphorylation of autophagy-related proteins in ASD but further investigation is needed to prove the full involvement of autophagy in ASD. Our study underscores the need for further examination into the functional consequences of the identified phosphorylation sites, which may offer potential novel therapeutic autophagy-related targets for ASD treatment.
Collapse
Affiliation(s)
- Eden Deri
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Karnik A, Joshi A. SARM1: The Checkpoint of Axonal Degeneration in the Nervous System Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04835-3. [PMID: 40097763 DOI: 10.1007/s12035-025-04835-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
Axons are metabolically active neuronal segments with well-controlled axonal degeneration and regeneration. External stress or injury displaces this equilibrium toward degeneration leading to axonal dysfunction observed in the pathology of several diseases. The demand and supply matrix of energy at the synapses are maintained by the axonal transport. Nicotinamide adenine dinucleotide (NAD+) is a major energy-driving coenzyme of cells that controls mitochondrial, cytoplasmic, and other organellar energy cycles generating high amounts of adenosine triphosphate (ATP). NAD+ participates in various cellular cycles and is consumed by several enzymes. One of the key enzymes targeting NAD+ is Sterile alpha and TIR motif-containing protein 1 (SARM1) which gets activated in response to external noxious stimuli. SARM1 is an octamer consisting of multiple domains of which the TIR domain governs NAD+ hydrolysis which eventually leads to axonal deficits. Besides its localization in neurons, SARM1 is also present in astrocytes, microglia, and macrophages in which it regulates inflammatory responses associated with disease pathology. SARM1 localization in the outer mitochondrial membrane is responsible for its association with mitochondrial dynamics. SARM1-mediated mitochondrial dysfunction further drives the axonal degeneration associated with peripheral and central nervous system disorders. Several genetic and pharmacological studies highlight the role of SARM1 in axonal degeneration. SARM1 is thus becoming a popular target for preventing axonal degeneration. Several small molecules consisting of isoquinoline, isothiazole, pyridine, and tryptoline acrylamide moieties have been tested for their activity against SARM1 with a promising foundation for drug discovery in targeting SARM1. In our review, we highlight the role of SARM1 in axonal degeneration associated with several disease pathologies focusing on genetic and pharmacological evaluation.
Collapse
Affiliation(s)
- Aaditi Karnik
- Department of Pharmacy, Birla Institute of Technology and Sciences-Pilani, Telangana State, Hyderabad Campus, Hyderabad City, India
| | - Abhijeet Joshi
- Department of Pharmacy, Birla Institute of Technology and Sciences-Pilani, Telangana State, Hyderabad Campus, Hyderabad City, India.
| |
Collapse
|
3
|
Luo X, Zhang J, Tolö J, Kügler S, Michel U, Bähr M, Koch JC. Axonal autophagic vesicle transport in the rat optic nerve in vivo under normal conditions and during acute axonal degeneration. Acta Neuropathol Commun 2024; 12:82. [PMID: 38812004 PMCID: PMC11134632 DOI: 10.1186/s40478-024-01791-2] [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: 01/26/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024] Open
Abstract
Neurons pose a particular challenge to degradative processes like autophagy due to their long and thin processes. Autophagic vesicles (AVs) are formed at the tip of the axon and transported back to the soma. This transport is essential since the final degradation of the vesicular content occurs only close to or in the soma. Here, we established an in vivo live-imaging model in the rat optic nerve using viral vector mediated LC3-labeling and two-photon-microscopy to analyze axonal transport of AVs. Under basal conditions in vivo, 50% of the AVs are moving with a majority of 85% being transported in the retrograde direction. Transport velocity is higher in the retrograde than in the anterograde direction. A crush lesion of the optic nerve results in a rapid breakdown of retrograde axonal transport while the anterograde transport stays intact over several hours. Close to the lesion site, the formation of AVs is upregulated within the first 6 h after crush, but the clearance of AVs and the levels of lysosomal markers in the adjacent axon are reduced. Expression of p150Glued, an adaptor protein of dynein, is significantly reduced after crush lesion. In vitro, fusion and colocalization of the lysosomal marker cathepsin D with AVs are reduced after axotomy. Taken together, we present here the first in vivo analysis of axonal AV transport in the mammalian CNS using live-imaging. We find that axotomy leads to severe defects of retrograde motility and a decreased clearance of AVs via the lysosomal system.
Collapse
Affiliation(s)
- Xiaoyue Luo
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Jiong Zhang
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Johan Tolö
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Uwe Michel
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Jan Christoph Koch
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany.
| |
Collapse
|
4
|
Wen J, Zellner A, Braun NC, Bajaj T, Gassen NC, Peitz M, Brüstle O. Loss of function of FIP200 in human pluripotent stem cell-derived neurons leads to axonal pathology and hyperactivity. Transl Psychiatry 2023; 13:143. [PMID: 37137886 PMCID: PMC10156752 DOI: 10.1038/s41398-023-02432-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/22/2023] [Accepted: 04/12/2023] [Indexed: 05/05/2023] Open
Abstract
FIP200 plays important roles in homeostatic processes such as autophagy and signaling pathways such as focal adhesion kinase (FAK) signaling. Furthermore, genetic studies suggest an association of FIP200 mutations with psychiatric disorders. However, its potential connections to psychiatric disorders and specific roles in human neurons are not clear. We set out to establish a human-specific model to study the functional consequences of neuronal FIP200 deficiency. To this end, we generated two independent sets of isogenic human pluripotent stem cell lines with homozygous FIP200KO alleles, which were then used for the derivation of glutamatergic neurons via forced expression of NGN2. FIP200KO neurons exhibited pathological axonal swellings, showed autophagy deficiency, and subsequently elevated p62 protein levels. Moreover, monitoring the electrophysiological activity of neuronal cultures on multi-electrode arrays revealed that FIP200KO resulted in a hyperactive network. This hyperactivity could be abolished by glutamatergic receptor antagonist CNQX, suggesting a strengthened glutamatergic synaptic activation in FIP200KO neurons. Furthermore, cell surface proteomic analysis revealed metabolic dysregulation and abnormal cell adhesion-related processes in FIP200KO neurons. Interestingly, an ULK1/2-specific autophagy inhibitor could recapitulate axonal swellings and hyperactivity in wild-type neurons, whereas inhibition of FAK signaling was able to normalize the hyperactivity of FIP200KO neurons. These results suggest that impaired autophagy and presumably also disinhibition of FAK can contribute to the hyperactivity of FIP200KO neuronal networks, whereas pathological axonal swellings are primarily due to autophagy deficiency. Taken together, our study reveals the consequences of FIP200 deficiency in induced human glutamatergic neurons, which might, in the end, help to understand cellular pathomechanisms contributing to neuropsychiatric conditions.
Collapse
Affiliation(s)
- Jianbin Wen
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andreas Zellner
- Research Group Neurohomeostasis, Clinic and Polyclinic for Psychiatry and Psychotherapy, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Nils Christian Braun
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Clinic and Polyclinic for Psychiatry and Psychotherapy, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Nils Christian Gassen
- Research Group Neurohomeostasis, Clinic and Polyclinic for Psychiatry and Psychotherapy, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany.
- Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany.
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
5
|
Xuan Z, Yang S, Clark B, Hill SE, Manning L, Colón-Ramos DA. The active zone protein Clarinet regulates synaptic sorting of ATG-9 and presynaptic autophagy. PLoS Biol 2023; 21:e3002030. [PMID: 37053235 PMCID: PMC10101500 DOI: 10.1371/journal.pbio.3002030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/08/2023] [Indexed: 04/14/2023] Open
Abstract
Autophagy is essential for cellular homeostasis and function. In neurons, autophagosome biogenesis is temporally and spatially regulated to occur near presynaptic sites, in part via the trafficking of autophagy transmembrane protein ATG-9. The molecules that regulate autophagy by sorting ATG-9 at synapses remain largely unknown. Here, we conduct forward genetic screens at single synapses of C. elegans neurons and identify a role for the long isoform of the active zone protein Clarinet (CLA-1L) in regulating sorting of autophagy protein ATG-9 at synapses, and presynaptic autophagy. We determine that disrupting CLA-1L results in abnormal accumulation of ATG-9 containing vesicles enriched with clathrin. The ATG-9 phenotype in cla-1(L) mutants is not observed for other synaptic vesicle proteins, suggesting distinct mechanisms that regulate sorting of ATG-9-containing vesicles and synaptic vesicles. Through genetic analyses, we uncover the adaptor protein complexes that genetically interact with CLA-1 in ATG-9 sorting. We also determine that CLA-1L extends from the active zone to the periactive zone and genetically interacts with periactive zone proteins in ATG-9 sorting. Our findings reveal novel roles for active zone proteins in the sorting of ATG-9 and in presynaptic autophagy.
Collapse
Affiliation(s)
- Zhao Xuan
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sisi Yang
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Benjamin Clark
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sarah E. Hill
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Laura Manning
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Daniel A. Colón-Ramos
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico, San Juan,Puerto Rico
- Wu Tsai Institute, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
6
|
Fathy YY, Jonkman LE, Bol JJ, Timmermans E, Jonker AJ, Rozemuller AJM, van de Berg WDJ. Axonal degeneration in the anterior insular cortex is associated with Alzheimer's co-pathology in Parkinson's disease and dementia with Lewy bodies. Transl Neurodegener 2022; 11:52. [PMID: 36474289 PMCID: PMC9728006 DOI: 10.1186/s40035-022-00325-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Axons, crucial for impulse transmission and cellular trafficking, are thought to be primary targets of neurodegeneration in Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Axonal degeneration occurs early, preceeding and exceeding neuronal loss, and contributes to the spread of pathology, yet is poorly described outside the nigrostriatal circuitry. The insula, a cortical brain hub, was recently discovered to be highly vulnerable to pathology and plays a role in cognitive deficits in PD and DLB. The aim of this study was to evaluate morphological features as well as burden of proteinopathy and axonal degeneration in the anterior insular sub-regions in PD, PD with dementia (PDD), and DLB. METHODS α-Synuclein, phosphorylated (p-)tau, and amyloid-β pathology load were evaluated in the anterior insular (agranular and dysgranular) subregions of post-mortem human brains (n = 27). Axonal loss was evaluated using modified Bielschowsky silver staining and quantified using stereology. Cytoskeletal damage was comprehensively studied using immunofluorescent multi-labelling and 3D confocal laser-scanning microscopy. RESULTS Compared to PD and PDD, DLB showed significantly higher α-synuclein and p-tau pathology load, argyrophilic grains, and more severe axonal loss, particularly in the anterior agranular insula. Alternatively, the dysgranular insula showed a significantly higher load of amyloid-β pathology and its axonal density correlated with cognitive performance. p-Tau contributed most to axonal loss in the DLB group, was highest in the anterior agranular insula and significantly correlated with CDR global scores for dementia. Neurofilament and myelin showed degenerative changes including swellings, demyelination, and detachment of the axon-myelin unit. CONCLUSIONS Our results highlight the selective vulnerability of the anterior insular sub-regions to various converging pathologies, leading to impaired axonal integrity in PD, PDD and DLB, disrupting their functional properties and potentially contributing to cognitive, emotional, and autonomic deficits.
Collapse
Affiliation(s)
- Yasmine Y. Fathy
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands ,grid.5645.2000000040459992XDepartment of Neurology, Erasmus Medical Center, Postbus 2040, 3000 CA Rotterdam, Netherlands
| | - Laura E. Jonkman
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - John J. Bol
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Evelien Timmermans
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Allert J. Jonker
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| | - Annemieke J. M. Rozemuller
- grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands ,grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Pathology, Amsterdam Neuroscience, Vrije University Amsterdam, De Boelelaan, Amsterdam, Netherlands
| | - Wilma D. J. van de Berg
- grid.12380.380000 0004 1754 9227Amsterdam UMC, Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam Neuroscience, Vrije University Amsterdam, O
- 2 Life Sciences building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands ,grid.484519.5Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Bai X, Jiang M, Wang J, Yang S, Liu Z, Zhang H, Zhu X. Cyanidin attenuates the apoptosis of rat nucleus pulposus cells and the degeneration of intervertebral disc via the JAK2/STAT3 signal pathway in vitro and in vivo. PHARMACEUTICAL BIOLOGY 2022; 60:427-436. [PMID: 35175176 PMCID: PMC8856032 DOI: 10.1080/13880209.2022.2035773] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
CONTEXT Cyanidin has been shown to have therapeutic potential in osteoarthritis. However, it is unclear whether cyanidin prevents the progression of intervertebral disc degeneration (IVDD). OBJECTIVE This study evaluates the effects of cyanidin on IVDD in vitro and in vivo. MATERIALS AND METHODS Nucleus pulposus cells (NPCs) isolated from lumbar IVD of 4-week-old male Sprague-Dawley (SD) rats were exposed to 20 ng/mL IL-1β, and then treated with different doses (0-120 µM) of cyanidin for 24 h. SD rats were classified into three groups (n = 8) and treated as follows: control (normal saline), IVDD (vehicle), IVDD + cyanidin (50 mg/kg). Cyanidin was administered intraperitoneally for 8 weeks. RESULTS The IC50 of cyanidin for NPCs was 94.78 µM, and cyanidin had no toxicity at concentrations up to 500 mg/kg in SD rats. Cyanidin inhibited the apoptosis of NPCs induced by IL-1β (12.73 ± 0.61% vs. 18.54 ± 0.60%), promoted collagen II (0.82-fold) and aggrecan (0.81-fold) expression, while reducing MMP-13 (1.02-fold) and ADAMTS-5 (1.40-fold) expression. Cyanidin increased the formation of autophagosomes in IL-1β-induced NPCs, and promoted LC3II/LC3I (0.83-fold) and beclin-1 (0.85-fold) expression, which could be reversed by chloroquine. Cyanidin inhibited the phosphorylation of JAK2 (0.47-fold) and STAT3 (0.53-fold) in IL-1β-induced NPCs. The effects of cyanidin could be enhanced by AG490. Furthermore, cyanidin mitigated disc degeneration in IVDD rats in vivo. DISCUSSION AND CONCLUSIONS Cyanidin improved the function of NPCs in IVDD by regulating the JAK2/STAT3 pathway, which may provide a novel alternative strategy for IVDD. The mechanism of cyanidin improving IVDD still needs further work for in-depth investigation.
Collapse
Affiliation(s)
- Xiaoliang Bai
- Department of Orthopaedics, Baoding NO.1 Central Hospital, Baoding, China
- Department of Spine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Meichao Jiang
- Department of Orthopaedics, Baoding NO.1 Central Hospital, Baoding, China
| | - Jie Wang
- Department of Orthopaedics, Baoding NO.1 Central Hospital, Baoding, China
| | - Shuai Yang
- Department of Orthopaedics, Baoding NO.1 Central Hospital, Baoding, China
- Department of Spine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhiwei Liu
- Department of Orthopaedics, Baoding NO.1 Central Hospital, Baoding, China
| | - Hongxin Zhang
- Department of Orthopaedics, Baoding NO.1 Central Hospital, Baoding, China
| | - Xiaojuan Zhu
- Department of Geratology, Baoding NO.1 Central Hospital, Baoding, China
- CONTACT Xiaojuan Zhu Department of Geratology, Baoding NO.1 Central Hospital, No.320 Great Wall North Street, Baoding, 071000, China
| |
Collapse
|
8
|
Zhao Q, Pan W, Li J, Yu S, Liu Y, Zhang X, Qu R, Zhang Q, Li B, Yan X, Ren X, Qiu Y. Effects of neuron autophagy induced by arsenic and fluoride on spatial learning and memory in offspring rats. CHEMOSPHERE 2022; 308:136341. [PMID: 36087721 DOI: 10.1016/j.chemosphere.2022.136341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
There are numerous studies showing that exposure to arsenic (As) or fluoride (F) damages the nervous system, but there is no literature investigating the effects of combined As and F exposure to induce autophagy on neurotoxicity in the offspring. In this study, we developed a rat model of As and/or F exposure through drinking water from before pregnancy to 90 days postnatal. The offspring rats were randomly divided into nine groups. Sodium arsenite (NaAsO2) (0, 35, 70 mg/L) and Sodium fluoride (NaF) (0, 50, 100 mg/L) were designed according to 3 × 3 factorial design. Our results suggested that the presence of F might antagonize the excretion of total As in urine, and As-F co-exposure led to severe pathological damage in brain tissue and reduced spatial learning and memory ability. At the same time, the experiments showed that As and F increased Beclin1 expression and LC3B ratio to activate autophagy; both P62 and Lamp2 expression were increased, suggesting that autophagy lysosomal degradation was blocked; SYN and JIP1 expression were significantly decreased, disrupting synaptic structure and function. Axonal autophagosome reverse transport regulation might be affected by combined As-F exposure, exacerbating neuronal synaptic damage and inducing neurotoxicity. Further analysis showed that there was an interaction between As and F exposure-induced changes in autolysosome-related proteins in the hippocampus, which showed antagonism, and the antagonism of the high As combined exposure groups were stronger than that of the low As combined exposure groups. In conclusion, our study showed that combined As and F exposure might induce reverse transport impairment of autophagy on axons, leading to autophagy defects, which in turn led to disruption of synaptic morphology and function, induced neurotoxicity, and there was an interaction between As and F, the type of its combined effect was antagonism.
Collapse
Affiliation(s)
- Qiuyi Zhao
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Weizhe Pan
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Jia Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Shengnan Yu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Yan Liu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Xiaoli Zhang
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China; Department of Microbiology Laboratory, Linfen Central Hospital, Linfen, China.
| | - Ruodi Qu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Qian Zhang
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Ben Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Xiaoyan Yan
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| | - Xuefeng Ren
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA; Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Yulan Qiu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
9
|
Wang S, Song M, Yong H, Zhang C, Kang K, Liu Z, Yang Y, Huang Z, Wang S, Ge H, Zhao X, Song F. Mitochondrial Localization of SARM1 in Acrylamide Intoxication Induces Mitophagy and Limits Neuropathy. Mol Neurobiol 2022; 59:7337-7353. [PMID: 36171479 DOI: 10.1007/s12035-022-03050-8] [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] [Received: 03/14/2022] [Accepted: 09/14/2022] [Indexed: 10/14/2022]
Abstract
Sterile α and toll/interleukin 1 receptor motif-containing protein 1 (SARM1) is the defining molecule and central executioner of programmed axon death, also known as Wallerian degeneration. SARM1 has a mitochondrial targeting sequence, and it can bind to and stabilize PTEN-induced putative kinase 1 (PINK1) for mitophagy induction, but the deletion of the mitochondrial localization sequence is found to disrupt the mitochondrial localization of SARM1 in neurons without altering its ability to promote axon degeneration after axotomy. The biological significance of SARM1 mitochondrial localization remains elusive. In this study, we observed that the pro-degeneration factor, SARM1, was upregulated in acrylamide (ACR) neuropathy, a slow, Wallerian-like, programmed axonal death process. The upregulated SARM1 accumulated on mitochondria, interfered with mitochondrial dynamics, and activated PINK1-mediated mitophagy. Importantly, rapamycin (RAPA) intervention eliminated mitochondrial accumulation of SARM1 and partly attenuated ACR neuropathy. Thus, mitochondrial localization of SARM1 may contribute to its clearance through the SARM1-PINK1 mitophagy pathway, which inhibits axonal degeneration through a negative feedback loop. The mitochondrial localization of SARM1 complements the coordinated activity of the pro-survival factor, nicotinamide mononucleotide adenyltransferase 2 (NMNAT2), and SARM1 and is part of the self-limiting molecular mechanisms underpinning programmed axon death in ACR neuropathy. Mitophagy clearance of SARM1 is complementary to the coordinated activity of NMNAT2 and SARM1 in ACR neuropathy.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Mingxue Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Hui Yong
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Cuiqin Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Kang Kang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Zhidan Liu
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yiyu Yang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Zhengcheng Huang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Shu'e Wang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haotong Ge
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Xiulan Zhao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
10
|
Berth SH, Rich DJ, Lloyd TE. The role of autophagic kinases in regulation of axonal function. Front Cell Neurosci 2022; 16:996593. [PMID: 36226074 PMCID: PMC9548526 DOI: 10.3389/fncel.2022.996593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
Autophagy is an essential process for maintaining cellular homeostasis. Highlighting the importance of proper functioning of autophagy in neurons, disruption of autophagy is a common finding in neurodegenerative diseases. In recent years, evidence has emerged for the role of autophagy in regulating critical axonal functions. In this review, we discuss kinase regulation of autophagy in neurons, and provide an overview of how autophagic kinases regulate axonal processes, including axonal transport and axonal degeneration and regeneration. We also examine mechanisms for disruption of this process leading to neurodegeneration, focusing on the role of TBK1 in pathogenesis of Amyotrophic Lateral Sclerosis.
Collapse
|
11
|
Zhang X, Qiao Y, Han R, Gao Y, Yang X, Zhang Y, Wan Y, Yu W, Pan X, Xing J. A Charcot-Marie-Tooth-Causing Mutation in HSPB1 Decreases Cell Adaptation to Repeated Stress by Disrupting Autophagic Clearance of Misfolded Proteins. Cells 2022; 11:cells11182886. [PMID: 36139461 PMCID: PMC9496658 DOI: 10.3390/cells11182886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is the most common inherited neurodegenerative disorder with selective degeneration of peripheral nerves. Despite advances in identifying CMT-causing genes, the underlying molecular mechanism, particularly of selective degeneration of peripheral neurons remains to be elucidated. Since peripheral neurons are sensitive to multiple stresses, we hypothesized that daily repeated stress might be an essential contributor to the selective degeneration of peripheral neurons induced by CMT-causing mutations. Here, we mainly focused on the biological effects of the dominant missense mutation (S135F) in the 27-kDa small heat-shock protein HSPB1 under repeated heat shock. HSPB1S135F presented hyperactive binding to both α-tubulin and acetylated α-tubulin during repeated heat shock when compared with the wild type. The aberrant interactions with tubulin prevented microtubule-based transport of heat shock-induced misfolded proteins for the formation of perinuclear aggresomes. Furthermore, the transport of autophagosomes along microtubules was also blocked. These results indicate that the autophagy pathway was disrupted, leading to an accumulation of ubiquitinated protein aggregates and a significant decrease in cell adaptation to repeated stress. Our findings provide novel insights into the molecular mechanisms of HSPB1S135F-induced selective degeneration of peripheral neurons and perspectives for targeting autophagy as a promising therapeutic strategy for CMT neuropathy.
Collapse
Affiliation(s)
- Xuelian Zhang
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Yaru Qiao
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ronglin Han
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Yingjie Gao
- Department of Medicine Chemistry, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xun Yang
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ying Zhang
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ying Wan
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Wei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
- Correspondence: (J.X.); (X.P.); (W.Y.)
| | - Xianchao Pan
- Department of Medicine Chemistry, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Correspondence: (J.X.); (X.P.); (W.Y.)
| | - Juan Xing
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
- Correspondence: (J.X.); (X.P.); (W.Y.)
| |
Collapse
|
12
|
Shi J, Xu J, Li Y, Li B, Ming H, Nice EC, Huang C, Li Q, Wang C. Drug repurposing in cancer neuroscience: From the viewpoint of the autophagy-mediated innervated niche. Front Pharmacol 2022; 13:990665. [PMID: 36105204 PMCID: PMC9464986 DOI: 10.3389/fphar.2022.990665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Based on the bidirectional interactions between neurology and cancer science, the burgeoning field “cancer neuroscience” has been proposed. An important node in the communications between nerves and cancer is the innervated niche, which has physical contact with the cancer parenchyma or nerve located in the proximity of the tumor. In the innervated niche, autophagy has recently been reported to be a double-edged sword that plays a significant role in maintaining homeostasis. Therefore, regulating the innervated niche by targeting the autophagy pathway may represent a novel therapeutic strategy for cancer treatment. Drug repurposing has received considerable attention for its advantages in cost-effectiveness and safety. The utilization of existing drugs that potentially regulate the innervated niche via the autophagy pathway is therefore a promising pharmacological approach for clinical practice and treatment selection in cancer neuroscience. Herein, we present the cancer neuroscience landscape with an emphasis on the crosstalk between the innervated niche and autophagy, while also summarizing the underlying mechanisms of candidate drugs in modulating the autophagy pathway. This review provides a strong rationale for drug repurposing in cancer treatment from the viewpoint of the autophagy-mediated innervated niche.
Collapse
Affiliation(s)
- Jiayan Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jia Xu
- Department of Pharmacology, Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
| | - Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hui Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Qifu Li
- Department of Neurology and Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, The First Affiliated Hospital, Hainan Medical University, Haikou, China
- *Correspondence: Qifu Li, ; Chuang Wang,
| | - Chuang Wang
- Department of Pharmacology, Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
- *Correspondence: Qifu Li, ; Chuang Wang,
| |
Collapse
|
13
|
Rezaeian AH, Wei W, Inuzuka H. The regulation of neuronal autophagy and cell survival by MCL1 in Alzheimer's disease. ACTA MATERIA MEDICA 2022; 1:42-55. [PMID: 35233562 DOI: 10.15212/amm-2021-0002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Maintaining neuronal integrity and functions requires precise mechanisms controlling organelle and protein quality. Alzheimer's disease (AD) is characterized by functional defects in the clearance and recycling of intracellular components. As such, neuronal homeostasis involves autophagy, mitophagy, and apoptosis. Compromised activity in these cellular processes may cause pathological phenotypes of AD. Dysfunction of mitochondria is one of the hallmarks of AD. Mitophagy is a critical mitochondria quality control system, and the impaired mitophagy is observed in AD. Myeloid cell leukemia 1 (MCL1), a member of the pro-survival B-cell lymphoma protein 2 (BCL2) family, is a mitochondria-targeted protein that contributes to maintaining mitochondrial integrity. Mcl1 knockout mice display peri-implantation lethality. The studies on conditional Mcl1 knockout mice demonstrate that MCL1 plays a central role in neurogenesis and neuronal survival during brain development. Accumulating evidence reveals the critical role of MCL1 as a regulator of neuronal autophagy, mitophagy, and survival. In this review, we discuss the emerging neuroprotective function of MCL1 and how dysregulation of MCL1 signaling is involved in the pathogenesis of AD. As the pro-survival BCL2 family of proteins are promising targets of pharmacological intervention with BH3 mimetic drugs, we also discuss the promise of MCL1-targeting therapy in AD.
Collapse
Affiliation(s)
- Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Zhang C, Kang K, Chen Y, Shan S, Xie K, Song F. Atg7 Knockout Alleviated the Axonal Injury of Neuro-2a Cells Induced by Tri-Ortho-Cresyl Phosphate. Neurotox Res 2021; 39:1076-1086. [PMID: 33650059 DOI: 10.1007/s12640-021-00344-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/09/2021] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
Autophagy is believed to be essential for the maintenance of axonal homeostasis in neurons. However, whether autophagy is causally related to the axon degeneration in organophosphorus-induced delayed neuropathy (OPIDN) still remains unclear. This research was designed to investigate the role of autophagy in axon degeneration following tri-ortho-cresyl phosphate (TOCP) in an in vitro model. Differentiated wild-type and Atg7-/- neuro-2a (N2a) cells were treated with TOCP for 24 h. Axonal degeneration in N2a cells was quantitatively analyzed; the key molecules responsible for axon degeneration and its upstream signaling pathway were determined by Western blotting and real-time PCR. The results found that Atg7-/- cells exhibited a higher resistance to TOCP insult than wild-type cells. Further study revealed that TOCP caused a significant decrease in pro-survival factors NMNATs and SCG10 and a significant increase in pro-degenerative factor SARM1 in both cells. Notably, Atg7-/- cells presented a higher level of pro-survival factors and a lower level of pro-degenerative factors than wild-type cells in the same setting of TOCP administration. Moreover, DLK-MAPK pathway was activated following TOCP. Altogether, our results suggest that autophagy is able to affect TOCP-induced axonal injury via regulating the balance between pro-survival and pro-degenerative factors, providing a promising avenue for the potential therapy for OPIDN patients.
Collapse
Affiliation(s)
- Cuiqin Zhang
- Institute of Toxicology, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Kang Kang
- Institute of Toxicology, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Yisi Chen
- Institute of Toxicology, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Shulin Shan
- Institute of Toxicology, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Keqin Xie
- Institute of Toxicology, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong, 250012, People's Republic of China
| | - Fuyong Song
- Institute of Toxicology, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
15
|
Behrendt L, Hoischen C, Kaether C. Disease-causing mutated ATLASTIN 3 is excluded from distal axons and reduces axonal autophagy. Neurobiol Dis 2021; 155:105400. [PMID: 34019998 DOI: 10.1016/j.nbd.2021.105400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/27/2021] [Accepted: 05/16/2021] [Indexed: 10/21/2022] Open
Abstract
Mutations in the ER-network forming GTPase atlastin3 (ATL3) can cause axon degeneration of sensory neurons by not fully understood mechanisms. We here show that the hereditary sensory and autonomous neuropathy (HSAN)-causing ATL3 Y192C or P338R are excluded from distal axons by a barrier at the axon initial segment (AIS). This barrier is selective for mutated ATL3, but not wildtype ATL3 or unrelated ER-membrane proteins. Actin-depolymerization partially restores the transport of ATL3 Y192C into distal axons. The results point to the existence of a selective diffusion barrier in the ER membrane at the AIS, analogous to the AIS-based barriers for plasma membrane and cytosolic proteins. Functionally, the absence of ATL3 at the distal axon reduces axonal autophagy and the ER network deformation in the soma causes a reduction in axonal lysosomes. Both could contribute to axonal degeneration and eventually to HSAN.
Collapse
Affiliation(s)
- Laura Behrendt
- Leibniz-Institut für Alternsforschung-Fritz-Lipmann-Institut, 07745 Jena, Germany
| | - Christian Hoischen
- Leibniz-Institut für Alternsforschung-Fritz-Lipmann-Institut, 07745 Jena, Germany
| | - Christoph Kaether
- Leibniz-Institut für Alternsforschung-Fritz-Lipmann-Institut, 07745 Jena, Germany.
| |
Collapse
|
16
|
Molecular Basis of Neuronal Autophagy in Ageing: Insights from Caenorhabditis elegans. Cells 2021; 10:cells10030694. [PMID: 33800981 PMCID: PMC8004021 DOI: 10.3390/cells10030694] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/19/2023] Open
Abstract
Autophagy is an evolutionarily conserved degradation process maintaining cell homeostasis. Induction of autophagy is triggered as a response to a broad range of cellular stress conditions, such as nutrient deprivation, protein aggregation, organelle damage and pathogen invasion. Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane organelle referred to as the autophagosome with subsequent degradation of its contents upon delivery to lysosomes. Autophagy plays critical roles in development, maintenance and survival of distinct cell populations including neurons. Consequently, age-dependent decline in autophagy predisposes animals for age-related diseases including neurodegeneration and compromises healthspan and longevity. In this review, we summarize recent advances in our understanding of the role of neuronal autophagy in ageing, focusing on studies in the nematode Caenorhabditis elegans.
Collapse
|
17
|
Wang B, Huang M, Shang D, Yan X, Zhao B, Zhang X. Mitochondrial Behavior in Axon Degeneration and Regeneration. Front Aging Neurosci 2021; 13:650038. [PMID: 33762926 PMCID: PMC7982458 DOI: 10.3389/fnagi.2021.650038] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are organelles responsible for bioenergetic metabolism, calcium homeostasis, and signal transmission essential for neurons due to their high energy consumption. Accumulating evidence has demonstrated that mitochondria play a key role in axon degeneration and regeneration under physiological and pathological conditions. Mitochondrial dysfunction occurs at an early stage of axon degeneration and involves oxidative stress, energy deficiency, imbalance of mitochondrial dynamics, defects in mitochondrial transport, and mitophagy dysregulation. The restoration of these defective mitochondria by enhancing mitochondrial transport, clearance of reactive oxidative species (ROS), and improving bioenergetic can greatly contribute to axon regeneration. In this paper, we focus on the biological behavior of axonal mitochondria in aging, injury (e.g., traumatic brain and spinal cord injury), and neurodegenerative diseases (Alzheimer's disease, AD; Parkinson's disease, PD; Amyotrophic lateral sclerosis, ALS) and consider the role of mitochondria in axon regeneration. We also compare the behavior of mitochondria in different diseases and outline novel therapeutic strategies for addressing abnormal mitochondrial biological behavior to promote axonal regeneration in neurological diseases and injuries.
Collapse
Affiliation(s)
- Biyao Wang
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Minghao Huang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Dehao Shang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xu Yan
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Baohong Zhao
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xinwen Zhang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
18
|
Wang P, Chen X, Wang Y, Jia C, Liu X, Wang Y, Wu H, Cai H, Shen HM, Le W. Essential role for autophagy protein VMP1 in maintaining neuronal homeostasis and preventing axonal degeneration. Cell Death Dis 2021; 12:116. [PMID: 33483473 PMCID: PMC7822891 DOI: 10.1038/s41419-021-03412-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/27/2022]
Abstract
Vacuole membrane protein 1 (VMP1), the endoplasmic reticulum (ER)-localized autophagy protein, plays a key role during the autophagy process in mammalian cells. To study the impact of VMP1-deficiency on midbrain dopaminergic (mDAergic) neurons, we selectively deleted VMP1 in the mDAergic neurons of VMP1fl/fl/DATCreERT2 bigenic mice using a tamoxifen-inducible CreERT2/loxp gene targeting system. The VMP1fl/fl/DATCreERT2 mice developed progressive motor deficits, concomitant with a profound loss of mDAergic neurons in the substantia nigra pars compacta (SNc) and a high presynaptic accumulation of α-synuclein (α-syn) in the enlarged terminals. Mechanistic studies showed that VMP1 deficiency in the mDAergic neurons led to the increased number of microtubule-associated protein 1 light chain 3-labeled (LC3) puncta and the accumulation of sequestosome 1/p62 aggregates in the SNc neurons, suggesting the impairment of autophagic flux in these neurons. Furthermore, VMP1 deficiency resulted in multiple cellular abnormalities, including large vacuolar-like structures (LVSs), damaged mitochondria, swollen ER, and the accumulation of ubiquitin+ aggregates. Together, our studies reveal a previously unknown role of VMP1 in modulating neuronal survival and maintaining axonal homeostasis, which suggests that VMP1 deficiency might contribute to mDAergic neurodegeneration via the autophagy pathway.
Collapse
Affiliation(s)
- Panpan Wang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Xi Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Yuanyuan Wang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Congcong Jia
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Xinyao Liu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Ying Wang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Haifeng Wu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, 999078, SAR, China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116011, China.
- Institute of Neurology and Department of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Medical School of UETSC, Chengdu, 610072, China.
| |
Collapse
|
19
|
Di Giaimo R, Penna E, Pizzella A, Cirillo R, Perrone-Capano C, Crispino M. Cross Talk at the Cytoskeleton-Plasma Membrane Interface: Impact on Neuronal Morphology and Functions. Int J Mol Sci 2020; 21:ijms21239133. [PMID: 33266269 PMCID: PMC7730950 DOI: 10.3390/ijms21239133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/18/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022] Open
Abstract
The cytoskeleton and its associated proteins present at the plasma membrane not only determine the cell shape but also modulate important aspects of cell physiology such as intracellular transport including secretory and endocytic pathways. Continuous remodeling of the cell structure and intense communication with extracellular environment heavily depend on interactions between cytoskeletal elements and plasma membrane. This review focuses on the plasma membrane-cytoskeleton interface in neurons, with a special emphasis on the axon and nerve endings. We discuss the interaction between the cytoskeleton and membrane mainly in two emerging topics of neurobiology: (i) production and release of extracellular vesicles and (ii) local synthesis of new proteins at the synapses upon signaling cues. Both of these events contribute to synaptic plasticity. Our review provides new insights into the physiological and pathological significance of the cytoskeleton-membrane interface in the nervous system.
Collapse
Affiliation(s)
- Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
- Correspondence: (R.D.G.); (M.C.)
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Raffaella Cirillo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Carla Perrone-Capano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
- Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, National Research Council (CNR), 80131 Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
- Correspondence: (R.D.G.); (M.C.)
| |
Collapse
|
20
|
Yilmaz EN, Bay S, Ozturk G, Ucisik MH. Neuroprotective Effects of Curcumin-Loaded Emulsomes in a Laser Axotomy-Induced CNS Injury Model. Int J Nanomedicine 2020; 15:9211-9229. [PMID: 33244233 PMCID: PMC7685369 DOI: 10.2147/ijn.s272931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/01/2020] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Curcumin, a polyphenol isolated from the rhizomes of turmeric, holds great potential as a neuroprotective agent in addition to its anti-inflammatory and antioxidant characteristics. The poor bioavailability and low stability of curcumin are the greatest barriers to its clinical use. This study aims to investigate the neuroprotective effect of curcumin on axonal injury, by delivering the lipophilic polyphenol to a primary hippocampal neuron culture by means of a lipid-based drug delivery system, named emulsomes. METHODS To study neuroregeneration ex vivo, an injury model was established through single-cell laser axotomy on hippocampal neurites. Upon treatment with curcumin-loaded emulsomes (CurcuEmulsomes), curcumin and CurcuEmulsome uptake into neurons was verified by three-dimensional Z-stack images acquired with confocal microscopy. Neuron survival after axonal injury was tracked by propidium iodide (PI) and Hoechst staining. Alterations in expression levels of physiological markers, such as anti-apoptotic marker Bcl2, apoptotic marker cleaved caspase 3, neuroprotective marker Wnt3a and the neuronal survival marker mTOR, were investigated by immunocytochemistry analyses. RESULTS The results indicated significant improvement in the survival rate of injured neurons upon CurcuEmulsome treatment. Bcl2 expression was significantly higher for injured neurons treated with curcumin or CurcuEmulsome. Reduction in caspase 3 expression was seen in both curcumin and CurcuEmulsome treatment, whereas there were no significant changes in Wnt3a and mTOR expression. CONCLUSION The established laser-axotomy model was proven as a reliable methodology to study neurodegenerative models ex vivo. CurcuEmulsomes delivered curcumin to primary hippocampal neurons successfully. Treated with CurcuEmulsomes, injured hippocampal neurons benefit from the neuroprotective effects of curcumin, exhibiting a higher survival rate and increased anti-apoptotic marker levels.
Collapse
Affiliation(s)
- Elif Nur Yilmaz
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Beykoz, Istanbul, Turkey
- Graduate School of Engineering and Natural Sciences, Istanbul Medipol University, Beykoz, Istanbul, Turkey
| | - Sadik Bay
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Beykoz, Istanbul, Turkey
| | - Gurkan Ozturk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Beykoz, Istanbul, Turkey
- Department of Physiology, International School of Medicine, Istanbul Medipol University, Beykoz, Istanbul, Turkey
| | - Mehmet Hikmet Ucisik
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Beykoz, Istanbul, Turkey
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Beykoz, Istanbul, Turkey
| |
Collapse
|
21
|
Axon Injury-Induced Autophagy Activation Is Impaired in a C. elegans Model of Tauopathy. Int J Mol Sci 2020; 21:ijms21228559. [PMID: 33202845 PMCID: PMC7696692 DOI: 10.3390/ijms21228559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a conserved pathway that plays a key role in cell homeostasis in normal settings, as well as abnormal and stress conditions. Autophagy dysfunction is found in various neurodegenerative diseases, although it remains unclear whether autophagy impairment is a contributor or consequence of neurodegeneration. Axonal injury is an acute neuronal stress that triggers autophagic responses in an age-dependent manner. In this study, we investigate the injury-triggered autophagy response in a C. elegans model of tauopathy. We found that transgenic expression of pro-aggregant Tau, but not the anti-aggregant Tau, abolished axon injury-induced autophagy activation, resulting in a reduced axon regeneration capacity. Furthermore, axonal trafficking of autophagic vesicles were significantly reduced in the animals expressing pro-aggregant F3ΔK280 Tau, indicating that Tau aggregation impairs autophagy regulation. Importantly, the reduced number of total or trafficking autophagic vesicles in the tauopathy model was not restored by the autophagy activator rapamycin. Loss of PTL-1, the sole Tau homologue in C. elegans, also led to impaired injury-induced autophagy activation, but with an increased basal level of autophagic vesicles. Therefore, we have demonstrated that Tau aggregation as well as Tau depletion both lead to disruption of injury-induced autophagy responses, suggesting that aberrant protein aggregation or microtubule dysfunction can modulate autophagy regulation in neurons after injury.
Collapse
|
22
|
Oliva Trejo JA, Tanida I, Suzuki C, Kakuta S, Tada N, Uchiyama Y. Characterization of starvation-induced autophagy in cerebellar Purkinje cells of pHluorin-mKate2-human LC3B transgenic mice. Sci Rep 2020; 10:9643. [PMID: 32541814 PMCID: PMC7295967 DOI: 10.1038/s41598-020-66370-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/19/2020] [Indexed: 12/25/2022] Open
Abstract
We generated a new transgenic mouse model that expresses a pHluorin-mKate2 fluorescent protein fused with human LC3B (PK-LC3 mice) for monitoring autophagy activity in neurons of the central nervous system. Histological analysis revealed fluorescent puncta in neurons of the cerebral cortex, hippocampus, cerebellar Purkinje cells, and anterior spinal regions. Using CLEM analysis, we confirmed that PK-LC3-positive puncta in the perikarya of Purkinje cells correspond to autophagic structures. To validate the usability of PK-LC3 mice, we quantified PK-LC3 puncta in Purkinje cells of mice kept in normal feeding conditions and of mice starved for 24 hours. Our results showed a significant increase in autophagosome number and in individual puncta areal size following starvation. To confirm these results, we used morphometry at the electron microscopic level to analyze the volume densities of autophagosomes and lysosomes/autolysosomes in Purkinje cells of PK-LC3 mice. The results revealed that the volume densities of autophagic structures increase significantly after starvation. Together, our data show that PK-LC3 mice are suitable for monitoring autophagy flux in Purkinje cells of the cerebellum, and potentially other areas in the central nervous system.
Collapse
Affiliation(s)
- Juan Alejandro Oliva Trejo
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Isei Tanida
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan.
| | - Chigure Suzuki
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Soichiro Kakuta
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Norihiro Tada
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan.
| |
Collapse
|
23
|
Yuan YS, Niu SP, Yu F, Zhang YJ, Han N, Lu H, Yin XF, Xu HL, Kou YH. Intraoperative single administration of neutrophil peptide 1 accelerates the early functional recovery of peripheral nerves after crush injury. Neural Regen Res 2020; 15:2108-2115. [PMID: 32394969 PMCID: PMC7716025 DOI: 10.4103/1673-5374.282270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neutrophil peptide 1 belongs to a family of peptides involved in innate immunity. Continuous intramuscular injection of neutrophil peptide 1 can promote the regeneration of peripheral nerves, but clinical application in this manner is not convenient. To this end, the effects of a single intraoperative administration of neutrophil peptide 1 on peripheral nerve regeneration were experimentally observed. A rat model of sciatic nerve crush injury was established using the clamp method. After model establishment, a normal saline group and a neutrophil peptide 1 group were injected with a single dose of normal saline or 10 μg/mL neutrophil peptide 1, respectively. A sham group, without sciatic nerve crush was also prepared as a control. Sciatic nerve function tests, neuroelectrophysiological tests, and hematoxylin-eosin staining showed that the nerve conduction velocity, sciatic functional index, and tibialis anterior muscle fiber cross-sectional area were better in the neutrophil peptide 1 group than in the normal saline group at 4 weeks after surgery. At 4 and 8 weeks after surgery, there were no differences in the wet weight of the tibialis anterior muscle between the neutrophil peptide 1 and saline groups. Histological staining of the sciatic nerve showed no significant differences in the number of myelinated nerve fibers or the axon cross-sectional area between the neutrophil peptide 1 and normal saline groups. The above data confirmed that a single dose of neutrophil peptide 1 during surgery can promote the recovery of neurological function 4 weeks after sciatic nerve injury. All the experiments were approved by the Medical Ethics Committee of Peking University People’s Hospital, China (approval No. 2015-50) on December 9, 2015.
Collapse
Affiliation(s)
- Yu-Song Yuan
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| | - Su-Ping Niu
- Office of Academic Research, Peking University People's Hospital, Beijing, China
| | - Fei Yu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing; National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Ya-Jun Zhang
- National Center for Trauma Medicine, Beijing, China
| | - Na Han
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education; Office of Academic Research, Peking University People's Hospital, Beijing, China
| | - Hao Lu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Diabetic Foot Treatment Center, Peking University People's Hospital, Peking University, Beijing, China
| | - Xiao-Feng Yin
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| | - Hai-Lin Xu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Diabetic Foot Treatment Center, Peking University People's Hospital, Peking University, Beijing, China
| | - Yu-Hui Kou
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
24
|
Horton RH, Wileman T, Rushworth SA. Autophagy Driven Extracellular Vesicles in the Leukaemic Microenvironment. Curr Cancer Drug Targets 2020; 20:501-512. [PMID: 32342819 DOI: 10.2174/1568009620666200428111051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 12/27/2019] [Accepted: 03/29/2020] [Indexed: 12/12/2022]
Abstract
The leukaemias are a heterogeneous group of blood cancers, which together, caused 310,000 deaths in 2016. Despite significant research into their biology and therapeutics, leukaemia is predicted to account for an increased 470,000 deaths in 2040. Many subtypes remain without targeted therapy, and therefore the mainstay of treatment remains generic cytotoxic drugs with bone marrow transplant the sole definitive option. In this review, we will focus on cellular mechanisms which have the potential for therapeutic exploitation to specifically target and treat this devastating disease. We will bring together the disciplines of autophagy and extracellular vesicles, exploring how the dysregulation of these mechanisms can lead to changes in the leukaemic microenvironment and the subsequent propagation of disease. The dual effect of these mechanisms in the disease microenvironment is not limited to leukaemia; therefore, we briefly explore their role in autoimmunity, inflammation and degenerative disease.
Collapse
Affiliation(s)
- Rebecca H Horton
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
| | - Tom Wileman
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
| | - Stuart A Rushworth
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
| |
Collapse
|
25
|
Autophagy lipidation machinery regulates axonal microtubule dynamics but is dispensable for survival of mammalian neurons. Nat Commun 2020; 11:1535. [PMID: 32210230 PMCID: PMC7093409 DOI: 10.1038/s41467-020-15287-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
Neurons maintain axonal homeostasis via employing a unique organization of the microtubule (MT) cytoskeleton, which supports axonal morphology and provides tracks for intracellular transport. Abnormal MT-based trafficking hallmarks the pathology of neurodegenerative diseases, but the exact mechanism regulating MT dynamics in axons remains enigmatic. Here we report on a regulation of MT dynamics by AuTophaGy(ATG)-related proteins, which previously have been linked to the autophagy pathway. We find that ATG proteins required for LC3 lipid conjugation are dispensable for survival of excitatory neurons and instead regulate MT stability via controlling the abundance of the MT-binding protein CLASP2. This function of ATGs is independent of their role in autophagy and requires the active zone protein ELKS1. Our results highlight a non-canonical role of ATG proteins in neurons and suggest that pharmacological activation of autophagy may not only promote the degradation of cytoplasmic material, but also impair axonal integrity via altering MT stability. In neurons, the microtubule cytoskeleton provides support and directionality of axons. Here, the authors report that microtubule dynamics in axons may be regulated by the autophagy proteins (ATGs) independently of their known role in autophagy.
Collapse
|
26
|
Li JN, Zhang Z, Wu GZ, Yao DB, Cui SS. Claudin-15 overexpression inhibits proliferation and promotes apoptosis of Schwann cells in vitro. Neural Regen Res 2020; 15:169-177. [PMID: 31535666 PMCID: PMC6862392 DOI: 10.4103/1673-5374.264463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Our previous experiments have discovered that Claudin-15 was up-regulated in Schwann cells of the distal nerve stumps of rat models of sciatic nerve injury. However, how Claudin-15 affects Schwann cell function is still unknown. This study aimed to identify the effects of Claudin-15 on proliferation and apoptosis of Schwann cells cultured in vitro and explore the underlying mechanisms. Primary Schwann cells were obtained from rats. Claudin-15 in Schwann cells was knocked down using siRNA (siRNA-1 group) compared with the negative control siRNA transfection group (negative control group). Claudin-15 in Schwann cells was overexpressed using pGV230-Claudin-15 plasmid (pGV230-Claudin-15 group). The pGV230 transfection group (pGV230 group) acted as the control of the pGV230-Claudin-15 group. Cell proliferation was analyzed with EdU assay. Cell apoptosis was analyzed with flow cytometric analysis. Cell migration was analyzed with Transwell inserts. The mRNA and protein expressions were analyzed with quantitative polymerase chain reaction assay and western blot assay. The results showed that compared with the negative control group, cell proliferation rate was up-regulated; p-AKT/AKT ratio, apoptotic rate, p-c-Jun/c-Jun ratio, mRNA expression of protein kinase C alpha, Bcl-2 and Bax were down-regulated; and mRNA expression of neurotrophins basic fibroblast growth factor and neurotrophin-3 were increased in the siRNA-1 group. No significant difference was found in cell migration between the negative control and siRNA-1 groups. Compared with the pGV230 group, the cell proliferation rate was down-regulated; apoptotic rate, p-c-Jun/c-Jun ratio and c-Fos protein expression increased; mRNA expression of protein kinase C alpha and Bax decreased; and mRNA expressions of neurotrophins basic fibroblast growth factor and neurotrophin-3 were up-regulated in the pGV230-Claudin-15 group. The above results demonstrated that overexpression of Claudin-15 inhibited Schwann cell proliferation and promoted Schwann cell apoptosis in vitro. Silencing of Claudin-15 had the reverse effect and provided neuroprotective effect. This study was approved by the Experimental Animal Ethics Committee of Jilin University of China (approval No. 2016-nsfc001) on March 5, 2016.
Collapse
Affiliation(s)
- Jian-Nan Li
- China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhan Zhang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Guang-Zhi Wu
- China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Deng-Bing Yao
- School of Life Sciences, Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Shu-Sen Cui
- China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
27
|
Telegina DV, Suvorov GK, Kozhevnikova OS, Kolosova NG. Mechanisms of Neuronal Death in the Cerebral Cortex during Aging and Development of Alzheimer's Disease-Like Pathology in Rats. Int J Mol Sci 2019; 20:E5632. [PMID: 31717998 PMCID: PMC6888576 DOI: 10.3390/ijms20225632] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the commonest type of late-life dementia and damages the cerebral cortex, a vulnerable brain region implicated in memory, emotion, cognition, and decision-making behavior. AD is characterized by progressive neuronal loss, but the mechanisms of cell death at different stages of the disease remain unknown. Here, by means of OXYS rats as an appropriate model of the most common (sporadic) AD form, we studied the main pathways of cell death during development of AD-like pathology, including the preclinical stage. We found that apoptosis is activated at the pre-symptomatic stage (age 20 days) correlating with the retardation of brain development in the OXYS strain early in life. Progression of the AD-like pathology was accompanied by activation of apoptosis and necroptosis resulting from a decline of autophagy-mediated proteostasis. Our results are consistent with the idea that the nature of changes in the pathways of apoptosis, autophagy, and necrosis depends on the stage of AD.
Collapse
Affiliation(s)
- Darya V. Telegina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia; (D.V.T.); (G.K.S.); (O.S.K.)
| | - Gleb K. Suvorov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia; (D.V.T.); (G.K.S.); (O.S.K.)
| | - Oyuna S. Kozhevnikova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia; (D.V.T.); (G.K.S.); (O.S.K.)
| | - Nataliya G. Kolosova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Pr. Lavrentyeva 10, Novosibirsk 630090, Russia; (D.V.T.); (G.K.S.); (O.S.K.)
- Novosibirsk State University, 1 Pirogova str., Novosibirsk 630090, Russia
| |
Collapse
|
28
|
Moss KR, Höke A. Targeting the programmed axon degeneration pathway as a potential therapeutic for Charcot-Marie-Tooth disease. Brain Res 2019; 1727:146539. [PMID: 31689415 DOI: 10.1016/j.brainres.2019.146539] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022]
Abstract
The programmed axon degeneration pathway has emerged as an important process contributing to the pathogenesis of several neurological diseases. The most crucial events in this pathway include activation of the central executioner SARM1 and NAD+ depletion, which leads to an energetic failure and ultimately axon destruction. Given the prevalence of this pathway, it is not surprising that inhibitory therapies are currently being developed in order to treat multiple neurological diseases with the same therapy. Charcot-Marie-Tooth disease (CMT) is a heterogeneous group of neurological diseases that may also benefit from this therapeutic approach. To evaluate the appropriateness of this strategy, the contribution of the programmed axon degeneration pathway to the pathogenesis of different CMT subtypes is being actively investigated. The subtypes CMT1A, CMT1B and CMT2D are the first to have been examined. Based on the results from these studies and advances in developing therapies to block the programmed axon degeneration pathway, promising therapeutics for CMT are now on the horizon.
Collapse
Affiliation(s)
- Kathryn R Moss
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Ahmet Höke
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
29
|
Wang H, Wang X, Zhang K, Wang Q, Cao X, Wang Z, Zhang S, Li A, Liu K, Fang Y. Rapid depletion of ESCRT protein Vps4 underlies injury-induced autophagic impediment and Wallerian degeneration. SCIENCE ADVANCES 2019; 5:eaav4971. [PMID: 30788439 PMCID: PMC6374107 DOI: 10.1126/sciadv.aav4971] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/02/2019] [Indexed: 06/01/2023]
Abstract
Injured axons undergo a controlled, self-destruction process, known as Wallerian degeneration. However, the underlying mechanism remains elusive. Using the Drosophila wing nerve as a model, we identify the ESCRT component Vps4 as a previously unidentified essential gene for axonal integrity. Up-regulation of Vps4 remarkably delays degeneration of injured axons. We further reveal that Vps4 is required and sufficient to promote autophagic flux in axons and mammalian cells. Moreover, using both in vitro and in vivo models, we show that the function of Vps4 in maintaining axonal autophagy and suppressing Wallerian degeneration is conserved in mammals. Last, we uncover that Vps4 protein is rapidly depleted in injured mouse axons, which may underlie the injury-induced autophagic impediment and the subsequent axonal degeneration. Together, Vps4 and ESCRT may represent a novel signal transduction mechanism in axon injury and Wallerian degeneration.
Collapse
Affiliation(s)
- Haiqiong Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Kai Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingyao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Cao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Shuang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Jinan University, Guangzhou 510632, China
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
30
|
Casares-Crespo L, Calatayud-Baselga I, García-Corzo L, Mira H. On the Role of Basal Autophagy in Adult Neural Stem Cells and Neurogenesis. Front Cell Neurosci 2018; 12:339. [PMID: 30349462 PMCID: PMC6187079 DOI: 10.3389/fncel.2018.00339] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/13/2018] [Indexed: 12/31/2022] Open
Abstract
Adult neurogenesis persists in the adult mammalian brain due to the existence of neural stem cell (NSC) reservoirs in defined niches, where they give rise to new neurons throughout life. Recent research has begun to address the implication of constitutive (basal) autophagy in the regulation of neurogenesis in the mature brain. This review summarizes the current knowledge on the role of autophagy-related genes in modulating adult NSCs, progenitor cells and their differentiation into neurons. The general function of autophagy in neurogenesis in several areas of the embryonic forebrain is also revisited. During development, basal autophagy regulates Wnt and Notch signaling and is mainly required for adequate neuronal differentiation. The available data in the adult indicate that the autophagy-lysosomal pathway regulates adult NSC maintenance, the activation of quiescent NSCs, the survival of the newly born neurons and the timing of their maturation. Future research is warranted to validate the results of these pioneering studies, refine the molecular mechanisms underlying the regulation of NSCs and newborn neurons by autophagy throughout the life-span of mammals and provide significance to the autophagic process in adult neurogenesis-dependent behavioral tasks, in physiological and pathological conditions. These lines of research may have important consequences for our understanding of stem cell dysfunction and neurogenic decline during healthy aging and neurodegeneration.
Collapse
Affiliation(s)
- Lucía Casares-Crespo
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| | - Isabel Calatayud-Baselga
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| | - Laura García-Corzo
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| | - Helena Mira
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| |
Collapse
|
31
|
Daniel S, Clark AF, McDowell CM. Subtype-specific response of retinal ganglion cells to optic nerve crush. Cell Death Discov 2018; 4:7. [PMID: 30062056 PMCID: PMC6054657 DOI: 10.1038/s41420-018-0069-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 06/03/2018] [Indexed: 01/09/2023] Open
Abstract
Glaucoma is a neurodegenerative disease with retinal ganglion cell (RGC) loss, optic nerve degeneration and subsequent vision loss. There are about 30 different subtypes of RGCs whose response to glaucomatous injury is not well characterized. The purpose of this study was to evaluate the response of 4 RGC subtypes in a mouse model of optic nerve crush (ONC). In this study, we also evaluated the pattern of axonal degeneration in RGC subtypes after nerve injury. We found that out of the 4 subtypes, transient-Off α RGCs are the most susceptible to injury followed by On-Off direction selective RGCs (DSGC). Non-image forming RGCs are more resilient with ipRGCs exhibiting the most resistance of them all. In contrast, axons degenerate irrespective of their retinal soma after ONC injury. In conclusion, we show that RGCs have subtype specific cell death response to ONC injury and that RGC axons disintegrate in an autonomous fashion undergoing Wallerian degeneration. These discoveries can further direct us towards effective diagnostic and therapeutic approaches to treat optic neuropathies, such as glaucoma.
Collapse
Affiliation(s)
- S. Daniel
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas United States
| | - AF Clark
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas United States
| | - CM McDowell
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas United States
| |
Collapse
|