1
|
Gao YH, Wen DT, Du ZR, Wang JF, Wang SJ. Muscle Psn gene combined with exercise contribute to healthy aging of skeletal muscle and lifespan by adaptively regulating Sirt1/PGC-1α and arm pathway. PLoS One 2024; 19:e0300787. [PMID: 38753634 PMCID: PMC11098322 DOI: 10.1371/journal.pone.0300787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/05/2024] [Indexed: 05/18/2024] Open
Abstract
The Presenilin (Psn) gene is closely related to aging, but it is still unclear the role of Psn genes in skeletal muscle. Here, the Psn-UAS/Mhc-GAL4 system in Drosophila was used to regulate muscle Psn overexpression(MPO) and muscle Psn knockdown(MPK). Drosophila were subjected to endurance exercise from 4 weeks to 5 weeks old. The results showed that MPO and exercise significantly increased climbing speed, climbing endurance, lifespan, muscle SOD activity, Psn expression, Sirt1 expression, PGC-1α expression, and armadillo (arm) expression in aged Drosophila, and they significantly decreased muscle malondialdehyde levels. Interestingly, when the Psn gene is knockdown by 0.78 times, the PGC-1α expression and arm expression were also down-regulated, but the exercise capacity and lifespan were increased. Furthermore, exercise combined with MPO further improved the exercise capacity and lifespan. MPK combined with exercise further improves the exercise capacity and lifespan. Thus, current results confirmed that the muscle Psn gene was a vital gene that contributed to the healthy aging of skeletal muscle since whether it was overexpressed or knocked down, the aging progress of skeletal muscle structure and function was slowed down by regulating the activity homeostasis of Sirt1/PGC-1α pathway and Psn/arm pathway. Exercise enhanced the function of the Psn gene to delay skeletal muscle aging by up regulating the activity of the Sirt1/PGC-1α pathway and Psn/arm pathway.
Collapse
Affiliation(s)
- Ying-hui Gao
- Ludong University, City Yantai, Shandong Province, China
| | - Deng-tai Wen
- Ludong University, City Yantai, Shandong Province, China
| | - Zhong-rui Du
- Ludong University, City Yantai, Shandong Province, China
| | - Jing-feng Wang
- Ludong University, City Yantai, Shandong Province, China
| | - Shi-jie Wang
- Ludong University, City Yantai, Shandong Province, China
| |
Collapse
|
2
|
Identification of a Novel Wnt Antagonist Based Therapeutic and Diagnostic Target for Alzheimer's Disease Using a Stem Cell-Derived Model. Bioengineering (Basel) 2023; 10:bioengineering10020192. [PMID: 36829686 PMCID: PMC9952699 DOI: 10.3390/bioengineering10020192] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 02/05/2023] Open
Abstract
Currently, all the existing treatments for Alzheimer's disease (AD) fail to stall progression due to longer duration of time between onset of the symptoms and diagnosis of the disease, raising the necessity of effective diagnostics and novel treatment. Specific molecular regulation of the onset and progression of disease is not yet elucidated. This warranted investigation of the role of Wnt signaling regulators which are thought to be involved in neurogenesis. The AD model was established using amyloid beta (Aβ) in human mesenchymal stem cells derived from amniotic membranes which were differentiated into neuronal cell types. In vivo studies were carried out with Aβ or a Wnt antagonist, AD201, belonging to the sFRP family. We further created an AD201-knockdown in vitro model to determine the role of Wnt antagonism. BACE1 upregulation, ChAT and α7nAChR downregulation with synapse and functionality loss with increases in ROS confirmed the neurodegeneration. Reduced β-catenin and increased AD201 expression indicated Wnt/canonical pathway inhibition. Similar results were exhibited in the in vivo study along with AD-associated behavioural and molecular changes. AD201-knockdown rescued neurons from Aβ-induced toxicity. We demonstrated for the first time a role of AD201 in Alzheimer's disease manifestation, which indicates a promising disease target and biomarker.
Collapse
|
3
|
Hogestyn JM, Salois G, Xie L, Apa C, Youngyunpipatkul J, Pröschel C, Mayer-Pröschel M. Expression of the human herpesvirus 6A latency-associated transcript U94A impairs cytoskeletal functions in human neural cells. Mol Cell Neurosci 2022; 123:103770. [PMID: 36055520 PMCID: PMC10124163 DOI: 10.1016/j.mcn.2022.103770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/12/2022] [Accepted: 08/20/2022] [Indexed: 12/30/2022] Open
Abstract
Many neurodegenerative diseases have a multifactorial etiology and variable course of progression that cannot be explained by current models. Neurotropic viruses have long been suggested to play a role in these diseases, although their exact contributions remain unclear. Human herpesvirus 6A (HHV-6A) is one of the most common viruses detected in the adult brain, and has been clinically associated with multiple sclerosis (MS), and, more recently, Alzheimer's disease (AD). HHV-6A is a ubiquitous viral pathogen capable of infecting glia and neurons. Primary infection in childhood is followed by the induction of latency, characterized by expression of the U94A viral transcript in the absence of viral replication. Here we examine the effects of U94A on cells of the central nervous system. We found that U94A expression inhibits the migration and impairs cytoplasmic maturation of human oligodendrocyte precursor cells (OPCs) without affecting their viability, a phenotype that may contribute to the failure of remyelination seen in many patients with MS. A subsequent proteomics analysis of U94A expression OPCs revealed altered expression of genes involved in tubulin associated cytoskeletal regulation. As HHV-6A seems to significantly be associated with early AD pathology, we extended our initially analysis of the impact of U94A on human derived neurons. We found that U94A expression inhibits neurite outgrowth of primary human cortical neurons and impairs synapse maturation. Based on these data we suggest that U94A expression by latent HHV-6A in glial cells and neurons renders them susceptible to dysfunction and degeneration. Therefore, latent viral infections of the brain represent a unique pathological risk factor that may contribute to disease processes.
Collapse
Affiliation(s)
- Jessica M Hogestyn
- Department of Biomedical Genetics, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA; Department of Neuroscience, School of Medicine and Dentistry, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA
| | - Garrick Salois
- Department of Biomedical Genetics, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA; Department of Neuroscience, School of Medicine and Dentistry, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA
| | - Li Xie
- Department of Biomedical Genetics, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA
| | - Connor Apa
- Department of Biomedical Genetics, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA; Stem cell and Regenerative Medicine Institute, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA
| | - Justin Youngyunpipatkul
- Department of Biomedical Genetics, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA
| | - Christoph Pröschel
- Department of Biomedical Genetics, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA; Stem cell and Regenerative Medicine Institute, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA
| | - Margot Mayer-Pröschel
- Department of Biomedical Genetics, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA; Department of Neuroscience, School of Medicine and Dentistry, 601 Elmwood Avenue, Box 633, University of Rochester, Rochester, NY 14642, USA,.
| |
Collapse
|
4
|
Ayabe T, Takahashi C, Ohya R, Ano Y. β-Lactolin improves mitochondrial function in Aβ-treated mouse hippocampal neuronal cell line and a human iPSC-derived neuronal cell model of Alzheimer's disease. FASEB J 2022; 36:e22277. [PMID: 35319792 DOI: 10.1096/fj.202101366rr] [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: 08/27/2021] [Revised: 03/06/2022] [Accepted: 03/14/2022] [Indexed: 11/11/2022]
Abstract
Mitochondrial dysfunctions are a key hallmark of Alzheimer's disease (AD). β-Lactolin, a whey-derived glycine-threonine-tryptophan-tyrosine tetrapeptide, has been previously reported to prevent AD-like pathologies in an AD mouse model via regulation of microglial functions. However, the direct effect of β-lactolin on neuronal cells and neuronal mitochondrial functions remains unknown. Here, we investigated the effects of β-lactolin on mitochondrial functions in amyloid β (Aβ)-treated mouse hippocampal neuronal HT22 cells and human induced-pluripotent cell (hiPSC)-derived AD model neurons. Adding β-lactolin to Aβ-treated HT22 cells increased both the oxygen consumption rate and cellular ATP concentrations, suggesting that β-lactolin improves mitochondrial respiration and energy production. Using high content image analysis, we found that β-lactolin improved mitochondrial fragmentation, membrane potential, and oxidative stress in Aβ-treated cells, eventually preventing neuronal cell death. From a mechanistic perspective, we found that β-lactolin increased gene expression of mitofusin-2, which contributes to mitochondrial fusion events. Finally, we showed that β-lactolin improves both mitochondrial morphologies and membrane potentials in hiPSC-derived AD model neurons. Taken together, β-lactolin improved mitochondrial functions AD-related neuronal cell models and prevented neuronal cell death. The dual function of β-lactolin on both neuron and microglia marks an advantage in maintaining neuronal health.
Collapse
Affiliation(s)
- Tatsuhiro Ayabe
- Kirin Central Research Institute, Kirin Holdings Company Limited, Fujisawa, Japan
| | - Chika Takahashi
- Kirin Central Research Institute, Kirin Holdings Company Limited, Fujisawa, Japan
| | - Rena Ohya
- Kirin Central Research Institute, Kirin Holdings Company Limited, Fujisawa, Japan
| | - Yasuhisa Ano
- Kirin Central Research Institute, Kirin Holdings Company Limited, Fujisawa, Japan
| |
Collapse
|
5
|
Xiong LL, Qiu DL, Xiu GH, Al-Hawwas M, Jiang Y, Wang YC, Hu Y, Chen L, Xia QJ, Wang TH. DPYSL2 is a novel regulator for neural stem cell differentiation in rats: revealed by Panax notoginseng saponin administration. Stem Cell Res Ther 2020; 11:155. [PMID: 32299503 PMCID: PMC7164273 DOI: 10.1186/s13287-020-01652-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/04/2020] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The limited neuronal differentiation of the endogenous or grafted neural stem cells (NSCs) after brain injury hampers the clinic usage of NSCs. Panax notoginseng saponins (PNS) were extensively used for their clinical value, such as in controlling blood pressure, blood glucose, and inhibiting neuronal apoptosis and enhancing neuronal protection, but whether or not it exerts an effect in promoting neuronal differentiation of the endogenous NSCs is completely unclear and the potential underlying mechanism requires further exploration. METHODS Firstly, we determined whether PNS could successfully induce NSCs to differentiate to neurons under the serum condition. Mass spectrometry and quantitative polymerase chain reaction (Q-PCR) were then performed to screen the differentially expressed proteins (genes) between the PNS + serum and serum control group, upon which dihydropyrimidinase-like 2 (DPYSL2), a possible candidate, was then selected for the subsequent research. To further investigate the actual role of DPYSL2 in the NSC differentiation, DPYSL2-expressing lentivirus was employed to obtain DPYSL2 overexpression in NSCs. DPYSL2-knockout rats were constructed to study its effects on hippocampal neural stem cells. Immunofluorescent staining was performed to identify the differentiation direction of NSCs after 7 days from DPYSL2 transfection, as well as those from DPYSL2-knockout rats. RESULTS Seven differentially expressed protein spots were detected by PD Quest, and DPYSL2 was found as one of the key factors of NSC differentiation in a PNS-treated condition. The results of immunostaining further showed that mainly Tuj1 and GFAP-positive cells increased in the DPYSL2-overexpressed group, while both were depressed in the hippocampal NSCs in the DPYSL2-knockout rat. CONCLUSIONS The present study revealed that the differentiation direction of NSCs could be enhanced through PNS administration, and the DPYSL2 is a key regulator in promoting NSC differentiation. These results not only emphasized the effect of PNS but also indicated DPYSL2 could be a novel target to enhance the NSC differentiation in future clinical trials.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - De-Lu Qiu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guang-Hui Xiu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mohammed Al-Hawwas
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Ya Jiang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650031, China
| | - You-Cui Wang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Hu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Chen
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing-Jie Xia
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Institute of Neuroscience, Kunming Medical University, Kunming, 650031, China.
| |
Collapse
|
6
|
A Large Panel of Isogenic APP and PSEN1 Mutant Human iPSC Neurons Reveals Shared Endosomal Abnormalities Mediated by APP β-CTFs, Not Aβ. Neuron 2019; 104:256-270.e5. [PMID: 31416668 DOI: 10.1016/j.neuron.2019.07.010] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 05/30/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022]
Abstract
Familial Alzheimer's disease (fAD) results from mutations in the amyloid precursor protein (APP) and presenilin (PSEN1 and PSEN2) genes. Here we leveraged recent advances in induced pluripotent stem cell (iPSC) and CRISPR/Cas9 genome editing technologies to generate a panel of isogenic knockin human iPSC lines carrying APP and/or PSEN1 mutations. Global transcriptomic and translatomic profiling revealed that fAD mutations have overlapping effects on the expression of AD-related and endocytosis-associated genes. Mutant neurons also increased Rab5+ early endosome size. APP and PSEN1 mutations had discordant effects on Aβ production but similar effects on APP β C-terminal fragments (β-CTFs), which accumulate in all mutant neurons. Importantly, endosomal dysfunction correlated with accumulation of β-CTFs, not Aβ, and could be rescued by pharmacological modulation of β-secretase (BACE). These data display the utility of our mutant iPSCs in studying AD-related phenotypes in a non-overexpression human-based system and support mounting evidence that β-CTF may be critical in AD pathogenesis.
Collapse
|
7
|
Kaindl J, Winner B. Disease Modeling of Neuropsychiatric Brain Disorders Using Human Stem Cell-Based Neural Models. Curr Top Behav Neurosci 2019; 42:159-183. [PMID: 31407242 DOI: 10.1007/7854_2019_111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Human pluripotent stem (PS) cells are a relevant platform to model human-specific neurological disorders. In this chapter, we focus on human stem cell models for neuropsychiatric disorders including induced pluripotent stem (iPS) cell-derived neural precursor cells (NPCs), neurons and cerebral organoids. We discuss crucial steps for planning human disease modeling experiments. We introduce the different strategies of human disease modeling including transdifferentiation, human embryonic stem (ES) cell-based models, iPS cell-based models and genome editing options. Analysis of disease-relevant phenotypes is discussed. In more detail, we provide exemplary insight into modeling of the neurodevelopmental defects in autism spectrum disorder (ASD) and the process of neurodegeneration in Alzheimer's disease (AD). Besides monogenic diseases, iPS cell-derived models also generated data from idiopathic and sporadic cases.
Collapse
Affiliation(s)
- Johanna Kaindl
- Department of Stem Cell Biology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
8
|
Human Neurospheroid Arrays for In Vitro Studies of Alzheimer's Disease. Sci Rep 2018; 8:2450. [PMID: 29402979 PMCID: PMC5799361 DOI: 10.1038/s41598-018-20436-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/17/2018] [Indexed: 01/11/2023] Open
Abstract
Neurospheroids are commonly used for in vitro disease modeling and drug screening. However, the heterogeneity in size of the neurospheroids mixtures available through current methods limits their utility when employed for basic mechanistic studies of neurodegenerative diseases or screening for new interventions. Here, we generate neurospheroids from immortalized neural progenitor cells and human induced pluripotent stem cells that are uniform in size, into large-scale arrays. In proof of concept experiments, we validate the neurospheroids array as a sensitive and robust tool for screening compounds over extended time. We show that when suspended in three-dimensional extracellular matrix up to several weeks, the stem cell-derived neurospheroids display extensive neurite outgrowth and extend thick bundles of dendrites outward. We also cultivate genetically-engineered stem cell-derived neurospheroids with familial Alzheimer's disease mutations for eight weeks in our microarray system. Interestingly, we observed robust accumulation of amyloid-β and phosphorylated tau, key hallmarks of Alzheimer's disease. Overall, our in vitro model for engineering neurospheroid arrays is a valuable tool for studying complex neurodegenerative diseases and accelerating drug discovery.
Collapse
|
9
|
Stanga S, Vrancx C, Tasiaux B, Marinangeli C, Karlström H, Kienlen-Campard P. Specificity of presenilin-1- and presenilin-2-dependent γ-secretases towards substrate processing. J Cell Mol Med 2017; 22:823-833. [PMID: 28994238 PMCID: PMC5783875 DOI: 10.1111/jcmm.13364] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/27/2017] [Indexed: 12/20/2022] Open
Abstract
The two presenilin‐1 (PS1) and presenilin‐2 (PS2) homologs are the catalytic core of the γ‐secretase complex, which has a major role in cell fate decision and Alzheimer's disease (AD) progression. Understanding the precise contribution of PS1‐ and PS2‐dependent γ‐secretases to the production of β‐amyloid peptide (Aβ) from amyloid precursor protein (APP) remains an important challenge to design molecules efficiently modulating Aβ release without affecting the processing of other γ‐secretase substrates. To that end, we studied PS1‐ and PS2‐dependent substrate processing in murine cells lacking presenilins (PSs) (PS1KO, PS2KO or PS1‐PS2 double‐KO noted PSdKO) or stably re‐expressing human PS1 or PS2 in an endogenous PS‐null (PSdKO) background. We characterized the processing of APP and Notch on both endogenous and exogenous substrates, and we investigated the effect of pharmacological inhibitors targeting the PSs activity (DAPT and L‐685,458). We found that murine PS1 γ‐secretase plays a predominant role in APP and Notch processing when compared to murine PS2 γ‐secretase. The inhibitors blocked more efficiently murine PS2‐ than murine PS1‐dependent processing. Human PSs, especially human PS1, expression in a PS‐null background efficiently restored APP and Notch processing. Strikingly, and contrary to the results obtained on murine PSs, pharmacological inhibitors appear to preferentially target human PS1‐ than human PS2‐dependent γ‐secretase activity.
Collapse
Affiliation(s)
- Serena Stanga
- Alzheimer Research group, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Céline Vrancx
- Alzheimer Research group, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Bernadette Tasiaux
- Alzheimer Research group, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Claudia Marinangeli
- Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT, University of Lille, Lille, France
| | - Helena Karlström
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Pascal Kienlen-Campard
- Alzheimer Research group, Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
10
|
Dan Q, Xia Q, Wang T. Interference chemically synthesized dsRNA decreased IL‐1β expression in PC12 cells and its functional implication. IBRAIN 2017. [DOI: 10.1002/j.2769-2795.2017.tb00014.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Qi‐Qin Dan
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduChina
| | - Qing‐Jie Xia
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduChina
| | - Ting‐Hua Wang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
11
|
Nishioka H, Tooi N, Isobe T, Nakatsuji N, Aiba K. BMS-708163 and Nilotinib restore synaptic dysfunction in human embryonic stem cell-derived Alzheimer's disease models. Sci Rep 2016; 6:33427. [PMID: 27641902 PMCID: PMC5027582 DOI: 10.1038/srep33427] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 08/26/2016] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. Cellular AD models derived from human pluripotent stem cells are promising tools in AD research. We recently developed human embryonic stem cell-derived AD models which overexpress mutant Presenilin1 genes, and which exhibit AD phenotypes, including synaptic dysfunction. In this study, we found that our AD models showed reduced levels of RAB3A and SV2B proteins in the pre-synapses, which is a possible cause of electrophysiological abnormalities. Through the screening of chemical compounds using our AD models, we have identified Aβ peptide inhibitors which decrease the concentration of Aβ in culture supernatant. Among these, BMS-708163 and Nilotinib were found to improve the expression levels of RAB3A and SV2B proteins and to recover the electrophysiological function in our AD models. These results suggest that the AD models we developed are promising materials for the discovery of AD drugs that target the expression of pre-synaptic proteins and synaptic function.
Collapse
Affiliation(s)
- Hisae Nishioka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Norie Tooi
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Takehisa Isobe
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Kazuhiro Aiba
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
12
|
Han XF, Zhang Y, Xiong LL, Xu Y, Zhang P, Xia QJ, Wang TH, Ba YC. Lentiviral-Mediated Netrin-1 Overexpression Improves Motor and Sensory Functions in SCT Rats Associated with SYP and GAP-43 Expressions. Mol Neurobiol 2016; 54:1684-1697. [PMID: 26873853 DOI: 10.1007/s12035-016-9723-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 01/13/2016] [Indexed: 02/05/2023]
Abstract
Spinal cord injury (SCI), as a major cause of disability, usually causes serious loss of motor and sensory functions. As a bifunctional axonal guidance cue, netrin-1 can attract axons via the deleted in colorectal cancer (DCC) receptors and repelling others via Unc5 receptors, but its exact role in the recovery of motor and sensory function has not well been studied, and the mechanisms remains elusive. The aim of this experiment is to determine whether lentiviral (LV)-mediated overexpression of netrin-1 or RNA interference (RNAi) can regulate the functional recovery in rats subjected to spinal cord transection (SCT). Firstly, two lentiviral vectors including Lv-exNtn-1 (netrin-1 open reading frame (ORF)) and Lv-shNtn-1 (netrin-1 sh) were constructed and injected into spinal cords rostral and caudal to the transected lesion site. Overexpressing netrin-1 enhanced significantly locomotor function, and reduced thermal and mechanical stimuli in vivo, compared with the control, while silencing netrin-1 did not significantly change the situation. Western blot and immunostaining analysis confirmed that netrin-1 ORF treatment not only effectively increased the expression level of netrin-1, also up-regulated the level of synaptophysin (SYP) in spinal cord rostral to the lesion, but also enhanced growth-associated protein-43 (GAP-43) expression in spinal cord caudal to the lesion site. Comparatively, knockdown of netrin-1 did not give rise to positive findings in our experimental condition. These findings therefore pointed that Lv-mediated netrin-1 overexpression could promote motor and sensory functional recoveries following SCT, and the underlying mechanisms were associated with SYP and GAP-43 expressions. The present study therefore provided a novel strategy for the treatment of SCI and explained the possible mechanisms for the functional improvement.
Collapse
Affiliation(s)
- Xue Fei Han
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China
| | - Yuan Zhang
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China
| | - Liu Lin Xiong
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yang Xu
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Piao Zhang
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China
| | - Qing Jie Xia
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Ting Hua Wang
- Department of Anesthesia, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Ying Chun Ba
- Institute of Neuroscience and Department of Anatomy, Kunming Medical University, Kunming, 650000, China.
| |
Collapse
|