1
|
Zhang Y, Yao T, Xu Y, Wang Y, Han S. Circulating RAC1 contributed to steroid-sensitive nephrotic syndrome: Mendelian randomization, single-cell RNA-sequencing, proteomic, and experimental evidence. Ren Fail 2024; 46:2416087. [PMID: 39422242 PMCID: PMC11492449 DOI: 10.1080/0886022x.2024.2416087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVES The small GTPase Rac1 (RAC1) has been linked to podocyte disorders and steroid-sensitive nephrotic syndrome (SSNS). The aim of this study was to explore and validate the potential causal association between circulating RAC1 and SSNS. METHODS The association between circulating RAC1 and SSNS at both gene expression and proteomic levels was investigated using Mendelian randomization analysis, and further validated by single-cell RNA-sequencing, proteomic analysis, and experimental studies. The genetic instruments comprised cis-expression quantitative trait loci (cis-eQTLs) associated with RAC1 gene expression and protein QTLs correlated with plasma RAC1 protein levels. Causal associations were estimated utilizing the inverse variance weighted and MR-PRESSO methods. Validation of RAC1 expression was conducted through single-cell RNA-sequencing of peripheral blood mononuclear cells from patients with SSNS and healthy controls. Proteomic analysis was performed among patients with minimal change nephrotic syndrome. Experimental validation was conducted using a puromycin aminonucleoside (PAN)-induced nephrosis model. RESULTS Increased expression of RAC1 was associated with a higher risk of SSNS (gene expression level: odds ratio [OR], 1.53; 95% confidence interval [CI], 1.02-2.28; protein level: OR, 1.82; 95% CI, 1.05-3.17). The results of MR-PRESSO were consistent (gene expression level: OR, 1.49; 95% CI, 1.17-1.92; protein level: OR, 1.81; 95% CI, 1.16-2.85). Single-cell RNA sequencing and proteomic analysis confirmed elevated RAC1 expression in patients with SSNS compared to healthy controls. Experimental data further supported increased RAC1 expression in PAN-induced nephropathy. CONCLUSIONS Increased expression of RAC1 might be causally associated with SSNS, suggesting that targeting RAC1 might represent a potential therapeutic strategy for SSNS.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianwen Yao
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqiu Xu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Wang
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shisheng Han
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Alves VC, Carro E, Figueiro-Silva J. Unveiling DNA methylation in Alzheimer's disease: a review of array-based human brain studies. Neural Regen Res 2024; 19:2365-2376. [PMID: 38526273 PMCID: PMC11090417 DOI: 10.4103/1673-5374.393106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/05/2023] [Indexed: 03/26/2024] Open
Abstract
The intricacies of Alzheimer's disease pathogenesis are being increasingly illuminated by the exploration of epigenetic mechanisms, particularly DNA methylation. This review comprehensively surveys recent human-centered studies that investigate whole genome DNA methylation in Alzheimer's disease neuropathology. The examination of various brain regions reveals distinctive DNA methylation patterns that associate with the Braak stage and Alzheimer's disease progression. The entorhinal cortex emerges as a focal point due to its early histological alterations and subsequent impact on downstream regions like the hippocampus. Notably, ANK1 hypermethylation, a protein implicated in neurofibrillary tangle formation, was recurrently identified in the entorhinal cortex. Further, the middle temporal gyrus and prefrontal cortex were shown to exhibit significant hypermethylation of genes like HOXA3, RHBDF2, and MCF2L, potentially influencing neuroinflammatory processes. The complex role of BIN1 in late-onset Alzheimer's disease is underscored by its association with altered methylation patterns. Despite the disparities across studies, these findings highlight the intricate interplay between epigenetic modifications and Alzheimer's disease pathology. Future research efforts should address methodological variations, incorporate diverse cohorts, and consider environmental factors to unravel the nuanced epigenetic landscape underlying Alzheimer's disease progression.
Collapse
Affiliation(s)
- Victoria Cunha Alves
- Neurodegenerative Diseases Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
- Neurotraumatology and Subarachnoid Hemorrhage Group, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Eva Carro
- Network Center for Biomedical Research, Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research Into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Joana Figueiro-Silva
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Science, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Hu J, Li H, Wang X, Cheng H, Zhu G, Yang S. Novel mechanisms of Anshen Dingzhi prescription against PTSD: Inhibiting DCC to modulate synaptic function and inflammatory responses. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118425. [PMID: 38848974 DOI: 10.1016/j.jep.2024.118425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anshen Dingzhi prescription (ADP), documented in "Yi Xue Xin Wu", is a famous prescription for treating panic-related mental disorders such as post-traumatic stress disorder (PTSD). However, the underlying mechanism remains unclear. AIM OF THE STUDY This study aimed to investigate the mechanisms by which ADP intervened in PTSD-like behaviors. METHODS A mouse model of single prolonged stress (SPS) was established to evaluate the ameliorative effects and mechanisms of ADP on PTSD. Behavioral tests were used to assess PTSD-like behaviors in mice; transmission electron microscopy was used to observe changes in the ultrastructure of hippocampal synapses, and western blot, immunofluorescence, and ELISA were used to detect the expression of hippocampal deleted in colorectal cancer (DCC) and downstream Ras-related C3 botulinum toxin substrate 1 (Rac1) - P21-activated kinase 1 (PAK1) signal, as well as levels of synaptic proteins and inflammatory factors. Molecular docking technology simulated the binding of potential brain-penetrating components of ADP to DCC. RESULTS SPS induced PTSD-like behaviors in mice and increased expression of hippocampal netrin-1 (NT-1) and DCC on the 14th day post-modeling, with concurrent elevation in serum NT-1 levels. Simultaneously, SPS also decreased p-Rac1 level and increased p-PAK1 level, the down-stream molecules of DCC. Lentiviral overexpression of DCC induced or exacerbated PTSD-like behaviors in control and SPS mice, respectively, whereas neutralization antibody against NT-1 reduced DCC activation and ameliorated PTSD-like behaviors in SPS mice. Interestingly, downstream Rac1-PAK1 signal was altered according to DCC expression. Moreover, DCC overexpression down-regulated N-methyl-d-aspartate (NMDA) receptor 2A (GluN2A) and postsynaptic density 95 (PSD95), up-regulated NMDA receptor 2B (GluN2B) and increased neuroinflammatory responses. Administration of ADP (36.8 mg/kg) improved PTSD-like behaviors in the SPS mice, suppressed hippocampal DCC, and downstream Rac1-PAK1 signal, upregulated GluN2A and PSD95, downregulated GluN2B, and reduced levels of inflammatory factors NOD-like receptor protein 3 (NLRP3), nuclear factor kappa-B (NF-κB) and interleukin-6 (IL-6). Importantly, DCC overexpression could also reduce the ameliorative effect of ADP on PTSD. Additionally, DCC demonstrated a favorable molecular docking pattern with the potential brain-penetrating components of ADP, further suggesting DCC as a potential target of ADP. CONCLUSION Our data indicate that DCC is a key target for the regulation of synaptic function and inflammatory response in the onset of PTSD, and ADP likely reduces DCC to prevent PTSD via modulating downstream Rac1-PAK1 pathway. This study provides a novel mechanism for the onset of PTSD and warrants the clinical application of ADP.
Collapse
Affiliation(s)
- Jiamin Hu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Haipeng Li
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Xuncui Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Hongliang Cheng
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230061, China.
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China.
| | - Shaojie Yang
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, 230061, China.
| |
Collapse
|
4
|
Berry JA, Guhle DC, Davis RL. Active forgetting and neuropsychiatric diseases. Mol Psychiatry 2024; 29:2810-2820. [PMID: 38532011 PMCID: PMC11420092 DOI: 10.1038/s41380-024-02521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
Recent and pioneering animal research has revealed the brain utilizes a variety of molecular, cellular, and network-level mechanisms used to forget memories in a process referred to as "active forgetting". Active forgetting increases behavioral flexibility and removes irrelevant information. Individuals with impaired active forgetting mechanisms can experience intrusive memories, distressing thoughts, and unwanted impulses that occur in neuropsychiatric diseases. The current evidence indicates that active forgetting mechanisms degrade, or mask, molecular and cellular memory traces created in synaptic connections of "engram cells" that are specific for a given memory. Combined molecular genetic/behavioral studies using Drosophila have uncovered a complex system of cellular active-forgetting pathways within engram cells that is regulated by dopamine neurons and involves dopamine-nitric oxide co-transmission and reception, endoplasmic reticulum Ca2+ signaling, and cytoskeletal remodeling machinery regulated by small GTPases. Some of these molecular cellular mechanisms have already been found to be conserved in mammals. Interestingly, some pathways independently regulate forgetting of distinct memory types and temporal phases, suggesting a multi-layering organization of forgetting systems. In mammals, active forgetting also involves modulation of memory trace synaptic strength by altering AMPA receptor trafficking. Furthermore, active-forgetting employs network level mechanisms wherein non-engram neurons, newly born-engram neurons, and glial cells regulate engram synapses in a state and experience dependent manner. Remarkably, there is evidence for potential coordination between the network and cellular level forgetting mechanisms. Finally, subjects with several neuropsychiatric diseases have been tested and shown to be impaired in active forgetting. Insights obtained from research on active forgetting in animal models will continue to enrich our understanding of the brain dysfunctions that occur in neuropsychiatric diseases.
Collapse
Affiliation(s)
- Jacob A Berry
- Department of Biological Sciences, University of Alberta, Edmonton, AL, T6G 2E9, Canada.
| | - Dana C Guhle
- Department of Biological Sciences, University of Alberta, Edmonton, AL, T6G 2E9, Canada
| | - Ronald L Davis
- Department of Neuroscience, UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA.
| |
Collapse
|
5
|
Yang S, Hu J, Chen Y, Zhang Z, Wang J, Zhu G. DCC, a potential target for controlling fear memory extinction and hippocampal LTP in male mice receiving single prolonged stress. Neurobiol Stress 2024; 32:100666. [PMID: 39224830 PMCID: PMC11366904 DOI: 10.1016/j.ynstr.2024.100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/29/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a severe stress-dependent psychiatric disorder characterized by impairment of fear memory extinction; however, biological markers to determine impaired fear memory extinction in PTSD remain unclear. In male mice with PTSD-like behaviors elicited by single prolonged stress (SPS), 19 differentially expressed proteins in the hippocampus were identified compared with controls. Among them, a biological macromolecular protein named deleted in colorectal cancer (DCC) was highly upregulated. Specific overexpression of DCC in the hippocampus induced similar impairment of long-term potentiation (LTP) and fear memory extinction as observed in SPS mice. The impairment of fear memory extinction in SPS mice was improved by inhibiting the function of hippocampal DCC using a neutralizing antibody. Mechanistic studies have shown that knocking down or inhibiting μ-calpain in hippocampal neurons increased DCC expression and induced impairment of fear memory extinction. Additionally, SPS-triggered impairment of hippocampal LTP and fear memory extinction could be rescued through activation of the Rac1-Pak1 signaling pathway. Our study provides evidence that calpain-mediated regulation of DCC controls hippocampal LTP and fear memory extinction in SPS mice, which likely through activation of the Rac1-Pak1 signaling pathway.
Collapse
Affiliation(s)
- Shaojie Yang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Shouchun Road 300, Hefei, 230061, China
| | - Jiamin Hu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| | - Yuzhuang Chen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| | - Zhengrong Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| | - Jingji Wang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Shouchun Road 300, Hefei, 230061, China
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Longzhihu Road 350, Hefei, 230012, China
| |
Collapse
|
6
|
Chen R, Wang Z, Lin Q, Hou X, Jiang Y, Le Q, Liu X, Ma L, Wang F. Destabilization of fear memory by Rac1-driven engram-microglia communication in hippocampus. Brain Behav Immun 2024; 119:621-636. [PMID: 38670239 DOI: 10.1016/j.bbi.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024] Open
Abstract
Rac1 is a key regulator of the cytoskeleton and neuronal plasticity, and is known to play a critical role in psychological and cognitive brain disorders. To elucidate the engram specific Rac1 signaling in fear memory, a doxycycline (Dox)-dependent robust activity marking (RAM) system was used to label dorsal dentate gyrus (DG) engram cells in mice during contextual fear conditioning. Rac1 mRNA and protein levels in DG engram cells were peaked at 24 h (day 1) after fear conditioning and were more abundant in the fear engram cells than in the non-engram cells. Optogenetic activation of Rac1 in a temporal manner in DG engram cells before memory retrieval decreased the freezing level in the fear context. Optogenetic activation of Rac1 increased autophagy protein 7 (ATG7) expression in the DG engram cells and activated DG microglia. Microglia-specific transcriptomics and fluorescence in situ hybridization revealed that overexpression of ATG7 in the fear engram cells upregulated the mRNA of Toll-like receptor TLR2/4 in DG microglia. Knockdown of microglial TLR2/4 rescued fear memory destabilization induced by ATG7 overexpression or Rac1 activation in DG engram cells. These results indicate that Rac1-driven communications between engram cells and microglia contributes to contextual fear memory destabilization, and is mediated by ATG7 and TLR2/4, and suggest a novel mechanistic framework for the cytoskeletal regulator in fear memory interference.
Collapse
Affiliation(s)
- Ruyan Chen
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Zhilin Wang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qing Lin
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Xutian Hou
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Yan Jiang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qiumin Le
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Xing Liu
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Lan Ma
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Feifei Wang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China.
| |
Collapse
|
7
|
Chiu J, Krupa JM, Seah C, Pasternak SH. Small GTPases control macropinocytosis of amyloid precursor protein and cleavage to amyloid-β. Heliyon 2024; 10:e31077. [PMID: 38799759 PMCID: PMC11126852 DOI: 10.1016/j.heliyon.2024.e31077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
The overproduction of the toxic peptide amyloid-beta (Aβ) generated from the cleavage of amyloid precursor protein (APP) is proposed to be a critical event in the development of Alzheimer's disease. Evidence suggests that the cleavage of APP occurs after its internalization from the cell surface. Previously, we identified a novel pathway for APP internalization, which trafficks cell surface APP directly to lysosomes by macropinocytosis, leading to its processing into Aβ. We also demonstrated that ADP-ribosylation factor 6 (Arf6) is required for the macropinocytosis of APP. Here, we characterized the roles of Arf6's downstream effectors Rac1, Cdc42 and RhoA. Both pharmacological inhibition and siRNA knockdown of these proteins reduced the amount of APP colocalized with LAMP1-labeled lysosomes without affecting APP transport to early endosomes. Decreases in the production of both Aβ40 and Aβ42 were also observed by ELISA in response to inhibitor treatment. These findings together demonstrate that Rac1, Cdc42 and RhoA are components of the mechanism regulating the macropinocytosis of APP and targeting these components can reduce the production of Aβ.
Collapse
Affiliation(s)
- Justin Chiu
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jordan M. Krupa
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Claudia Seah
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen H. Pasternak
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
8
|
Bailly C, Degand C, Laine W, Sauzeau V, Kluza J. Implication of Rac1 GTPase in molecular and cellular mitochondrial functions. Life Sci 2024; 342:122510. [PMID: 38387701 DOI: 10.1016/j.lfs.2024.122510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Rac1 is a member of the Rho GTPase family which plays major roles in cell mobility, polarity and migration, as a fundamental regulator of actin cytoskeleton. Signal transduction by Rac1 occurs through interaction with multiple effector proteins, and its activity is regulated by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). The small protein is mainly anchored to the inner side of the plasma membrane but it can be found in endocellular compartments, notably endosomes and cell nuclei. The protein localizes also into mitochondria where it contributes to the regulation of mitochondrial dynamics, including both mitobiogenesis and mitophagy, in addition to signaling processes via different protein partners, such as the proapoptotic protein Bcl-2 and chaperone sigma-1 receptor (σ-1R). The mitochondrial form of Rac1 (mtRac1) has been understudied thus far, but it is as essential as the nuclear or plasma membrane forms, via its implication in regulation of oxidative stress and DNA damages. Rac1 is subject to diverse post-translational modifications, notably to a geranylgeranylation which contributes importantly to its mitochondrial import and its anchorage to mitochondrial membranes. In addition, Rac1 contributes to the mitochondrial translocation of other proteins, such as p53. The mitochondrial localization and functions of Rac1 are discussed here, notably in the context of human diseases such as cancers. Inhibitors of Rac1 have been identified (NSC-23766, EHT-1864) and some are being developed for the treatment of cancer (MBQ-167) or central nervous system diseases (JK-50561). Their effects on mtRac1 warrant further investigations. An overview of mtRac1 is provided here.
Collapse
Affiliation(s)
- Christian Bailly
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France; University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; OncoWitan, Consulting Scientific Office, Lille (Wasquehal) 59290, France.
| | - Claire Degand
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - William Laine
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Vincent Sauzeau
- Université de Nantes, CHU Nantes, CNRS, INSERM, Institut du thorax, Nantes, France
| | - Jérôme Kluza
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| |
Collapse
|
9
|
Zang C, Liu H, Ning J, Chen Q, Jiang Y, Shang M, Yang Y, Ma J, Dong Y, Wang J, Li F, Bao X, Zhang D. Emerging role and mechanism of HACE1 in the pathogenesis of neurodegenerative diseases: A promising target. Biomed Pharmacother 2024; 172:116204. [PMID: 38364733 DOI: 10.1016/j.biopha.2024.116204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/18/2024] Open
Abstract
HACE1 is a member of the HECT domain-containing E3 ligases with 909 amino acid residues, containing N-terminal ankyrin-repeats (ANK) and C-terminal HECT domain. Previously, it was shown that HACE1 is inactive in human tumors and plays a crucial role in the initiation, progression, and invasion of malignant tumors. Recent studies indicated that HACE1 might be closely involved in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. HACE1 interacts with its substrates, including Ras-related C3 botulinum toxin substrate 1 (Rac1), nuclear factor erythroid 2-related factor 2 (Nrf2), tumor necrosis factor receptor (TNFR), and optineurin (OPTN), through which participates in several pathophysiological processes, such as oxidative stress, autophagy and inflammation. Therefore, in this review, we elaborately describe the essential substrates of HACE1 and illuminate the pathophysiological processes by which HACE1 is involved in neurodegenerative diseases. We provide a new molecular target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Caixia Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Hui Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Jingwen Ning
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Qiuzhu Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Yueqi Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Meiyu Shang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Yang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Jingwei Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Yirong Dong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Jinrong Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Fangfang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Xiuqi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China.
| |
Collapse
|
10
|
Ohno M. A Strategy for Allowing Earlier Diagnosis and Rigorous Evaluation of BACE1 Inhibitors in Preclinical Alzheimer's Disease. J Alzheimers Dis 2024; 99:431-445. [PMID: 38701146 DOI: 10.3233/jad-231451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Given continued failure of BACE1 inhibitor programs at symptomatic and prodromal stages of Alzheimer's disease (AD), clinical trials need to target the earlier preclinical stage. However, trial design is complex in this population with negative diagnosis of classical hippocampal amnesia on standard memory tests. Besides recent advances in brain imaging, electroencephalogram, and fluid-based biomarkers, new cognitive markers should be established for earlier diagnosis that can optimize recruitment to BACE1 inhibitor trials in presymptomatic AD. Notably, accelerated long-term forgetting (ALF) is emerging as a sensitive cognitive measure that can discriminate between asymptomatic individuals with high risks for developing AD and healthy controls. ALF is a form of declarative memory impairment characterized by increased forgetting rates over longer delays (days to months) despite normal storage within the standard delays of testing (20-60 min). Therefore, ALF may represent a harbinger of preclinical dementia and the impairment of systems memory consolidation, during which memory traces temporarily stored in the hippocampus become gradually integrated into cortical networks. This review provides an overview of the utility of ALF in a rational design of next-generation BACE1 inhibitor trials in preclinical AD. I explore potential mechanisms underlying ALF and relevant early-stage biomarkers useful for BACE1 inhibitor evaluation, including synaptic protein alterations, astrocytic dysregulation and neuron hyperactivity in the hippocampal-cortical network. Furthermore, given the physiological role of the isoform BACE2 as an AD-suppressor gene, I also discuss the possible association between the poor selectivity of BACE1 inhibitors and their side effects (e.g., cognitive worsening) in prior clinical trials.
Collapse
Affiliation(s)
- Masuo Ohno
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
11
|
Zhu M, Liu Y, Chen C, Chen H, Ni W, Song Y, Lv B, Hua F, Cui G, Zhang Z. TLR4/Rac1/NLRP3 Pathway Mediates Amyloid-β-Induced Neuroinflammation in Alzheimer's Disease. J Alzheimers Dis 2024; 99:911-925. [PMID: 38728187 DOI: 10.3233/jad-240012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
Background Neuroinflammation plays a crucial part in the initial onset and progression of Alzheimer's disease (AD). NLRP3 inflammasome was demonstrated to get involved in amyloid-β (Aβ)-induced neuroinflammation. However, the mechanism of Aβ-triggered activation of NLRP3 inflammasome remains poorly understood. Objective Based on our previous data, the study aimed to identify the downstream signals that bridge the activation of TLR4 and NLRP3 inflammasome associated with Aβ. Methods BV-2 cells were transfected with TLR4siRNA or pretreated with a CLI-095 or NSC23766, followed by Aβ1-42 treatment. APP/PS1 mice were injected intraperitoneally with CLI-095 or NSC23766. NLRP3 inflammasome and microglia activation was detected with immunostaining and western blot. G-LISA and Rac1 pull-down activation test were performed to investigate the activation of Rac1. Real-time PCR and ELISA were used to detect the inflammatory cytokines. Aβ plaques were assessed by western blotting and immunofluorescence staining. Morris water maze test was conducted to determine the spatial memory in mice. Results Rac1 and NLRP3 inflammasome were activated by Aβ in both in vitro and in vivo experiments. Inhibition of TLR4 reduced the activity of Rac1 and NLRP3 inflammasome induced by Aβ1-42. Furthermore, inhibition of Rac1 blocked NLRP3 inflammasome activation mediated by TLR4. Blocking the pathway by CLI095 or NSC23766 suppressed Aβ1-42-triggered activation of microglia, reduced the expression of pro-inflammatory mediators and ameliorated the cognition deficits in APP/PS1 mice. Conclusions Our study demonstrated that TLR4/Rac1/NLRP3 pathway mediated Aβ-induced neuroinflammation, which unveiled a novel pathway and key contributors underlying the pathogenic mechanism of Aβ.
Collapse
Affiliation(s)
- Mengxin Zhu
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yang Liu
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, Xi'an People's hospital, Xi'an, China
| | - Chen Chen
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hao Chen
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wanyan Ni
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanjian Song
- Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Bingchen Lv
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fang Hua
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guiyun Cui
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zuohui Zhang
- Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
12
|
Zhu M, Xiao B, Xue T, Qin S, Ding J, Wu Y, Tang Q, Huang M, Zhao N, Ye Y, Zhang Y, Zhang B, Li J, Guo F, Jiang Y, Zhang L, Zhang L. Cdc42GAP deficiency contributes to the Alzheimer's disease phenotype. Brain 2023; 146:4350-4365. [PMID: 37254741 DOI: 10.1093/brain/awad184] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 06/01/2023] Open
Abstract
Alzheimer's disease, the most common cause of dementia, is a chronic degenerative disease with typical pathological features of extracellular senile plaques and intracellular neurofibrillary tangles and a significant decrease in the density of neuronal dendritic spines. Cdc42 is a member of the small G protein family that plays an important role in regulating synaptic plasticity and is regulated by Cdc42GAP, which switches Cdc42 from active GTP-bound to inactive GDP-bound states regulating downstream pathways via effector proteins. However, few studies have focused on Cdc42 in the progression of Alzheimer's disease. In a heterozygous Cdc42GAP mouse model that exhibited elevated Cdc42-GTPase activity accompanied by increased Cdc42-PAK1-cofilin signalling, we found impairments in cognitive behaviours, neuron senescence, synaptic loss with depolymerization of F-actin and the pathological phenotypes of Alzheimer's disease, including phosphorylated tau (p-T231, AT8), along with increased soluble and insoluble Aβ1-42 and Aβ1-40, which are consistent with typical Alzheimer's disease mice. Interestingly, these impairments increased significantly with age. Furthermore, the results of quantitative phosphoproteomic analysis of the hippocampus of 11-month-old GAP mice suggested that Cdc42GAP deficiency induces and accelerates Alzheimer's disease-like phenotypes through activation of GSK-3β by dephosphorylation at Ser9, Ser389 and/or phosphorylation at Tyr216. In addition, overexpression of dominant-negative Cdc42 in the primary hippocampal and cortical neurons of heterozygous Cdc42GAP mice reversed synaptic loss and tau hyperphosphorylation. Importantly, the Cdc42 signalling pathway, Aβ1-42, Aβ1-40 and GSK-3β activity were increased in the cortical sections of Alzheimer's disease patients compared with those in healthy controls. Together, these data indicated that Cdc42GAP is involved in regulating Alzheimer's disease-like phenotypes such as cognitive deficits, dendritic spine loss, phosphorylated tau (p-T231, AT8) and increased soluble and insoluble Aβ1-42 and Aβ1-40, possibly through the activation of GSK-3β, and these impairments increased significantly with age. Thus, we provide the first evidence that Cdc42 is involved in the progression of Alzheimer's disease-like phenotypes, which may provide new targets for Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Mengjuan Zhu
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Xiao
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tao Xue
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Sifei Qin
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Yue Wu
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qingqiu Tang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Mengfan Huang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Na Zhao
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Yingshan Ye
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuning Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Boya Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Juan Li
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Center for Orthopedic Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, Cincinnati, OH 45229-3026, USA
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin Zhang
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Center for Orthopedic Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
13
|
Guskjolen A, Cembrowski MS. Engram neurons: Encoding, consolidation, retrieval, and forgetting of memory. Mol Psychiatry 2023; 28:3207-3219. [PMID: 37369721 PMCID: PMC10618102 DOI: 10.1038/s41380-023-02137-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
Tremendous strides have been made in our understanding of the neurobiological substrates of memory - the so-called memory "engram". Here, we integrate recent progress in the engram field to illustrate how engram neurons transform across the "lifespan" of a memory - from initial memory encoding, to consolidation and retrieval, and ultimately to forgetting. To do so, we first describe how cell-intrinsic properties shape the initial emergence of the engram at memory encoding. Second, we highlight how these encoding neurons preferentially participate in synaptic- and systems-level consolidation of memory. Third, we describe how these changes during encoding and consolidation guide neural reactivation during retrieval, and facilitate memory recall. Fourth, we describe neurobiological mechanisms of forgetting, and how these mechanisms can counteract engram properties established during memory encoding, consolidation, and retrieval. Motivated by recent experimental results across these four sections, we conclude by proposing some conceptual extensions to the traditional view of the engram, including broadening the view of cell-type participation within engrams and across memory stages. In collection, our review synthesizes general principles of the engram across memory stages, and describes future avenues to further understand the dynamic engram.
Collapse
Affiliation(s)
- Axel Guskjolen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Mark S Cembrowski
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Wang W, Wang Z, Cao J, Dong Y, Chen Y. Roles of Rac1-Dependent Intrinsic Forgetting in Memory-Related Brain Disorders: Demon or Angel. Int J Mol Sci 2023; 24:10736. [PMID: 37445914 DOI: 10.3390/ijms241310736] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Animals are required to handle daily massive amounts of information in an ever-changing environment, and the resulting memories and experiences determine their survival and development, which is critical for adaptive evolution. However, intrinsic forgetting, which actively deletes irrelevant information, is equally important for memory acquisition and consolidation. Recently, it has been shown that Rac1 activity plays a key role in intrinsic forgetting, maintaining the balance of the brain's memory management system in a controlled manner. In addition, dysfunctions of Rac1-dependent intrinsic forgetting may contribute to memory deficits in neurological and neurodegenerative diseases. Here, these new findings will provide insights into the neurobiology of memory and forgetting, pathological mechanisms and potential therapies for brain disorders that alter intrinsic forgetting mechanisms.
Collapse
Affiliation(s)
- Wei Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Zixu Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jing Cao
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yulan Dong
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yaoxing Chen
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, Beijing Laboratory of Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| |
Collapse
|
15
|
Wang C, Yang Y, Zhang X, Shi Z, Gao H, Zhong M, Fan Y, Zhang H, Liu B, Qing G. Secreted endogenous macrosomes reduce Aβ burden and ameliorate Alzheimer's disease. SCIENCE ADVANCES 2023; 9:eade0293. [PMID: 37235655 DOI: 10.1126/sciadv.ade0293] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
Innovative therapeutic strategies are urgently needed for Alzheimer's disease (AD) due to the increasing size of the aging population and the lack of effective drug treatment. Here, we report the therapeutic effects of extracellular vesicles (EVs) secreted by microglia, including macrosomes and small EVs, on AD-associated pathology. Macrosomes strongly inhibited β-amyloid (Aβ) aggregation and rescued cells from Aβ misfolding-induced cytotoxicity. Furthermore, macrosome administration reduced Aβ plaques and ameliorated cognitive impairment in mice with AD. In contrast, small EVs slightly promoted Aβ aggregation and did not improve AD pathology. Proteomic analysis of small EVs and macrosomes revealed that macrosomes harbor several important neuroprotective proteins that inhibit Aβ misfolding. In particular, the small integral membrane protein 10-like protein 2B in macrosomes has been shown to inhibit Aβ aggregation. Our observations provide an alternative therapeutic strategy for the treatment of AD over conventional ineffective drug treatments.
Collapse
Affiliation(s)
- Cunli Wang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian 116024, P. R. China
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Yiming Yang
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian 116024, P. R. China
| | - Xiaoyu Zhang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Zhenqiang Shi
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Huiling Gao
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, P. R. China
| | - Manli Zhong
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, P. R. China
| | - Yonggang Fan
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System, China Medical University, Shenyang, 110122, P. R. China
| | - Hongyan Zhang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
| | - Bo Liu
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Lingshui Road, Dalian 116024, P. R. China
| | - Guangyan Qing
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, P. R. China
- Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| |
Collapse
|
16
|
Ohno M. Accelerated long-term forgetting: A sensitive paradigm for detecting subtle cognitive impairment and evaluating BACE1 inhibitor efficacy in preclinical Alzheimer's disease. FRONTIERS IN DEMENTIA 2023; 2:1161875. [PMID: 39081986 PMCID: PMC11285641 DOI: 10.3389/frdem.2023.1161875] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/27/2023] [Indexed: 08/02/2024]
Abstract
Given a long preclinical stage of Alzheimer's disease (AD) continuum before the onset of dementia, there is a growing demand for tools capable of detecting the earliest feature of subtle cognitive impairment and optimizing recruitment to clinical trials for potentially disease-modifying therapeutic interventions such as BACE1 inhibitors. Now that all BACE1 inhibitor programs in symptomatic and prodromal AD populations have ended in failure, trials need to shift to target the earlier preclinical stage. However, evaluating cognitive efficacy (if any) in asymptomatic AD individuals is a great challenge. In this context, accelerated long-term forgetting (ALF) is emerging as a sensitive cognitive measure that can discriminate between presymptomatic individuals with high risks for developing AD and healthy controls. ALF is characterized by increased forgetting rates over extended delays (e.g., days, weeks, months) despite normal learning and short-term retention on standard memory assessments that typically use around 30-min delays. This review provides an overview of recent progress in animal model and clinical studies on this topic, focusing on the utility and underlying mechanism of ALF that may be applicable to earlier diagnosis and BACE1 inhibitor efficacy evaluation at a preclinical stage of AD.
Collapse
Affiliation(s)
- Masuo Ohno
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
| |
Collapse
|
17
|
Pham AQ, Dore K. Novel approaches to increase synaptic resilience as potential treatments for Alzheimer's disease. Semin Cell Dev Biol 2023; 139:84-92. [PMID: 35370089 DOI: 10.1016/j.semcdb.2022.03.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022]
Abstract
A significant proportion of brains with Alzheimer's disease pathology are obtained from patients that were cognitively normal, suggesting that differences within the brains of these individuals made them resilient to the disease. Here, we describe recent approaches that specifically increase synaptic resilience, as loss of synapses is considered to be the first change in the brains of Alzheimer's patients. We start by discussing studies showing benefit from increased expression of neurotrophic factors and protective genes. Methods that effectively make dendritic spines stronger, specifically by acting through actin network proteins, scaffolding proteins and inhibition of phosphatases are described next. Importantly, the therapeutic strategies presented in this review tackle Alzheimer's disease not by targeting plaques and tangles, but instead by making synapses resilient to the pathology associated with Alzheimer's disease, which has tremendous potential.
Collapse
Affiliation(s)
- Andrew Q Pham
- Department of Neurosciences, Center for Neural Circuits and Behavior, UCSD, La Jolla 92093, United States
| | - Kim Dore
- Department of Neurosciences, Center for Neural Circuits and Behavior, UCSD, La Jolla 92093, United States.
| |
Collapse
|
18
|
Yang Q, Zhou J, Wang L, Hu W, Zhong Y, Li Q. Spontaneous recovery of reward memory through active forgetting of extinction memory. Curr Biol 2023; 33:838-848.e3. [PMID: 36731465 DOI: 10.1016/j.cub.2023.01.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 12/16/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023]
Abstract
Learned behavior can be suppressed by the extinction procedure. Such extinguished memory often returns spontaneously over time, making it difficult to treat diseases such as addiction. However, the biological mechanisms underlying such spontaneous recovery remain unclear. Here, we report that the extinguished reward memory in Drosophila recovers spontaneously because extinction training forms an aversive memory that can be actively forgotten via the Rac1/Dia pathway. Manipulating Rac1 activity does not affect sugar-reward memory and its immediate extinction effect but bidirectionally regulates spontaneous recovery-the decay process of extinction. Experiments using thermogenetic inhibition and functional imaging support that such extinction appears to be coded as an aversive experience. Genetic and pharmacological inhibition of formin Dia, a downstream effector of Rac1, specifically prevents spontaneous recovery after extinction in both behavioral performance and corresponding physiological traces. Together, our data suggest that spontaneous recovery is caused by active forgetting of the opposing extinction memory.
Collapse
Affiliation(s)
- Qi Yang
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jun Zhou
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Lingling Wang
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Wantong Hu
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yi Zhong
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| | - Qian Li
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
19
|
Ding J, Huang J, Tang X, Shen L, Hu S, He J, Liu T, Yu Z, Liu Y, Wang Q, Wang J, Zhao N, Qi X, Huang J. Low and high dose methamphetamine differentially regulate synaptic structural plasticity in cortex and hippocampus. Front Cell Neurosci 2022; 16:1003617. [PMID: 36406748 PMCID: PMC9666390 DOI: 10.3389/fncel.2022.1003617] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/19/2022] [Indexed: 03/24/2024] Open
Abstract
Psychostimulants, such as methamphetamine (METH) can induce structural remodeling of synapses by remodeling presynaptic and postsynaptic morphology. Escalating or long-lasting high dose METH accounts for neurodegeneration by targeting multiple neurotransmitters. However, the effects of low dose METH on synaptic structure and the modulation mechanism remain elusive. This study aims to assess the effects of low dose (2 mg/kg) and high dose (10 mg/kg) of METH on synaptic structure alternation in hippocampus and prefrontal cortex (PFC) and to reveal the underlying mechanism involved in the process. Low dose METH promoted spine formation, synaptic number increase, post-synaptic density length elongation, and memory function. High dose of METH induced synaptic degeneration, neuronal number loss and memory impairment. Moreover, high dose, but not low dose, of METH caused gliosis in PFC and hippocampus. Mechanism-wise, low dose METH inactivated ras-related C3 botulinum toxin substrate 1 (Rac1) and activated cell division control protein 42 homolog (Cdc42); whereas high dose METH inactivated Cdc42 and activated Rac1. We provided evidence that low and high doses of METH differentially regulate synaptic plasticity in cortex and hippocampus.
Collapse
Affiliation(s)
- Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jian Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiang Tang
- Department of Children Rehabilitation, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Lingyi Shen
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Shanshan Hu
- Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaojiao He
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Zhixing Yu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Yubo Liu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Qiyan Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Na Zhao
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Jiang Huang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
| |
Collapse
|
20
|
Wurz AI, Schulz AM, O’Bryant CT, Sharp JF, Hughes RM. Cytoskeletal dysregulation and neurodegenerative disease: Formation, monitoring, and inhibition of cofilin-actin rods. Front Cell Neurosci 2022; 16:982074. [PMID: 36212686 PMCID: PMC9535683 DOI: 10.3389/fncel.2022.982074] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The presence of atypical cytoskeletal dynamics, structures, and associated morphologies is a common theme uniting numerous diseases and developmental disorders. In particular, cytoskeletal dysregulation is a common cellular feature of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. While the numerous activators and inhibitors of dysregulation present complexities for characterizing these elements as byproducts or initiators of the disease state, it is increasingly clear that a better understanding of these anomalies is critical for advancing the state of knowledge and plan of therapeutic attack. In this review, we focus on the hallmarks of cytoskeletal dysregulation that are associated with cofilin-linked actin regulation, with a particular emphasis on the formation, monitoring, and inhibition of cofilin-actin rods. We also review actin-associated proteins other than cofilin with links to cytoskeleton-associated neurodegenerative processes, recognizing that cofilin-actin rods comprise one strand of a vast web of interactions that occur as a result of cytoskeletal dysregulation. Our aim is to present a current perspective on cytoskeletal dysregulation, connecting recent developments in our understanding with emerging strategies for biosensing and biomimicry that will help shape future directions of the field.
Collapse
Affiliation(s)
- Anna I. Wurz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Anna M. Schulz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Collin T. O’Bryant
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Josephine F. Sharp
- Department of Chemistry, Notre Dame College, South Euclid, OH, United States
| | - Robert M. Hughes
- Department of Chemistry, East Carolina University, Greenville, NC, United States
- *Correspondence: Robert M. Hughes,
| |
Collapse
|
21
|
Blok LER, Boon M, van Reijmersdal B, Höffler KD, Fenckova M, Schenck A. Genetics, molecular control and clinical relevance of habituation learning. Neurosci Biobehav Rev 2022; 143:104883. [PMID: 36152842 DOI: 10.1016/j.neubiorev.2022.104883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
Habituation is the most fundamental form of learning. As a firewall that protects our brain from sensory overload, it is indispensable for cognitive processes. Studies in humans and animal models provide increasing evidence that habituation is affected in autism and related monogenic neurodevelopmental disorders (NDDs). An integrated application of habituation assessment in NDDs and their animal models has unexploited potential for neuroscience and medical care. With the aim to gain mechanistic insights, we systematically retrieved genes that have been demonstrated in the literature to underlie habituation. We identified 258 evolutionarily conserved genes across species, describe the biological processes they converge on, and highlight regulatory pathways and drugs that may alleviate habituation deficits. We also summarize current habituation paradigms and extract the most decisive arguments that support the crucial role of habituation for cognition in health and disease. We conclude that habituation is a conserved, quantitative, cognition- and disease-relevant process that can connect preclinical and clinical work, and hence is a powerful tool to advance research, diagnostics, and treatment of NDDs.
Collapse
Affiliation(s)
- Laura Elisabeth Rosalie Blok
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Marina Boon
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Boyd van Reijmersdal
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Kira Daniela Höffler
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands; Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| |
Collapse
|
22
|
Liu X, Wang J. NMDA receptors mediate synaptic plasticity impairment of hippocampal neurons due to arsenic exposure. Neuroscience 2022; 498:300-310. [PMID: 35905926 DOI: 10.1016/j.neuroscience.2022.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/08/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022]
Abstract
Endemic arsenism is a worldwide health problem. Chronic arsenic exposure results in cognitive dysfunction due to arsenic and its metabolites accumulating in hippocampus. As the cellular basis of cognition, synaptic plasticity is pivotal in arsenic-induced cognitive dysfunction. N-methyl-D-aspartate receptors (NMDARs) serve physiological functions in synaptic transmission. However, excessive NMDARs activity contributes to exitotoxicity and synaptic plasticity impairment. Here, we provide an overview of the mechanisms that NMDARs and their downstream signaling pathways mediate synaptic plasticity impairment due to arsenic exposure in hippocampal neurons, ways of arsenic exerting on NMDARs, as well as the potential therapeutic targets except for water improvement.
Collapse
Affiliation(s)
- Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University(23618504), Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, China, 150081
| | - Jing Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, National Health Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, Harbin Medical University(23618504), Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, China, 150081.
| |
Collapse
|
23
|
Zhang H, Ben Zablah Y, Zhang H, Liu A, Gugustea R, Lee D, Luo X, Meng Y, Li S, Zhou C, Xin T, Jia Z. Inhibition of Rac1 in ventral hippocampal excitatory neurons improves social recognition memory and synaptic plasticity. Front Aging Neurosci 2022; 14:914491. [PMID: 35936771 PMCID: PMC9354987 DOI: 10.3389/fnagi.2022.914491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/27/2022] [Indexed: 11/28/2022] Open
Abstract
Rac1 is critically involved in the regulation of the actin cytoskeleton, neuronal structure, synaptic plasticity, and memory. Rac1 overactivation is reported in human patients and animal models of Alzheimer’s disease (AD) and contributes to their spatial memory deficits, but whether Rac1 dysregulation is also important in other forms of memory deficits is unknown. In addition, the cell types and synaptic mechanisms involved remain unclear. In this study, we used local injections of AAV virus containing a dominant-negative (DN) Rac1 under the control of CaMKIIα promoter and found that the reduction of Rac1 hyperactivity in ventral hippocampal excitatory neurons improves social recognition memory in APP/PS1 mice. Expression of DN Rac1 also improves long-term potentiation, a key synaptic mechanism for memory formation. Our results suggest that overactivation of Rac1 in hippocampal excitatory neurons contributes to social memory deficits in APP/PS1 mice and that manipulating Rac1 activity may provide a potential therapeutic strategy to treat social deficits in AD.
Collapse
Affiliation(s)
- Haiwang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan, China
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Youssif Ben Zablah
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Haorui Zhang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - An Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Radu Gugustea
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Dongju Lee
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Xiao Luo
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Yanghong Meng
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Song Li
- Department of Neurosurgery, Caoxian People’s Hospital, Caoxian, China
| | - Changxi Zhou
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Beijing, China
- *Correspondence: Changxi Zhou,
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan, China
- Tao Xin,
| | - Zhengping Jia
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Zhengping Jia,
| |
Collapse
|
24
|
Jiang L, Liu C, Zhao B, Ma C, Yin Y, Zhou Q, Xu L, Mao R. Time of Day-Dependent Alteration of Hippocampal Rac1 Activation Regulates Contextual Fear Memory in Rats. Front Mol Neurosci 2022; 15:871679. [PMID: 35782392 PMCID: PMC9245039 DOI: 10.3389/fnmol.2022.871679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/13/2022] [Indexed: 12/03/2022] Open
Abstract
Fear memory in species varies according to the time of the day. Although the underlying molecular mechanisms have been extensively explored, they remain largely unknown. Here, we report that hippocampal Rac1 activity undergoes a time of day-dependent alteration both in nocturnal rats and diurnal tree shrews and that training at the lower hippocampal Rac1 activation period during the night leads to better contextual fear memory in rats. Furthermore, day and night reversion by 24 h darkness/24 h light housing inverses the external clock time of hippocampal Rac1 activation, but the better contextual fear memory still coincides with the lower Rac1 activation in rats during the night. Interestingly, exogenous melatonin treatment promotes hippocampal Rac1 activity and impairs better contextual fear memory acquired at the lower Rac1 activation period during the night, and Rac1-specific inhibitor NSC23766 compromises the effect of melatonin. These results suggest that the time of day-dependent alteration of hippocampal Rac1 activation regulates contextual fear memory in rats by forgetting.
Collapse
Affiliation(s)
- Lizhu Jiang
- CAS Key Laboratory of Animal Models and Human Disease Mechanisms, KIZ-SU Joint Laboratory of Animal Model and Drug Development, Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Department of Clinical Psychology, The Third People’s Hospital of Yunnan Province, Kunming, China
- Department of Neuropsychopathy, Clinical Medical School, Dali University, Dali, China
| | - Chao Liu
- CAS Key Laboratory of Animal Models and Human Disease Mechanisms, KIZ-SU Joint Laboratory of Animal Model and Drug Development, Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Baizhen Zhao
- CAS Key Laboratory of Animal Models and Human Disease Mechanisms, KIZ-SU Joint Laboratory of Animal Model and Drug Development, Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Chen Ma
- CAS Key Laboratory of Animal Models and Human Disease Mechanisms, KIZ-SU Joint Laboratory of Animal Model and Drug Development, Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Yan Yin
- Department of Clinical Psychology, The Third People’s Hospital of Yunnan Province, Kunming, China
| | - Qixin Zhou
- CAS Key Laboratory of Animal Models and Human Disease Mechanisms, KIZ-SU Joint Laboratory of Animal Model and Drug Development, Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Lin Xu
- CAS Key Laboratory of Animal Models and Human Disease Mechanisms, KIZ-SU Joint Laboratory of Animal Model and Drug Development, Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
- CAS Centre for Excellence in Brain Science and Intelligent Technology, Shanghai, China
- *Correspondence: Lin Xu,
| | - RongRong Mao
- Department of Pathology and Pathophysiology, School of Basic Medical Science, Kunming Medical University, Kunming, China
- RongRong Mao,
| |
Collapse
|
25
|
Santana DA, Bedrat A, Puga RD, Turecki G, Mechawar N, Faria TC, Gigek CO, Payão SL, Smith MA, Lemos B, Chen ES. The role of H3K9 acetylation and gene expression in different brain regions of Alzheimer's disease patients. Epigenomics 2022; 14:651-670. [PMID: 35588246 DOI: 10.2217/epi-2022-0096] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To evaluate H3K9 acetylation and gene expression profiles in three brain regions of Alzheimer's disease (AD) patients and elderly controls, and to identify AD region-specific abnormalities. Methods: Brain samples of auditory cortex, hippocampus and cerebellum from AD patients and controls underwent chromatin immunoprecipitation sequencing, RNA sequencing and network analyses. Results: We found a hyperacetylation of AD cerebellum and a slight hypoacetylation of AD hippocampus. The transcriptome revealed differentially expressed genes in the hippocampus and auditory cortex. Network analysis revealed Rho GTPase-mediated mechanisms. Conclusions: These findings suggest that some crucial mechanisms, such as Rho GTPase activity and cytoskeletal organization, are differentially dysregulated in brain regions of AD patients at the epigenetic and transcriptomic levels, and might contribute toward future research on AD pathogenesis.
Collapse
Affiliation(s)
- Daliléia A Santana
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil
| | - Amina Bedrat
- Department of Environmental Health & Molecular & Integrative Physiological Sciences Program, Harvard TH Chan School of Public Health, Boston, MA 02115-5810, USA
| | - Renato D Puga
- Hermes Pardini Institute, São Paulo, SP, 04038-030, Brazil
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Hospital Research Center, McGill University, Montreal, QC, H4H1R3, Canada
| | - Naguib Mechawar
- Department of Psychiatry, Douglas Hospital Research Center, McGill University, Montreal, QC, H4H1R3, Canada
| | - Tathyane C Faria
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil
| | - Carolina O Gigek
- Department of Pathology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, 04023-062, Brazil
| | - Spencer Lm Payão
- Department of Genetics, Blood Center, Faculdade de Medicina de Marília (FAMEMA), Marília, SP, 17519-050, Brazil
| | - Marília Ac Smith
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil
| | - Bernardo Lemos
- Department of Environmental Health & Molecular & Integrative Physiological Sciences Program, Harvard TH Chan School of Public Health, Boston, MA 02115-5810, USA
| | - Elizabeth S Chen
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil.,Department of Environmental Health & Molecular & Integrative Physiological Sciences Program, Harvard TH Chan School of Public Health, Boston, MA 02115-5810, USA
| |
Collapse
|
26
|
Mai Le N, Li J. Ras-related C3 botulinum toxin substrate 1 role in Pathophysiology of Neurological diseases. BRAIN HEMORRHAGES 2022. [DOI: 10.1016/j.hest.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
27
|
Lei B, Lv L, Hu S, Tang Y, Zhong Y. Social experiences switch states of memory engrams through regulating hippocampal Rac1 activity. Proc Natl Acad Sci U S A 2022; 119:e2116844119. [PMID: 35377811 PMCID: PMC9169661 DOI: 10.1073/pnas.2116844119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/06/2022] [Indexed: 11/18/2022] Open
Abstract
In pathological or artificial conditions, memory can be formed as silenced engrams that are unavailable for retrieval by presenting conditioned stimuli but can be artificially switched into the latent state so that natural recall is allowed. However, it remains unclear whether such different states of engrams bear any physiological significance and can be switched through physiological mechanisms. Here, we show that an acute social reward experience switches the silent memory engram into the latent state. Conversely, an acute social stress causes transient forgetting via turning a latent memory engram into a silent state. Such emotion-driven bidirectional switching between latent and silent states of engrams is mediated through regulation of Rac1 activity–dependent reversible forgetting in the hippocampus, as stress-activated Rac1 suppresses retrieval, while reward recovers silenced memory under amnesia by inhibiting Rac1. Thus, data presented reveal hippocampal Rac1 activity as the basis for emotion-mediated switching between latent and silent engrams to achieve emotion-driven behavioral flexibility.
Collapse
Affiliation(s)
- Bo Lei
- School of Life Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
- McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Li Lv
- School of Life Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
- McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, People’s Republic of China
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, People’s Republic of China
- Peking University–Tsinghua University–National Institute Biological Science Joint Graduate Program, Beijing 100084, People's Republic of China
| | - Shiqiang Hu
- School of Life Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
- McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, People’s Republic of China
- Peking University–Tsinghua University–National Institute Biological Science Joint Graduate Program, Beijing 100084, People's Republic of China
| | - Yikai Tang
- School of Life Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
- McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, People’s Republic of China
| | - Yi Zhong
- School of Life Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
- McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, People’s Republic of China
- Peking University–Tsinghua University–National Institute Biological Science Joint Graduate Program, Beijing 100084, People's Republic of China
- Tsinghua–Peking Center for Life Sciences, Beijing 100084, People’s Republic of China
- Ministry of Education Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, People’s Republic of China
| |
Collapse
|
28
|
Ryan TJ, Frankland PW. Forgetting as a form of adaptive engram cell plasticity. Nat Rev Neurosci 2022; 23:173-186. [PMID: 35027710 DOI: 10.1038/s41583-021-00548-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2021] [Indexed: 12/30/2022]
Abstract
One leading hypothesis suggests that memories are stored in ensembles of neurons (or 'engram cells') and that successful recall involves reactivation of these ensembles. A logical extension of this idea is that forgetting occurs when engram cells cannot be reactivated. Forms of 'natural forgetting' vary considerably in terms of their underlying mechanisms, time course and reversibility. However, we suggest that all forms of forgetting involve circuit remodelling that switches engram cells from an accessible state (where they can be reactivated by natural recall cues) to an inaccessible state (where they cannot). In many cases, forgetting rates are modulated by environmental conditions and we therefore propose that forgetting is a form of neuroplasticity that alters engram cell accessibility in a manner that is sensitive to mismatches between expectations and the environment. Moreover, we hypothesize that disease states associated with forgetting may hijack natural forgetting mechanisms, resulting in reduced engram cell accessibility and memory loss.
Collapse
Affiliation(s)
- Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland. .,Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland. .,Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, Victoria, Australia. .,Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada.
| | - Paul W Frankland
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada. .,Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada. .,Department of Psychology, University of Toronto, Toronto, Ontario, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, Canada. .,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Duman JG, Blanco FA, Cronkite CA, Ru Q, Erikson KC, Mulherkar S, Saifullah AB, Firozi K, Tolias KF. Rac-maninoff and Rho-vel: The symphony of Rho-GTPase signaling at excitatory synapses. Small GTPases 2022; 13:14-47. [PMID: 33955328 PMCID: PMC9707551 DOI: 10.1080/21541248.2021.1885264] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.
Collapse
Affiliation(s)
- Joseph G. Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Francisco A. Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. Cronkite
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kelly C. Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
30
|
Kaldun JC, Lone SR, Humbert Camps AM, Fritsch C, Widmer YF, Stein JV, Tomchik SM, Sprecher SG. Dopamine, sleep, and neuronal excitability modulate amyloid-β-mediated forgetting in Drosophila. PLoS Biol 2021; 19:e3001412. [PMID: 34613972 PMCID: PMC8523056 DOI: 10.1371/journal.pbio.3001412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 10/18/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022] Open
Abstract
Alzheimer disease (AD) is one of the main causes of age-related dementia and neurodegeneration. However, the onset of the disease and the mechanisms causing cognitive defects are not well understood. Aggregation of amyloidogenic peptides is a pathological hallmark of AD and is assumed to be a central component of the molecular disease pathways. Pan-neuronal expression of Aβ42Arctic peptides in Drosophila melanogaster results in learning and memory defects. Surprisingly, targeted expression to the mushroom bodies, a center for olfactory memories in the fly brain, does not interfere with learning but accelerates forgetting. We show here that reducing neuronal excitability either by feeding Levetiracetam or silencing of neurons in the involved circuitry ameliorates the phenotype. Furthermore, inhibition of the Rac-regulated forgetting pathway could rescue the Aβ42Arctic-mediated accelerated forgetting phenotype. Similar effects are achieved by increasing sleep, a critical regulator of neuronal homeostasis. Our results provide a functional framework connecting forgetting signaling and sleep, which are critical for regulating neuronal excitability and homeostasis and are therefore a promising mechanism to modulate forgetting caused by toxic Aβ peptides.
Collapse
Affiliation(s)
- Jenifer C. Kaldun
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Shahnaz R. Lone
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Department of Animal Sciences, Central University of Punjab, Bathinda, India
| | | | - Cornelia Fritsch
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Yves F. Widmer
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Jens V. Stein
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Seth M. Tomchik
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Simon G. Sprecher
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- * E-mail:
| |
Collapse
|
31
|
Zhang H, Ben Zablah Y, Zhang H, Jia Z. Rho Signaling in Synaptic Plasticity, Memory, and Brain Disorders. Front Cell Dev Biol 2021; 9:729076. [PMID: 34671600 PMCID: PMC8520953 DOI: 10.3389/fcell.2021.729076] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Memory impairments are associated with many brain disorders such as autism, Alzheimer's disease, and depression. Forming memories involves modifications of synaptic transmission and spine morphology. The Rho family small GTPases are key regulators of synaptic plasticity by affecting various downstream molecules to remodel the actin cytoskeleton. In this paper, we will review recent studies on the roles of Rho proteins in the regulation of hippocampal long-term potentiation (LTP) and long-term depression (LTD), the most extensively studied forms of synaptic plasticity widely regarded as cellular mechanisms for learning and memory. We will also discuss the involvement of Rho signaling in spine morphology, the structural basis of synaptic plasticity and memory formation. Finally, we will review the association between brain disorders and abnormalities of Rho function. It is expected that studying Rho signaling at the synapse will contribute to the understanding of how memory is formed and disrupted in diseases.
Collapse
Affiliation(s)
- Haorui Zhang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Youssif Ben Zablah
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Haiwang Zhang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zhengping Jia
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Walker CK, Herskowitz JH. Dendritic Spines: Mediators of Cognitive Resilience in Aging and Alzheimer's Disease. Neuroscientist 2021; 27:487-505. [PMID: 32812494 PMCID: PMC8130863 DOI: 10.1177/1073858420945964] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cognitive resilience is often defined as the ability to remain cognitively normal in the face of insults to the brain. These insults can include disease pathology, such as plaques and tangles associated with Alzheimer's disease, stroke, traumatic brain injury, or other lesions. Factors such as physical or mental activity and genetics may contribute to cognitive resilience, but the neurobiological underpinnings remain ill-defined. Emerging evidence suggests that dendritic spine structural plasticity is one plausible mechanism. In this review, we highlight the basic structure and function of dendritic spines and discuss how spine density and morphology change in aging and Alzheimer's disease. We note evidence that spine plasticity mediates resilience to stress, and we tackle dendritic spines in the context of cognitive resilience to Alzheimer's disease. Finally, we examine how lifestyle and genetic factors may influence dendritic spine plasticity to promote cognitive resilience before discussing evidence for actin regulatory kinases as therapeutic targets for Alzheimer's disease.
Collapse
Affiliation(s)
- Courtney K. Walker
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Jeremy H. Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| |
Collapse
|
33
|
Cheng KC, Chen YH, Wu CL, Lee WP, Cheung CHA, Chiang HC. Rac1 and Akt Exhibit Distinct Roles in Mediating Aβ-Induced Memory Damage and Learning Impairment. Mol Neurobiol 2021; 58:5224-5238. [PMID: 34273104 DOI: 10.1007/s12035-021-02471-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022]
Abstract
Accumulated beta-amyloid (Aβ) in the brain is the hallmark of Alzheimer's disease (AD). Despite Aβ accumulation is known to trigger cellular dysfunctions and learning and memory damage, the detailed molecular mechanism remains elusive. Recent studies have shown that the onset of memory impairment and learning damage in the AD animal is different, suggesting that the underlying mechanism of the development of memory impairment and learning damage may not be the same. In the current study, with the use of Aβ42 transgenic flies as models, we found that Aβ induces memory damage and learning impairment via differential molecular signaling pathways. In early stage, Aβ activates both Ras and PI3K to regulate Rac1 activity, which affects mostly on memory performance. In later stage, PI3K-Akt is strongly activated by Aβ, which leads to learning damage. Moreover, reduced Akt, but not Rac1, activity promotes cell viability in the Aβ42 transgenic flies, indicating that Akt and Rac1 exhibit differential roles in Aβ regulating toxicity. Taken together, different molecular and cellular mechanisms are involved in Aβ-induced learning damage and memory decline; thus, caution should be taken during the development of therapeutic intervention in the future.
Collapse
Affiliation(s)
- Kuan-Chung Cheng
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Ying-Hao Chen
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wang-Pao Lee
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun Hei Antonio Cheung
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Hsueh-Cheng Chiang
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.
| |
Collapse
|
34
|
Wang Z, Xia P, Hu J, Huang Y, Zhang F, Li L, Wang E, Guo Q, Ye Z. LncRNA MEG3 Alleviates Diabetic Cognitive Impairments by Reducing Mitochondrial-Derived Apoptosis through Promotion of FUNDC1-Related Mitophagy via Rac1-ROS Axis. ACS Chem Neurosci 2021; 12:2280-2307. [PMID: 33843209 DOI: 10.1021/acschemneuro.0c00682] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction and elevated ROS generation are predominant contributors of neuronal death that is responsible for the diabetes-related cognitive impairments. Emerging evidence has demonstrated that long noncoding RNA-MEG3 can serve as an important regulator in the pathogenesis of diabetes. However, the underlying mechanisms remain to be further clarified. Here, it was observed that MEG3 was significantly down-regulated in STZ (streptozotocin)-induced diabetic rats. MEG3 overexpression noticeably improved diabetes-induced cognitive dysfunctions, accompanied by the abatement of Rac1 activation and ROS production, as well as the inhibition of mitochondria-associated apoptosis. Furthermore, either MEG3 overexpression or Rac1 inhibition promoted FUNDC1 dephosphorylation and suppressed oxidative stress and neuro-inflammation. Similarly, in vitro studies confirmed that hyperglycemia also down-regulated MEG3 expression in PC12 cells. MEG3 reintroduction protected PC12 cells against hyperglycemia-triggered neurotoxicity by improving mitochondrial fitness and repressing mitochondria-mediated apoptosis. Moreover, these neuroprotective effects of MEG3 relied on FUNDC1-related mitophagy, since silencing of FUNDC1 abolished these beneficial outcomes. Additionally, MEG3 rescued HG-induced neurotoxicity was involved in inhibiting Rac1 expression via interaction with Rac1 3'UTR. Conversely, knockdown of MEG3 showed opposite effects. NSC23766, a specific inhibitor of Rac1, fully abolished harmful effects of MEG3 depletion. Consistently, knockdown of Rac1 potentiated FUNDC1-associated mitophagy. Meanwhile, colocalization of Rac1 and FUNDC1 was found in mitochondria under hyperglycemia, which was interrupted by MEG3 overexpression. Furthermore, silencing of Rac1 promoted PGAM5 expression, and FUNDC1 strongly interacted with LC3 in Rac1-deleted cells. Altogether, our findings suggested that the Rac1/ROS axis may be a downstream signaling pathway for MEG3-induced neuroprotection, which was involved in FUNDC1-associated mitophagy.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Anesthesiology, Hainan General Hospital, Haikou 570311, China
| | - Pingping Xia
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Jie Hu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Yan Huang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
| | - Fan Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha 410008, China
| |
Collapse
|
35
|
Zhang F, Liu Y, You Q, Yang E, Liu B, Wang H, Xu S, Nawaz W, Chen D, Wu Z. NSC23766 and Ehop016 Suppress Herpes Simplex Virus-1 Replication by Inhibiting Rac1 Activity. Biol Pharm Bull 2021; 44:1263-1271. [PMID: 34162786 DOI: 10.1248/bpb.b21-00054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Herpes simplex virus-1 (HSV-1) infection of the eyes leads to herpes simplex virus keratitis (HSK), the main cause of infectious blindness in the world. As the current therapeutics for HSV-1 infection are rather limited and prolonged use of acyclovir (ACV)/ganciclovir (GCV) and in immunocompromised patients lead to the rise of drug resistant mutants, it underlines the urgent need for new antiviral agents with distinct mechanisms. Our study attempted to establish ras-related C3 botulinum toxin substrate 1 (Rac1) as a new therapeutic target for HSV-1 infection by using Rac1-specific inhibitors to evaluate the in vitro inhibition of virus growth. Our results showed that increased Rac1 activity facilitated HSV-1 replication and inhibition of Rac1 activity by NSC23766 and Ehop016 significantly reduced HSV-1 replication. Thus, we identified NSC23766 and Ehop016 as possessing potent anti-HSV-1 activities by suppressing the Rac1 activity, suggesting that Rac1 is a potential target for treating HSV-1-related diseases.
Collapse
Affiliation(s)
- Fang Zhang
- Center for Public Health Research, Medical School of Nanjing University.,Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University
| | - Ye Liu
- Center for Public Health Research, Medical School of Nanjing University.,Department of Ophthalmology, JinLing Hospital, Medical School of Nanjing University
| | - Qiao You
- Center for Public Health Research, Medical School of Nanjing University
| | - Enhui Yang
- Nanjing Children's Hospital, Nanjing Medical University
| | - Bingxin Liu
- Center for Public Health Research, Medical School of Nanjing University
| | - Huanru Wang
- Center for Public Health Research, Medical School of Nanjing University
| | - Shijie Xu
- Center for Public Health Research, Medical School of Nanjing University
| | - Waqas Nawaz
- Center for Public Health Research, Medical School of Nanjing University.,Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University
| | - Deyan Chen
- Center for Public Health Research, Medical School of Nanjing University
| | - Zhiwei Wu
- Center for Public Health Research, Medical School of Nanjing University.,Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University.,School of Life Sciences, Ningxia University
| |
Collapse
|
36
|
ABCC1 regulates cocaine-associated memory, spine plasticity and GluA1 and GluA2 surface expression. Neuroreport 2021; 32:833-839. [PMID: 34029289 DOI: 10.1097/wnr.0000000000001657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
ATP-binding cassettes C1 (ABCC1s) are expressed in the neurons of the brain, but their function in neurological diseases is far from clear. In this study, we investigated the role of ABCC1 in the hippocampus in cocaine-associated memory and spine plasticity. We also investigated the role of ABCC1 in AMPA receptors (AMPARs) surface expression in primary prefrontal cortex (PFC) neurons following dopamine treatment, which was used to mimic exposure to cocaine. We found that cocaine increased ABCC1 expression in the hippocampus, and ABCC1-siRNA blocked cocaine-induced place preference. Furthermore, a morphological study showed that ABCC1-siRNA reduced the total spine density, including thin, stubby and mushroom spines in both cocaine and basal treatments compared with controls. Meanwhile, in vitro tests showed that ABCC1-siRNA decreased GluA1 and GluA2 surface expression induced by dopamine, while a decreased number of synapses in primary PFC neurons was observed following dopamine treatment. The data show that ABCC1 in the hippocampus is critically involved in cocaine-associated memory and spine plasticity and that dopamine induces AMPARs surface expression in primary PFC neurons. ABCC1 is thus presented as a new signaling molecule involved in cocaine addiction, which may provide a new target for the treatment of cocaine addiction.
Collapse
|
37
|
Ji B, Skup M. Roles of palmitoylation in structural long-term synaptic plasticity. Mol Brain 2021; 14:8. [PMID: 33430908 PMCID: PMC7802216 DOI: 10.1186/s13041-020-00717-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are important cellular mechanisms underlying learning and memory processes. N-Methyl-d-aspartate receptor (NMDAR)-dependent LTP and LTD play especially crucial roles in these functions, and their expression depends on changes in the number and single channel conductance of the major ionotropic glutamate receptor α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) located on the postsynaptic membrane. Structural changes in dendritic spines comprise the morphological platform and support for molecular changes in the execution of synaptic plasticity and memory storage. At the molecular level, spine morphology is directly determined by actin cytoskeleton organization within the spine and indirectly stabilized and consolidated by scaffold proteins at the spine head. Palmitoylation, as a uniquely reversible lipid modification with the ability to regulate protein membrane localization and trafficking, plays significant roles in the structural and functional regulation of LTP and LTD. Altered structural plasticity of dendritic spines is also considered a hallmark of neurodevelopmental disorders, while genetic evidence strongly links abnormal brain function to impaired palmitoylation. Numerous studies have indicated that palmitoylation contributes to morphological spine modifications. In this review, we have gathered data showing that the regulatory proteins that modulate the actin network and scaffold proteins related to AMPAR-mediated neurotransmission also undergo palmitoylation and play roles in modifying spine architecture during structural plasticity.
Collapse
Affiliation(s)
- Benjun Ji
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| | - Małgorzata Skup
- Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland.
| |
Collapse
|
38
|
Zhang H, Karisetty BC, Bhatnagar A, Armour EM, Beaver M, Roach TV, Mortazavi S, Mandloi S, Elefant F. Tip60 protects against amyloid-β-induced transcriptomic alterations via different modes of action in early versus late stages of neurodegeneration. Mol Cell Neurosci 2020; 109:103570. [PMID: 33160016 DOI: 10.1016/j.mcn.2020.103570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/24/2020] [Accepted: 10/31/2020] [Indexed: 10/23/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder hallmarked by amyloid-β (Aβ) plaque accumulation, neuronal cell death, and cognitive deficits that worsen during disease progression. Histone acetylation dysregulation, caused by an imbalance between reduced histone acetyltransferases (HAT) Tip60 and increased histone deacetylase 2 (HDAC2) levels, can directly contribute to AD pathology. However, whether such AD-associated neuroepigenetic alterations occur in response to Aβ peptide production and can be protected against by increasing Tip60 levels over the course of neurodegenerative progression remains unknown. Here we profile Tip60 HAT/HDAC2 dynamics and transcriptome-wide changes across early and late stage AD pathology in the Drosophila brain produced solely by human amyloid-β42. We show that early Aβ42 induction leads to disruption of Tip60 HAT/HDAC2 balance during early neurodegenerative stages preceding Aβ plaque accumulation that persists into late AD stages. Correlative transcriptome-wide studies reveal alterations in biological processes we classified as transient (early-stage only), late-onset (late-stage only), and constant (both). Increasing Tip60 HAT levels in the Aβ42 fly brain protects against AD functional pathologies that include Aβ plaque accumulation, neural cell death, cognitive deficits, and shorter life-span. Strikingly, Tip60 protects against Aβ42-induced transcriptomic alterations via distinct mechanisms during early and late stages of neurodegeneration. Our findings reveal distinct modes of neuroepigenetic gene changes and Tip60 neuroprotection in early versus late stages in AD that can serve as early biomarkers for AD, and support the therapeutic potential of Tip60 over the course of AD progression.
Collapse
Affiliation(s)
- Haolin Zhang
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | | | - Akanksha Bhatnagar
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Ellen M Armour
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Mariah Beaver
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Tiffany V Roach
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Sina Mortazavi
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Shreya Mandloi
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Felice Elefant
- Department of Biology, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
39
|
Lu W, Lin J, Zheng D, Hong C, Ke L, Wu X, Chen P. Overexpression of MicroRNA-133a Inhibits Apoptosis and Autophagy in a Cell Model of Parkinson's Disease by Downregulating Ras-Related C3 Botulinum Toxin Substrate 1 (RAC1). Med Sci Monit 2020; 26:e922032. [PMID: 32713934 PMCID: PMC7409387 DOI: 10.12659/msm.922032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Parkinson’s disease (PD) is a movement disorder. microRNA (miR)-133 expression is reduced in PD patients and in mice with a dopamine neuron deficiency. We aimed to identify the mechanism of miR-133a in apoptosis and autophagy in PD. Material/Methods The optimal concentration of MPP+ (1-methyl-4-phenylpyridinium ion) was initially determined to construct a PD cell model. Gain-of function experiments were carried out to evaluate the role of miR-133a in PD. The levels of miR-133a, RAC1 (Ras-related C3 botulinum toxin substrate 1), apoptosis-related factors, and autophagy-related factors were detected after detection of cell proliferation, cell cycle, and apoptosis. Transmission electron microscopy was applied to observe autophagosomes, and immunofluorescence staining was performed to detect LC3 and further analyze the effect of miR-133a on autophagy in a PD cell model. Results Low miR-133a expression was detected in a cell model of MPP+-induced PD. After overexpressing miR-133a, cell proliferation increased, and apoptosis (cleaved caspase-3 and Bax levels decreased, while Bcl2 levels increased) and autophagy was inhibited (LC3II/I and Beclin-1 levels decreased, while p62 levels increased). MiR-133a targeted RAC1. RACY upregulation attenuated the inhibitory effects of miR-133a on PC12 cell apoptosis and autophagy. Conclusions Our data highlighted that miR-133a overexpression prevented apoptosis and autophagy in a cell model of MPP+-induced PD by inhibiting RAC1 expression.
Collapse
Affiliation(s)
- Wusheng Lu
- Department of Neurology, The 909th Hospital of (People's Liberation Army) PLA, Zhangzhou, Fujian, China (mainland)
| | - Jinhuang Lin
- Department of Neurology, The 909th Hospital of (People's Liberation Army) PLA, Zhangzhou, Fujian, China (mainland)
| | - Dequan Zheng
- Department of Neurology, The 909th Hospital of (People's Liberation Army) PLA, Zhangzhou, Fujian, China (mainland)
| | - Chunyong Hong
- Department of Neurology, The 909th Hospital of (People's Liberation Army) PLA, Zhangzhou, Fujian, China (mainland)
| | - Laishun Ke
- Department of Neurology, The 909th Hospital of (People's Liberation Army) PLA, Zhangzhou, Fujian, China (mainland)
| | - Xinyu Wu
- Department of Neurology, The 909th Hospital of (People's Liberation Army) PLA, Zhangzhou, Fujian, China (mainland)
| | - Peineng Chen
- Department of Neurology, The 909th Hospital of (People's Liberation Army) PLA, Zhangzhou, Fujian, China (mainland)
| |
Collapse
|
40
|
Moreno A. Molecular mechanisms of forgetting. Eur J Neurosci 2020; 54:6912-6932. [DOI: 10.1111/ejn.14839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Andrea Moreno
- Danish Institute of Translational Neuroscience (DANDRITE) Aarhus University Aarhus C Denmark
| |
Collapse
|
41
|
Teravskis PJ, Ashe KH, Liao D. The Accumulation of Tau in Postsynaptic Structures: A Common Feature in Multiple Neurodegenerative Diseases? Neuroscientist 2020; 26:503-520. [PMID: 32389059 DOI: 10.1177/1073858420916696] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increasingly, research suggests that neurodegenerative diseases and dementias are caused not by unique, solitary cellular mechanisms, but by multiple contributory mechanisms manifesting as heterogeneous clinical presentations. However, diverse neurodegenerative diseases also share common pathological hallmarks and cellular mechanisms. One such mechanism involves the redistribution of the microtubule associated protein tau from the axon into the somatodendritic compartment of neurons, followed by the mislocalization of tau into dendritic spines, resulting in postsynaptic functional deficits. Here we review various signaling pathways that trigger the redistribution of tau to the cell body and dendritic tree, and its mislocalization to dendritic spines. The convergence of multiple pathways in different disease models onto this final common pathway suggests that it may be an attractive pathway to target for developing new treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter J Teravskis
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.,University of Minnesota Medical School, Minneapolis, MN, USA
| | - Karen H Ashe
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Budd Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA.,Geriatric Research Education and Clinical Center, Veterans Affairs Medical Center, Minneapolis, MN, USA
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
42
|
Costa JF, Dines M, Lamprecht R. The Role of Rac GTPase in Dendritic Spine Morphogenesis and Memory. Front Synaptic Neurosci 2020; 12:12. [PMID: 32362820 PMCID: PMC7182350 DOI: 10.3389/fnsyn.2020.00012] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/04/2020] [Indexed: 11/21/2022] Open
Abstract
The ability to form memories in the brain is needed for daily functions, and its impairment is associated with human mental disorders. Evidence indicates that long-term memory (LTM)-related processes such as its consolidation, extinction and forgetting involve changes of synaptic efficacy produced by alterations in neural transmission and morphology. Modulation of the morphology and number of dendritic spines has been proposed to contribute to changes in neuronal transmission mediating such LTM-related processes. Rac GTPase activity is regulated by synaptic activation and it can affect spine morphology by controlling actin-regulatory proteins. Recent evidence shows that changes in Rac GTPase activity affect memory consolidation, extinction, erasure and forgetting and can affect spine morphology in brain areas that mediate these behaviors. Altered Rac GTPase activity is associated with abnormal spine morphology and brain disorders. By affecting Rac GTPase activity we can further understand the roles of spine morphogenesis in memory. Moreover, manipulation of Rac GTPase activity may serve as a therapeutic tool for the treatment of memory-related brain diseases.
Collapse
Affiliation(s)
| | | | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
43
|
Genetic Dissection of Alzheimer's Disease Using Drosophila Models. Int J Mol Sci 2020; 21:ijms21030884. [PMID: 32019113 PMCID: PMC7037931 DOI: 10.3390/ijms21030884] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 01/26/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD), a main cause of dementia, is the most common neurodegenerative disease that is related to abnormal accumulation of the amyloid β (Aβ) protein. Despite decades of intensive research, the mechanisms underlying AD remain elusive, and the only available treatment remains symptomatic. Molecular understanding of the pathogenesis and progression of AD is necessary to develop disease-modifying treatment. Drosophila, as the most advanced genetic model, has been used to explore the molecular mechanisms of AD in the last few decades. Here, we introduce Drosophila AD models based on human Aβ and summarize the results of their genetic dissection. We also discuss the utility of functional genomics using the Drosophila system in the search for AD-associated molecular mechanisms in the post-genomic era.
Collapse
|