1
|
Wang Q, Xu X, Chen S, Lu R, Li L, Lo CH, Liu Z, Ning K, Li T, Kowal TJ, Wang B, Hartnett ME, Wang S, Qi LS, Sun Y. dCasMINI-mediated therapy rescues photoreceptors degeneration in a mouse model of retinitis pigmentosa. SCIENCE ADVANCES 2024; 10:eadn7540. [PMID: 39693439 DOI: 10.1126/sciadv.adn7540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024]
Abstract
Retinitis pigmentosa (RP) is characterized by degeneration of rod and cone photoreceptors that progresses to irreversible blindness. Now, there are no mutation-agnostic approaches to treat RP. Here, we utilized a single adeno-associated virus (AAV)-based CRISPR activation system to activate phosphodiesterase 6B (Pde6b) to mitigate the severe degeneration in Pde6anmf363 mice. We demonstrate that transcriptional activation of Pde6b can rescue the loss of Pde6a, with preservation of retinal structure, restoration of electroretinography responses, and improvement of visual function as assessed by optokinetic response and looming-induced escape behaviors. These findings demonstrate the therapeutic potential of a dCasMINI-mediated activation strategy that provides a mutation-independent treatment for retinal degeneration. This study offers a promising therapeutic approach for RP and potentially other forms of genetic diseases.
Collapse
Affiliation(s)
- Qing Wang
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Xiaoshu Xu
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Siyu Chen
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Rui Lu
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, USA
| | - Liang Li
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Chien-Hui Lo
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Zhiquan Liu
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Ke Ning
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Tingting Li
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Tia J Kowal
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Biao Wang
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Mary E Hartnett
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Sui Wang
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA 94158, USA
| | - Yang Sun
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
- Maternal Child Health Research Institute at Stanford, Stanford University School of Medicine, Palo Alto, CA, USA
- BioX, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
2
|
Joshi N, Vaidya B, Sharma SS. Transient receptor potential channels as an emerging target for the treatment of Alzheimer's disease: Unravelling the potential of pharmacological interventions. Basic Clin Pharmacol Toxicol 2024; 135:375-400. [PMID: 39209323 DOI: 10.1111/bcpt.14073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/09/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Alzheimer's disease (AD) is a devastating disorder with a multifaceted aetiology characterized by dementia, which later progresses to cognitive impairment. Significant efforts have been made to develop pharmacological interventions that slow down the pathogenesis of AD. However, conventional drugs have failed to satisfactorily treat AD and are more focussed towards symptomatic management. Thus, there is a gap in the literature regarding novel targets and modulators targeting them for the effective treatment of AD. Recent studies have demonstrated that modulation of transient receptor potential (TRP) channels has the potential to halt AD pathogenesis at an early stage and rescue hippocampal neurons from death. Amongst several members, TRP channels like TRPA1, TRPC6, TRPM2 and TRPV2 have shown promising results in the attenuation of neurobehavioural cognitive deficits as well as signalling pathways governing such cognitive decline. Furthermore, as these channels govern the ionic balance in the cell, their beneficial effects have also been known to maintain the homeostasis of Ca2+, which is the major culprit eliciting the vicious cycle of excitotoxicity, mitochondrial dysfunction, ROS generation and neurodegeneration. Despite such tremendous potential of TRP channel modulators, their clinical investigation remains elusive. Therefore, in the present review, we have discussed such agents in the light of TRP channels as molecular targets for the amelioration of AD both at the preclinical and clinical levels.
Collapse
Affiliation(s)
- Nishit Joshi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| | - Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, India
| |
Collapse
|
3
|
Wang J, Chen L, Wang Z, Zhang S, Ding D, Lin G, Zhang H, Boda VK, Kong D, Ortyl TC, Wang X, Lu L, Zhou FM, Bezprozvanny I, Du J, Wu Z, Li W, Liao FF. TRPC3 suppression ameliorates synaptic dysfunctions and memory deficits in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.611061. [PMID: 39345364 PMCID: PMC11430068 DOI: 10.1101/2024.09.16.611061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Transient receptor potential canonical (TRPC) channels are widely expressed in the brain; however, their precise roles in neurodegeneration, such as Alzheimer's disease (AD) remain elusive. Bioinformatic analysis of the published single-cell RNA-seq data collected from AD patient cohorts indicates that the Trpc3 gene is uniquely upregulated in excitatory neurons. TRPC3 expression is also upregulated in post-mortem AD brains, and in both acute and chronic mouse models of AD. Functional screening of TRPC3 antagonists resulted in a lead inhibitor JW-65, which completely rescued Aβ-induced neurotoxicity, impaired synaptic plasticity (e.g., LTP), and learning memory in acute and chronic experimental AD models. In cultured rat hippocampal neurons, we found that treatment with soluble β-amyloid oligomers (AβOs) induces rapid and sustained upregulation of the TRPC3 expression selectively in excitatory neurons. This aberrantly upregulated TRPC3 contributes to AβOs-induced Ca 2+ overload through the calcium entry and store-release mechanisms. The neuroprotective action of JW-65 is primarily mediated via restoring AβOs-impaired Ca 2+ /calmodulin-mediated signaling pathways, including calmodulin kinases CaMKII/IV and calcineurin (CaN). The synaptic protective mechanism via TRPC3 inhibition was further supported by hippocampal RNA-seq data from the symptomatic 5xFAD mice after chronic treatment with JW-65. Overall, these findings not only validate TRPC3 as a novel therapeutic target for treating synaptic dysfunction of AD but most importantly, disclose a distinct role of upregulated TRPC3 in AD pathogenesis in mediating Ca 2+ dyshomeostasis.
Collapse
|
4
|
Rezzani R, Favero G, Gianò M, Pinto D, Labanca M, van Noorden CJ, Rinaldi F. Transient Receptor Potential Channels in the Healthy and Diseased Blood-Brain Barrier. J Histochem Cytochem 2024; 72:199-231. [PMID: 38590114 PMCID: PMC11020746 DOI: 10.1369/00221554241246032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
The large family of transient receptor potential (TRP) channels are integral membrane proteins that function as environmental sensors and act as ion channels after activation by mechanical (touch), physical (heat, pain), and chemical stimuli (pungent compounds such as capsaicin). Most TRP channels are localized in the plasma membrane of cells but some of them are localized in membranes of organelles and function as intracellular Ca2+-ion channels. TRP channels are involved in neurological disorders but their precise role(s) and relevance in these disorders are not clear. Endothelial cells of the blood-brain barrier (BBB) express TRP channels such as TRP vanilloid 1-4 and are involved in thermal detection by regulating BBB permeability. In neurological disorders, TRP channels in the BBB are responsible for edema formation in the brain. Therefore, drug design to modulate locally activity of TRP channels in the BBB is a hot topic. Today, the application of TRP channel antagonists against neurological disorders is still limited.
Collapse
Affiliation(s)
- Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
| | - Marzia Gianò
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, Milan, Italy
| | - Mauro Labanca
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, Milan, Italy
| |
Collapse
|
5
|
Zhou M, Jiao Q, Wu Z, Li W, Liu G, Wang R, Tang Y. Uncovering the Oxidative Stress Mechanisms and Targets in Alzheimer's Disease by Integrating Phenotypic Screening Data and Polypharmacology Networks. J Alzheimers Dis 2024; 99:S139-S156. [PMID: 36744334 DOI: 10.3233/jad-220727] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background The oxidative stress hypothesis is challenging the dominant position of amyloid-β (Aβ) in the field of understanding the mechanisms of Alzheimer's disease (AD), a complicated and untreatable neurodegenerative disease. Objective The goal of the present study was to uncover the oxidative stress mechanisms causing AD, as well as the potential therapeutic targets and neuroprotective drugs against oxidative stress mechanisms. Methods In this study, a systematic workflow combining pharmacological experiments and computational prediction was proposed. 222 drugs and natural products were collected first and then tested on SH-SY5Y cells to obtain phenotypic screening data on neuroprotection. The preliminary screening data were integrated with drug-target interactions (DTIs) and multi-scale biomedical data, which were analyzed with statistical tests and gene set enrichment analysis. A polypharmacology network was further constructed for investigation. Results 340 DTIs were matched in multiple databases, and 222 cell viability ratios were calculated for experimental compounds. We identified significant potential therapeutic targets based on oxidative stress mechanisms for AD, including NR3C1, SHBG, ESR1, PGR, and AVPR1A, which might be closely related to neuroprotective effects and pathogenesis. 50% of the top 14 enriched pathways were found to correlate with AD, such as arachidonic acid metabolism and neuroactive ligand-receptor interaction. Several approved drugs in this research were also found to exert neuroprotective effects against oxidative stress mechanisms, including beclometasone, methylprednisolone, and conivaptan. Conclusion Our results indicated that NR3C1, SHBG, ESR1, PGR, and AVPR1A were promising therapeutic targets and several drugs may be repurposed from the perspective of oxidative stress and AD.
Collapse
Affiliation(s)
- Moran Zhou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Qian Jiao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zengrui Wu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Rui Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
6
|
Baracaldo-Santamaría D, Avendaño-Lopez SS, Ariza-Salamanca DF, Rodriguez-Giraldo M, Calderon-Ospina CA, González-Reyes RE, Nava-Mesa MO. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24109067. [PMID: 37240413 DOI: 10.3390/ijms24109067] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most frequent cause of progressive dementia in senior adults. It is characterized by memory loss and cognitive impairment secondary to cholinergic dysfunction and N-methyl-D-aspartate (NMDA)-mediated neurotoxicity. Intracellular neurofibrillary tangles, extracellular plaques composed of amyloid-β (Aβ), and selective neurodegeneration are the anatomopathological hallmarks of this disease. The dysregulation of calcium may be present in all the stages of AD, and it is associated with other pathophysiological mechanisms, such as mitochondrial failure, oxidative stress, and chronic neuroinflammation. Although the cytosolic calcium alterations in AD are not completely elucidated, some calcium-permeable channels, transporters, pumps, and receptors have been shown to be involved at the neuronal and glial levels. In particular, the relationship between glutamatergic NMDA receptor (NMDAR) activity and amyloidosis has been widely documented. Other pathophysiological mechanisms involved in calcium dyshomeostasis include the activation of L-type voltage-dependent calcium channels, transient receptor potential channels, and ryanodine receptors, among many others. This review aims to update the calcium-dysregulation mechanisms in AD and discuss targets and molecules with therapeutic potential based on their modulation.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Sara Sofia Avendaño-Lopez
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Daniel Felipe Ariza-Salamanca
- Medical and Health Sciences Education Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mateo Rodriguez-Giraldo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos A Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Grupo de Investigación en Ciencias Biomédicas Aplicadas (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
7
|
Saqib U, Munjuluri S, Sarkar S, Biswas S, Mukherjee O, Satsangi H, Baig MS, Obukhov AG, Hajela K. Transient Receptor Potential Canonical 6 (TRPC6) Channel in the Pathogenesis of Diseases: A Jack of Many Trades. Inflammation 2023:10.1007/s10753-023-01808-3. [PMID: 37072606 PMCID: PMC10112830 DOI: 10.1007/s10753-023-01808-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/18/2023] [Accepted: 03/23/2023] [Indexed: 04/20/2023]
Abstract
The mammalian Transient Receptor Potential Canonical (TRPC) subfamily comprises seven transmembrane proteins (TRPC1-7) forming cation channels in the plasma membrane of mammalian cells. TRPC channels mediate Ca2+ and Na+ influx into the cells. Amongst TRPCs, TRPC6 deficiency or increased activity due to gain-of-function mutations has been associated with a multitude of diseases, such as kidney disease, pulmonary disease, and neurological disease. Indeed, the TRPC6 protein is expressed in various organs and is involved in diverse signalling pathways. The last decade saw a surge in the investigative studies concerning the physiological roles of TRPC6 and describing the development of new pharmacological tools modulating TRPC6 activity. The current review summarizes the progress achieved in those investigations.
Collapse
Affiliation(s)
- Uzma Saqib
- School of Life Sciences, Devi Ahilya Vishwavidyalaya, Vigyan Bhawan, Khandwa Road Campus, Indore, 452 001, MP, India
| | - Sreepadaarchana Munjuluri
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sutripta Sarkar
- Post Graduate Department of Food and Nutrition, Barrackpore Rastraguru Surendranath College, 85, Middle Road, Barrackpore, 700120, West Bengal, India
| | - Subir Biswas
- Ramky One Galaxia, Nallagandla, Hyderabad, 500019, Telangana, India
| | - Oyshi Mukherjee
- Post Graduate Department of Food and Nutrition, Barrackpore Rastraguru Surendranath College, 85, Middle Road, Barrackpore, 700120, West Bengal, India
| | | | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Krishnan Hajela
- School of Life Sciences, Devi Ahilya Vishwavidyalaya, Vigyan Bhawan, Khandwa Road Campus, Indore, 452 001, MP, India.
| |
Collapse
|
8
|
Zhang YY, Li XS, Ren KD, Peng J, Luo XJ. Restoration of metal homeostasis: a potential strategy against neurodegenerative diseases. Ageing Res Rev 2023; 87:101931. [PMID: 37031723 DOI: 10.1016/j.arr.2023.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Metal homeostasis is critical to normal neurophysiological activity. Metal ions are involved in the development, metabolism, redox and neurotransmitter transmission of the central nervous system (CNS). Thus, disturbance of homeostasis (such as metal deficiency or excess) can result in serious consequences, including neurooxidative stress, excitotoxicity, neuroinflammation, and nerve cell death. The uptake, transport and metabolism of metal ions are highly regulated by ion channels. There is growing evidence that metal ion disorders and/or the dysfunction of ion channels contribute to the progression of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Therefore, metal homeostasis-related signaling pathways are emerging as promising therapeutic targets for diverse neurological diseases. This review summarizes recent advances in the studies regarding the physiological and pathophysiological functions of metal ions and their channels, as well as their role in neurodegenerative diseases. In addition, currently available metal ion modulators and in vivo quantitative metal ion imaging methods are also discussed. Current work provides certain recommendations based on literatures and in-depth reflections to improve neurodegenerative diseases. Future studies should turn to crosstalk and interactions between different metal ions and their channels. Concomitant pharmacological interventions for two or more metal signaling pathways may offer clinical advantages in treating the neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xi-Sheng Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013,China
| | - Kai-Di Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013,China.
| |
Collapse
|
9
|
Zhu T, Guo J, Wu Y, Lei T, Zhu J, Chen H, Kala S, Wong KF, Cheung CP, Huang X, Zhao X, Yang M, Sun L. The mechanosensitive ion channel Piezo1 modulates the migration and immune response of microglia. iScience 2023; 26:105993. [PMID: 36798430 PMCID: PMC9926228 DOI: 10.1016/j.isci.2023.105993] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/28/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Microglia are the brain's resident immune cells, performing surveillance to promote homeostasis and healthy functioning. While microglial chemical signaling is well-studied, mechanical cues regulating their function are less well-understood. Here, we investigate the role of the mechanosensitive ion channel Piezo1 in microglia migration, pro-inflammatory cytokine production, and stiffness sensing. In Piezo1 knockout transgenic mice, we demonstrated the functional expression of Piezo1 in microglia and identified genes whose expression was consequently affected. Functional assays revealed that Piezo1 deficiency in microglia enhanced migration toward amyloid β-protein, and decreased levels of pro-inflammatory cytokines produced upon stimulation by lipopolysaccharide, both in vitro and in vivo. The phenomenon could be mimicked or reversed chemically using a Piezo1-specific agonist or antagonist. Finally, we also showed that Piezo1 mediated the effect of substrate stiffness-induced migration and cytokine expression. Altogether, we show that Piezo1 is an important molecular mediator for microglia, its activation modulating microglial migration and immune responses.
Collapse
Affiliation(s)
- Ting Zhu
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Jinghui Guo
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Yong Wu
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Ting Lei
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Jiejun Zhu
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Hui Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China,Cell-gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shashwati Kala
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Kin Fung Wong
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Chi Pong Cheung
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Xiaohui Huang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Xinyi Zhao
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Minyi Yang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China
| | - Lei Sun
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR 999077, P. R. China,Corresponding author
| |
Collapse
|
10
|
Liu Y, Yang G, Cui W, Zhang Y, Liang X. Regulatory mechanisms of tetramethylpyrazine on central nervous system diseases: A review. Front Pharmacol 2022; 13:948600. [PMID: 36133805 PMCID: PMC9483103 DOI: 10.3389/fphar.2022.948600] [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: 05/20/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) diseases can lead to motor, sensory, speech, cognitive dysfunction, and sometimes even death. These diseases are recognized to cause a substantial socio-economic impact on a global scale. Tetramethylpyrazine (TMP) is one of the main active ingredients extracted from the Chinese herbal medicine Ligusticum striatum DC. (Chuan Xiong). Many in vivo and in vitro studies have demonstrated that TMP has a certain role in the treatment of CNS diseases through inhibiting calcium ion overload and glutamate excitotoxicity, anti-oxidative/nitrification stress, mitigating inflammatory response, anti-apoptosis, protecting the integrity of the blood-brain barrier (BBB) and facilitating synaptic plasticity. In this review, we summarize the roles and mechanisms of action of TMP on ischemic cerebrovascular disease, spinal cord injury, Parkinson’s disease, Alzheimer’s disease, cognitive impairments, migraine, and depression. Our review will provide new insights into the clinical applications of TMP and the development of novel therapeutics.
Collapse
Affiliation(s)
- Yue Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guang Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenqiang Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunling Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yunling Zhang, ; Xiao Liang,
| | - Xiao Liang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yunling Zhang, ; Xiao Liang,
| |
Collapse
|
11
|
Liu W, Li J, Yang M, Ke X, Dai Y, Lin H, Wang S, Chen L, Tao J. Chemical genetic activation of the cholinergic basal forebrain hippocampal circuit rescues memory loss in Alzheimer's disease. Alzheimers Res Ther 2022; 14:53. [PMID: 35418161 PMCID: PMC9006585 DOI: 10.1186/s13195-022-00994-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/27/2022] [Indexed: 11/18/2022]
Abstract
Background The degeneration of the cholinergic circuit from the basal forebrain to the hippocampus contributes to memory loss in patients suffering from Alzheimer’s disease (AD). However, the internal relationships between the acetylcholine (Ach) cycle and memory decline during the early stages of AD currently remain unknown. Here, we investigate the mechanisms underlying the activation of the cholinergic circuit and its impact on learning and memory using APP/PS1 mice models. Methods Novel object recognition and Morris water maze tests were used to measure learning and memory function. Magnetic resonance spectrum (MRS) imaging was applied to longitudinally track changes in neurochemical metabolism in APP/PS1 mice aged 2, 4, 6, and 8 months. The number of neurons and the deposition of Aβ plaques were measured using Nissl, immunohistochemistry, and Thioflavin S staining. We then employed a chemogenetic strategy to selectively activate the cholinergic circuit from the medial septal nucleus (MS) and the vertical limb of the diagonal band nucleus (VDB) on the basal forebrain to the hippocampus. MRS and immunoblotting techniques were used to measure the neurochemical metabolism levels and cholinergic-related proteins, respectively. Results We found that the levels of choline (Cho) in the basal forebrain were markedly higher compared to other brain regions and that its decrease along with N-acetyl aspartate (NAA) levels in the hippocampus was accompanied by memory deficits in APP/PS1 mice aged 4, 6, and 8 months. In terms of pathology, we observed that the deposition of Aβ plaques gradually aggravated throughout the cerebral cortex and hippocampus in APP/PS1 mice aged 6 and 8 months, while no Aβ deposition was detected in the basal forebrain. In contrast, the activity of choline acetyltransferase (ChAT) enzyme in the basal forebrain was decreased at 6 months of age and the cholinergic neurons were lost in the basal forebrain at 8 months of age. In addition, the activation of the cholinergic circuit from the MS and VDB to the hippocampus using chemical genetics is able to improve learning and reduce memory impairment in APP/PS1 mice. Similarly, the levels of Cho in the basal forebrain; NAA in the hippocampus, as well as the expression of ChAT and vesicular acetylcholine transporter (vAchT) in the basal forebrain; and muscarinic acetylcholine receptor 2 (CHRM2) in the hippocampus all increased. Conclusions These findings demonstrate that the neurochemical Cho and NAA of the cholinergic circuit can be used as biomarkers to enable the early diagnosis of AD. In addition, memory impairment in APP/PS1 mice can be attenuated using chemical genetics-driven Ach cycle activity of the cholinergic circuit. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00994-w.
Collapse
Affiliation(s)
- Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.,The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Jianhong Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Minguang Yang
- The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Xiaohua Ke
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Yaling Dai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Huawei Lin
- Rehabilitation Medical Technology Joint National Local Engineering Research Center, Fuzhou, 350122, Fujian, China
| | - Sinuo Wang
- Rehabilitation Medical Technology Joint National Local Engineering Research Center, Fuzhou, 350122, Fujian, China
| | - Lidian Chen
- The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
12
|
Ren R, Qi J, Lin S, Liu X, Yin P, Wang Z, Tang R, Wang J, Huang Q, Li J, Xie X, Hu Y, Cui S, Zhu Y, Yu X, Wang P, Zhu Y, Wang Y, Huang Y, Hu Y, Wang Y, Li C, Zhou M, Wang G. The China Alzheimer Report 2022. Gen Psychiatr 2022; 35:e100751. [PMID: 35372787 PMCID: PMC8919463 DOI: 10.1136/gpsych-2022-100751] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/23/2022] [Indexed: 01/22/2023] Open
Abstract
China’s population has rapidly aged over the recent decades of social and economic development as neurodegenerative disorders have proliferated, especially Alzheimer’s disease (AD) and related dementias (ADRD). AD’s incidence rate, morbidity, and mortality have steadily increased to make it presently the fifth leading cause of death among urban and rural residents in China and magnify the resulting financial burdens on individuals, families and society. The ‘Healthy China Action’ plan of 2019–2030 promotes the transition from disease treatment to health maintenance for this expanding population with ADRD. This report describes related epidemiological trends, evaluates the economic burden of the disease, outlines current clinical diagnosis and treatment status and delineates existing available public health resources. More specifically, it examines the public health impact of ADRD, including prevalence, mortality, costs, usage of care, and the overall effect on caregivers and society. In addition, this special report presents technical guidance and supports for the prevention and treatment of AD, provides expertise to guide relevant governmental healthcare policy development and suggests an information platform for international exchange and cooperation.
Collapse
Affiliation(s)
- Rujing Ren
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinlei Qi
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shaohui Lin
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinya Liu
- School of Public Health, Fudan University, Shanghai, China
- NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Peng Yin
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhihui Wang
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ran Tang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jintao Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianping Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Xie
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongbo Hu
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shishuang Cui
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Zhu
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Yu
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengfei Wang
- School of Public Health, Fudan University, Shanghai, China
- NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Yikang Zhu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiran Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyan Huang
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China
| | - Yisong Hu
- National Survey Research Center, Renmin University of China, Beijing, China
| | - Ying Wang
- School of Public Health, Fudan University, Shanghai, China
- NHC Key Laboratory of Health Technology Assessment, Fudan University, Shanghai, China
| | - Chunbo Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maigeng Zhou
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Gang Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
He C, Li Q, Cui Y, Gao P, Shu W, Zhou Q, Wang L, Li L, Lu Z, Zhao Y, Ma H, Chen X, Jia H, Zheng H, Yang G, Liu D, Tepel M, Zhu Z. Recurrent moderate hypoglycemia accelerates the progression of cognitive deficits through impairment of TRPC6/GLUT3 pathway in diabetic APP/PS1 mice. JCI Insight 2022; 7:154595. [PMID: 35077394 PMCID: PMC8983129 DOI: 10.1172/jci.insight.154595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/20/2022] [Indexed: 11/17/2022] Open
Abstract
Currently, the most effective strategy for dealing with Alzheimer’s disease (AD) is delaying the onset of dementia. Severe hypoglycemia is strongly associated with dementia; however, the effects of recurrent moderate hypoglycemia (RH) on the progression of cognitive deficits in patients with diabetes with genetic susceptibility to AD remain unclear. Here, we report that insulin-controlled hyperglycemia slightly aggravated AD-type pathologies and cognitive impairment; however, RH significantly increased neuronal hyperactivity and accelerated the progression of cognitive deficits in streptozotocin-induced (STZ-induced) diabetic APP/PS1 mice. Glucose transporter 3–mediated (GLUT3-mediated) neuronal glucose uptake was not significantly altered under hyperglycemia but was markedly reduced by RH, which induced excessive mitochondrial fission in the hippocampus. Overexpression of GLUT3, specifically in the dentate gyrus (DG) area of the hippocampus, enhanced mitochondrial function and improved cognitive deficits. Activation of the transient receptor potential channel 6 (TRPC6) increased GLUT3-mediated glucose uptake in the brain and alleviated RH-induced cognitive deficits, and inactivation of the Ca2+/AMPK pathway was responsible for TRPC6-induced GLUT3 inhibition. Taken together, RH impairs brain GLUT3-mediated glucose uptake and further provokes neuronal mitochondrial dysfunction by inhibiting TRPC6 expression, which then accelerates progression of cognitive deficits in diabetic APP/PS1 mice. Avoiding RH is essential for glycemic control in patients with diabetes, and TRPC6/GLUT3 represents potent targets for delaying the onset of dementia in patients with diabetes.
Collapse
Affiliation(s)
- Chengkang He
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Qiang Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Yuanting Cui
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Wentao Shu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Qing Zhou
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Lijuan Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Yu Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Huan Ma
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Xiaowei Chen
- Brain Research Center, Army Medical University, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Hongbo Jia
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Gangyi Yang
- Endocrine Department, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Martin Tepel
- Odense University Hospital, Department of Nephrology, University of Southern Denmark, Institute for Molecular Medicine, Cardiovascular and Renal Research, Institute of Clinical Research, Odense, Denmark
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing Institute for Brain and Intelligence, Chongqing, China
| |
Collapse
|
14
|
Tedeschi V, Sisalli MJ, Pannaccione A, Piccialli I, Molinaro P, Annunziato L, Secondo A. Na +/Ca 2+ exchanger isoform 1 (NCX1) and canonical transient receptor potential channel 6 (TRPC6) are recruited by STIM1 to mediate Store-Operated Calcium Entry in primary cortical neurons. Cell Calcium 2022; 101:102525. [PMID: 34995919 DOI: 10.1016/j.ceca.2021.102525] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/05/2021] [Accepted: 12/26/2021] [Indexed: 02/01/2023]
Abstract
Excessive calcium (Ca2+) release from the endoplasmic reticulum (ER) represents an important hallmark of several neurodegenerative diseases. ER is recharged from Ca2+ through the so-called Store-Operated Calcium Entry (SOCE) thus providing Ca2+ signals to regulate critical cell functions. Single transmembrane-spanning domain protein stromal interacting molecule 1 (STIM1), mainly residing in the ER, and plasmalemmal channel Orai1 represent the SOCE key components at neuronal level. However, many other proteins participate to ER Ca2+ refilling including the Na+/Ca2+ exchanger isoform 1 (NCX1), whose regulation by ER remains unknown. In this study, we tested the possibility that neuronal NCX1 may take part to SOCE through the interaction with STIM1. In rat primary cortical neurons and in nerve growth factor (NGF)-differentiated PC12 cells NCX1 knocking down by siRNA strategy significantly prevented SOCE as well as SOCE pharmacological inhibition by SKF-96365 and 2-APB. A significant reduction of SOCE was recorded also in synaptosomes from ncx1-/- mice brain compared with ncx1+/+ mice. Double labeling confocal experiments showed a large co-localization between NCX1 and STIM1 in rat primary cortical neurons. Accordingly, NCX1 and STIM1 co-immunoprecipitated and functionally interacted each other during ischemic preconditioning, a phenomenon inducing ischemic tolerance. However, STIM1 knocking down reduced NCX1 activity recorded by either patch-clamp electrophysiology or Fura-2 single-cell microfluorimetry. Furthermore, canonical transient receptor potential channel 6 (TRPC6) was identified as the mechanism mediating local increase of sodium (Na+) useful to drive NCX1 reverse mode and, therefore, NCX1-mediated Ca2+ refilling. In fact, TRPC6 not only interacted with STIM1, as shown by the co-localization and co-immunoprecipitation with the ER Ca2+ sensor, but it also mediated 1,3-Benzenedicarboxylic acid, 4,4'-[1,4,10-trioxa-7,13-diazacyclopentadecane-7,13-diylbis(5-methoxy-6,12-benzofurandiyl)]bis-, tetrakis[(acetyloxy)methyl] ester (SBFI)-monitored Na+ increase elicited by thapsigargin in primary cortical neurons. Accordingly, efficient TRPC6 knockdown prevented thapsigargin-induced intracellular Na+ elevation and SOCE. Collectively, we identify NCX1 as a new partner of STIM1 in mediating SOCE, whose activation in the reverse mode may be facilitated by the local increase of Na+ concentration due to the interaction between STIM1 and TRPC6 in primary cortical neurons.
Collapse
Affiliation(s)
- Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | - Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | - Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | - Pasquale Molinaro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | | | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via S. Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
15
|
Mutations in trpγ, the homologue of TRPC6 autism candidate gene, causes autism-like behavioral deficits in Drosophila. Mol Psychiatry 2022; 27:3328-3342. [PMID: 35501408 PMCID: PMC9708601 DOI: 10.1038/s41380-022-01555-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 03/15/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022]
Abstract
Autism Spectrum Disorder (ASD) is characterized by impaired social communication, restricted interests, and repetitive and stereotyped behaviors. The TRPC6 (transient receptor potential channel 6) represents an ASD candidate gene under an oligogenic/multifactorial model based on the initial description and cellular characterization of an individual with ASD bearing a de novo heterozygous mutation disrupting TRPC6, together with the enrichment of disruptive TRPC6 variants in ASD cases as compared to controls. Here, we perform a clinical re-evaluation of the initial non-verbal patient, and also present eight newly reported individuals ascertained for ASD and bearing predicted loss-of-function mutations in TRPC6. In order to understand the consequences of mutations in TRPC6 on nervous system function, we used the fruit fly, Drosophila melanogaster, to show that null mutations in transient receptor gamma (trpγ; the fly gene most similar to TRPC6), cause a number of behavioral defects that mirror features seen in ASD patients, including deficits in social interactions (based on courtship behavior), impaired sleep homeostasis (without affecting the circadian control of sleep), hyperactivity in both young and old flies, and defects in learning and memory. Some defects, most notably in sleep, differed in severity between males and females and became normal with age. Interestingly, hyperforin, a TRPC6 agonist and the primary active component of the St. John's wort antidepressant, attenuated many of the deficits expressed by trpγ mutant flies. In summary, our results provide further evidence that the TRPC6 gene is a risk factor for ASD. In addition, they show that the behavioral defects caused by mutations in TRPC6 can be modeled in Drosophila, thereby establishing a paradigm to examine the impact of mutations in other candidate genes.
Collapse
|
16
|
Roy P, Martinelli I, Moruzzi M, Maggi F, Amantini C, Micioni Di Bonaventura MV, Cifani C, Amenta F, Tayebati SK, Tomassoni D. Ion channels alterations in the forebrain of high-fat diet fed rats. Eur J Histochem 2021; 65:3305. [PMID: 34814650 PMCID: PMC8636841 DOI: 10.4081/ejh.2021.3305] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/27/2021] [Indexed: 12/22/2022] Open
Abstract
Evidence suggests that transient receptor potential (TRP) ion channels dysfunction significantly contributes to the physiopathology of metabolic and neurological disorders. Dysregulation in functions and expression in genes encoding the TRP channels cause several inherited diseases in humans (the so-called 'TRP channelopathies'), which affect the cardiovascular, renal, skeletal, and nervous systems. This study aimed to evaluate the expression of ion channels in the forebrain of rats with diet-induced obesity (DIO). DIO rats were studied after 17 weeks under a hypercaloric diet (high-fat diet, HFD) and were compared to the control rats with a standard diet (CHOW). To determine the systemic effects of HFD exposure, we examined food intake, fat mass content, fasting glycemia, insulin levels, cholesterol, and triglycerides. qRT-PCR, Western blot, and immunochemistry analysis were performed in the frontal cortex (FC) and hippocampus (HIP). After 17 weeks of HFD, DIO rats increased their body weight significantly compared to the CHOW rats. In DIO rats, TRPC1 and TRPC6 were upregulated in the HIP, while they were downregulated in the FC. In the case of TRPM2 expression, instead was increased both in the HIP and in the FC. These could be related to the increase of proteins and nucleic acid oxidation. TRPV1 and TRPV2 gene expression showed no differences both in the FC and HIP. In general, qRT-PCR analyses were confirmed by Western blot analysis. Immunohistochemical procedures highlighted the expression of the channels in the cell body of neurons and axons, particularly for the TRPC1 and TRPC6. The alterations of TRP channel expression could be related to the activation of glial cells or the neurodegenerative process presented in the brain of the DIO rat highlighted with post synaptic protein (PSD 95) alterations. The availability of suitable animal models may be useful for studying possible pharmacological treatments to counter obesity-induced brain injury. The identified changes in DIO rats may represent the first insight to characterize the neuronal alterations occurring in obesity. Further investigations are necessary to characterize the role of TRP channels in the regulation of synaptic plasticity and obesity-related cognitive decline.
Collapse
Affiliation(s)
- Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | - Federica Maggi
- Department of Molecular Medicine, La Sapienza University of Rome.
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | | | | | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino.
| |
Collapse
|
17
|
Xiang J, Yang F, Zhu W, Cai M, Li XT, Zhang JS, Yu ZH, Zhang W, Cai DF. Bilobalide inhibits inflammation and promotes the expression of Aβ degrading enzymes in astrocytes to rescue neuronal deficiency in AD models. Transl Psychiatry 2021; 11:542. [PMID: 34671017 PMCID: PMC8528910 DOI: 10.1038/s41398-021-01594-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
The pathogenesis of Alzheimer's disease (AD) involves multiple cell types including endothelial cells, glia, and neurons. It suggests that therapy against single target in single cell type may not be sufficient to treat AD and therapies with protective effects in multiple cell types may be more effective. Here, we comprehensively investigated the effects of bilobalide on neuroinflammation and Aβ degrading enzymes in AD cell model and mouse model. We find that bilobalide inhibits Aβ-induced and STAT3-dependent expression of TNF-α, IL-1β, and IL-6 in primary astrocyte culture. Bilobalide also induces robust expression of Aβ degrading enzymes like NEP, IDE, and MMP2 to facilitate astrocyte-mediated Aβ clearance. Moreover, bilobalide treatment of astrocyte rescues neuronal deficiency in co-cultured APP/PS1 neurons. Most importantly, bilobalide reduces amyloid and inflammation in AD mouse brain. Taken together, the protective effects of bilobalide in in vitro cultures were fully recapitulated in in vivo AD mouse model. Our study supports that bilobalide has therapeutic potential for AD treatment.
Collapse
Affiliation(s)
- Jun Xiang
- grid.8547.e0000 0001 0125 2443Department of Integrative Medicine, Zhongshan Hospital, Fudan University, 200032 Shanghai, China ,grid.8547.e0000 0001 0125 2443Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, 200032 Shanghai, China
| | - Feng Yang
- grid.8547.e0000 0001 0125 2443Department of Integrative Medicine, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Wen Zhu
- grid.8547.e0000 0001 0125 2443Department of Integrative Medicine, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Min Cai
- grid.8547.e0000 0001 0125 2443Department of Integrative Medicine, Zhongshan Hospital, Fudan University, 200032 Shanghai, China ,grid.8547.e0000 0001 0125 2443Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, 200032 Shanghai, China
| | - Xiang-Ting Li
- grid.8547.e0000 0001 0125 2443Department of Integrative Medicine, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Jing-Si Zhang
- grid.412585.f0000 0004 0604 8558Department of Neurology, Shuguang hospital affiliated to Shanghai University of TCM, 201203 Shanghai, China
| | - Zhong-Hai Yu
- grid.412528.80000 0004 1798 5117Department of Traditional Chinese Medicine, the Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, 200233 Shanghai, China
| | - Wen Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China. .,Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, 200032, Shanghai, China.
| | - Ding-Fang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, 200032, Shanghai, China. .,Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
18
|
miR-193a Directly Targets PSEN1 and Inhibits Gastric Cancer Cell Growth, the Activation of PI3K/Akt Signaling Pathway, and the Epithelial-to-Mesenchymal Transition. JOURNAL OF ONCOLOGY 2021; 2021:2804478. [PMID: 34335753 PMCID: PMC8298175 DOI: 10.1155/2021/2804478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/05/2021] [Indexed: 11/25/2022]
Abstract
Background Gastric cancer, a kind of gastrointestinal malignancy, is the second type of leading death cancer. miR-193a is a key tumor suppressor in several diseases. PSEN1 is mainly related to Alzheimer's disease and may be involved in the cleavage of the Notch receptor. Material and Methods. RT-PCR and western blot were applied to evaluate miR-193a and the expression level of PSEN1. Luciferase reporter assay was applied to verify whether PSEN1 was a target of miR-193a. The Kaplan–Meier method was employed to calculate the 5-year overall survival of gastric cancer patients. Results miR-193a was downregulated in gastric cancer tissues and cell lines, and downregulation of miR-193a predicted poor 5-year overall survival of gastric cancer. miR-193a inhibited the proliferation and the activation of the PI3K/AKT signaling pathway in gastric cancer cells. miR-193a inhibited gastric cancer tumor growth in vivo. miR-193a impaired cell invasion and epithelial-to-mesenchymal transition (EMT) in HGC-27 cells. In addition, PSEN1 was a direct target of miR-193a and PSEN1 reversed partial functions of miR-193a in cell proliferation and invasion. Conclusion miR-193a prominently decreased the proliferation, invasion, and activation of the PI3K/Akt signaling pathway and the abilities of epithelial-to-mesenchymal transition in gastric cancer cells. The newly identified miR-193a/PSEN1 axis provides novel insight into the pathogenesis of gastric cancer.
Collapse
|
19
|
Sun Y, Liu Z, Pi Z, Song F, Wu J, Liu S. Poria cocos could ameliorate cognitive dysfunction in APP/PS1 mice by restoring imbalance of Aβ production and clearance and gut microbiota dysbiosis. Phytother Res 2021; 35:2678-2690. [PMID: 33432644 DOI: 10.1002/ptr.7014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder. Amyloid beta-protein (Aβ) plaques, which are the hallmark of AD, are formed from the imbalance of Aβ production and clearance accompanied by neuroinflammation, gut dysbiosis, and metabolite dysfunction. All of these processes give rise to neurochemical deficiencies and synaptic dysfunction, which ultimately contribute to recognition dysfunction. Poria cocos (PC), which contains multiple active ingredients, plays a significant role in the treatment of multiple-pathogenesis senile diseases such as AD. Nevertheless, there are only very few investigations on the intricate action mechanism of PC for the treatment of AD. In this study, we evaluate the multi-target cure effect of PC on APP/PS1 mice by behavioral, immunohistochemical (IHC), targeted metabolomics, and 16S rRNA sequencing experiments. Mice treated with PC showed significant improvements in cognitive function as evaluated by the behavioral experiment. IHC revealed that PC treatment relieved Aβ deposition by reducing the formation of Aβ and increasing its clearance. Moreover, PC treatment improved gut dysbiosis, which reversed the metabolite dysfunction of bile acid. These findings reveal that PC is a promising therapeutic agent, which might ameliorate the cognitive function of AD by restoring the imbalance of Aβ production and clearance and gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Yufei Sun
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun, Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Zhiqiang Liu
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun, Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Zifeng Pi
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun, Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Fengrui Song
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun, Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Jianlin Wu
- State Key Laboratory for Quality Research of Chinese Medicines, Macau University of Science and Technology, Taipa, China
| | - Shu Liu
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun, Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
20
|
Canonical transient receptor potential channels and their modulators: biology, pharmacology and therapeutic potentials. Arch Pharm Res 2021; 44:354-377. [PMID: 33763843 PMCID: PMC7989688 DOI: 10.1007/s12272-021-01319-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
Canonical transient receptor potential channels (TRPCs) are nonselective, high calcium permeability cationic channels. The TRPCs family includes TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, and TRPC7. These channels are widely expressed in the cardiovascular and nervous systems and exist in many other human tissues and cell types, playing several crucial roles in the human physiological and pathological processes. Hence, the emergence of TRPCs modulators can help investigate these channels’ applications in health and disease. It is worth noting that the TRPCs subfamilies have structural and functional similarities, which presents a significant difficulty in screening and discovering of TRPCs modulators. In the past few years, only a limited number of selective modulators of TRPCs were detected; thus, additional research on more potent and more selective TRPCs modulators is needed. The present review focuses on the striking desired therapeutic effects of TRPCs modulators, which provides intel on the structural modification of TRPCs modulators and further pharmacological research. Importantly, TRPCs modulators can significantly facilitate future studies of TRPCs and TRPCs related diseases.
Collapse
|
21
|
Lee K, Jo YY, Chung G, Jung JH, Kim YH, Park CK. Functional Importance of Transient Receptor Potential (TRP) Channels in Neurological Disorders. Front Cell Dev Biol 2021; 9:611773. [PMID: 33748103 PMCID: PMC7969799 DOI: 10.3389/fcell.2021.611773] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential (TRP) channels are transmembrane protein complexes that play important roles in the physiology and pathophysiology of both the central nervous system (CNS) and the peripheral nerve system (PNS). TRP channels function as non-selective cation channels that are activated by several chemical, mechanical, and thermal stimuli as well as by pH, osmolarity, and several endogenous or exogenous ligands, second messengers, and signaling molecules. On the pathophysiological side, these channels have been shown to play essential roles in the reproductive system, kidney, pancreas, lung, bone, intestine, as well as in neuropathic pain in both the CNS and PNS. In this context, TRP channels have been implicated in several neurological disorders, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and epilepsy. Herein, we focus on the latest involvement of TRP channels, with a special emphasis on the recently identified functional roles of TRP channels in neurological disorders related to the disruption in calcium ion homeostasis.
Collapse
Affiliation(s)
- Kihwan Lee
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Youn Yi Jo
- Department of Anesthesiology and Pain Medicine, Gil Medical Center, Gachon University, Incheon, South Korea
| | - Gehoon Chung
- Department of Oral Physiology and Program in Neurobiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jung Hoon Jung
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
22
|
Kinoshita PF, Orellana AMM, Nakao VW, de Souza Port's NM, Quintas LEM, Kawamoto EM, Scavone C. The Janus face of ouabain in Na + /K + -ATPase and calcium signalling in neurons. Br J Pharmacol 2021; 179:1512-1524. [PMID: 33644859 DOI: 10.1111/bph.15419] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 12/20/2022] Open
Abstract
Na+ /K+ -ATPase, a transmembrane protein essential for maintaining the electrochemical gradient across the plasma membrane, acts as a receptor for cardiotonic steroids such as ouabain. Cardiotonic steroids binding to Na+ /K+ -ATPase triggers signalling pathways or inhibits Na+ /K+ -ATPas activity in a concentration-dependent manner, resulting in a modulation of Ca2+ levels, which are essential for homeostasis in neurons. However, most of the pharmacological strategies for avoiding neuronal death do not target Na+ /K+ -ATPase activity due to its complexity and the poor understanding of the mechanisms involved in Na+ /K+ -ATPase modulation. The present review aims to discuss two points regarding the interplay between Na+ /K+ -ATPase and Ca2+ signalling in the brain. One, Na+ /K+ -ATPase impairment causing illness and neuronal death due to Ca2+ signalling and two, benefits to the brain by modulating Na+ /K+ -ATPase activity. These interactions play an essential role in neuronal cell fate determination and are relevant to find new targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Maria Marques Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vinicius Watanabe Nakao
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natacha Medeiros de Souza Port's
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luis Eduardo Menezes Quintas
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Health Sciences Centre Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
23
|
Hwang SM, Lee JY, Park CK, Kim YH. The Role of TRP Channels and PMCA in Brain Disorders: Intracellular Calcium and pH Homeostasis. Front Cell Dev Biol 2021; 9:584388. [PMID: 33585474 PMCID: PMC7876282 DOI: 10.3389/fcell.2021.584388] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Brain disorders include neurodegenerative diseases (NDs) with different conditions that primarily affect the neurons and glia in the brain. However, the risk factors and pathophysiological mechanisms of NDs have not been fully elucidated. Homeostasis of intracellular Ca2+ concentration and intracellular pH (pHi) is crucial for cell function. The regulatory processes of these ionic mechanisms may be absent or excessive in pathological conditions, leading to a loss of cell death in distinct regions of ND patients. Herein, we review the potential involvement of transient receptor potential (TRP) channels in NDs, where disrupted Ca2+ homeostasis leads to cell death. The capability of TRP channels to restore or excite the cell through Ca2+ regulation depending on the level of plasma membrane Ca2+ ATPase (PMCA) activity is discussed in detail. As PMCA simultaneously affects intracellular Ca2+ regulation as well as pHi, TRP channels and PMCA thus play vital roles in modulating ionic homeostasis in various cell types or specific regions of the brain where the TRP channels and PMCA are expressed. For this reason, the dysfunction of TRP channels and/or PMCA under pathological conditions disrupts neuronal homeostasis due to abnormal Ca2+ and pH levels in the brain, resulting in various NDs. This review addresses the function of TRP channels and PMCA in controlling intracellular Ca2+ and pH, which may provide novel targets for treating NDs.
Collapse
Affiliation(s)
- Sung-Min Hwang
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Ji Yeon Lee
- Gil Medical Center, Department of Anesthesiology and Pain Medicine, Gachon University, Incheon, South Korea
| | - Chul-Kyu Park
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Yong Ho Kim
- Gachon Pain Center, Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
24
|
Jeon J, Bu F, Sun G, Tian JB, Ting SM, Li J, Aronowski J, Birnbaumer L, Freichel M, Zhu MX. Contribution of TRPC Channels in Neuronal Excitotoxicity Associated With Neurodegenerative Disease and Ischemic Stroke. Front Cell Dev Biol 2021; 8:618663. [PMID: 33490083 PMCID: PMC7820370 DOI: 10.3389/fcell.2020.618663] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
The seven canonical members of transient receptor potential (TRPC) proteins form cation channels that evoke membrane depolarization and intracellular calcium concentration ([Ca2+] i ) rise, which are not only important for regulating cell function but their deregulation can also lead to cell damage. Recent studies have implicated complex roles of TRPC channels in neurodegenerative diseases including ischemic stroke. Brain ischemia reduces oxygen and glucose supply to neurons, i.e., Oxygen and Glucose Deprivation (OGD), resulting in [Ca2+] i elevation, ion dyshomeostasis, and excitotoxicity, which are also common in many forms of neurodegenerative diseases. Although ionotropic glutamate receptors, e.g., N-methyl-D-aspartate receptors, are well established to play roles in excitotoxicity, the contribution of metabotropic glutamate receptors and their downstream effectors, i.e., TRPC channels, should not be neglected. Here, we summarize the current findings about contributions of TRPC channels in neurodegenerative diseases, with a focus on OGD-induced neuronal death and rodent models of cerebral ischemia/reperfusion. TRPC channels play both detrimental and protective roles to neurodegeneration depending on the TRPC subtype and specific pathological conditions involved. When illustrated the mechanisms by which TRPC channels are involved in neuronal survival or death seem differ greatly, implicating diverse and complex regulation. We provide our own data showing that TRPC1/C4/C5, especially TRPC4, may be generally detrimental in OGD and cerebral ischemia/reperfusion. We propose that although TRPC channels significantly contribute to ischemic neuronal death, detailed mechanisms and specific roles of TRPC subtypes in brain injury at different stages of ischemia/reperfusion and in different brain regions need to be carefully and systematically investigated.
Collapse
Affiliation(s)
- Jaepyo Jeon
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Fan Bu
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Guanghua Sun
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jin-Bin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shun-Ming Ting
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jun Li
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Lutz Birnbaumer
- Institute for Biomedical Research (BIOMED UCA-CONICET), Buenos Aires, Argentina.,School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, Argentina.,Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Marc Freichel
- Department of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
25
|
Chami M, Checler F. Alterations of the Endoplasmic Reticulum (ER) Calcium Signaling Molecular Components in Alzheimer's Disease. Cells 2020; 9:cells9122577. [PMID: 33271984 PMCID: PMC7760721 DOI: 10.3390/cells9122577] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Sustained imbalance in intracellular calcium (Ca2+) entry and clearance alters cellular integrity, ultimately leading to cellular homeostasis disequilibrium and cell death. Alzheimer’s disease (AD) is the most common cause of dementia. Beside the major pathological features associated with AD-linked toxic amyloid beta (Aβ) and hyperphosphorylated tau (p-tau), several studies suggested the contribution of altered Ca2+ handling in AD development. These studies documented physical or functional interactions of Aβ with several Ca2+ handling proteins located either at the plasma membrane or in intracellular organelles including the endoplasmic reticulum (ER), considered the major intracellular Ca2+ pool. In this review, we describe the cellular components of ER Ca2+ dysregulations likely responsible for AD. These include alterations of the inositol 1,4,5-trisphosphate receptors’ (IP3Rs) and ryanodine receptors’ (RyRs) expression and function, dysfunction of the sarco-endoplasmic reticulum Ca2+ ATPase (SERCA) activity and upregulation of its truncated isoform (S1T), as well as presenilin (PS1, PS2)-mediated ER Ca2+ leak/ER Ca2+ release potentiation. Finally, we highlight the functional consequences of alterations of these ER Ca2+ components in AD pathology and unravel the potential benefit of targeting ER Ca2+ homeostasis as a tool to alleviate AD pathogenesis.
Collapse
Affiliation(s)
- Mounia Chami
- Correspondence: ; Tel.: +33-4939-53457; Fax: +33-4939-53408
| | | |
Collapse
|
26
|
Prikhodko V, Chernyuk D, Sysoev Y, Zernov N, Okovityi S, Popugaeva E. Potential Drug Candidates to Treat TRPC6 Channel Deficiencies in the Pathophysiology of Alzheimer's Disease and Brain Ischemia. Cells 2020; 9:cells9112351. [PMID: 33114455 PMCID: PMC7692306 DOI: 10.3390/cells9112351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease and cerebral ischemia are among the many causative neurodegenerative diseases that lead to disabilities in the middle-aged and elderly population. There are no effective disease-preventing therapies for these pathologies. Recent in vitro and in vivo studies have revealed the TRPC6 channel to be a promising molecular target for the development of neuroprotective agents. TRPC6 channel is a non-selective cation plasma membrane channel that is permeable to Ca2+. Its Ca2+-dependent pharmacological effect is associated with the stabilization and protection of excitatory synapses. Downregulation as well as upregulation of TRPC6 channel functions have been observed in Alzheimer’s disease and brain ischemia models. Thus, in order to protect neurons from Alzheimer’s disease and cerebral ischemia, proper TRPC6 channels modulators have to be used. TRPC6 channels modulators are an emerging research field. New chemical structures modulating the activity of TRPC6 channels are being currently discovered. The recent publication of the cryo-EM structure of TRPC6 channels should speed up the discovery process even more. This review summarizes the currently available information about potential drug candidates that may be used as basic structures to develop selective, highly potent TRPC6 channel modulators to treat neurodegenerative disorders, such as Alzheimer’s disease and cerebral ischemia.
Collapse
Affiliation(s)
- Veronika Prikhodko
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Daria Chernyuk
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Yurii Sysoev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 St. Petersburg, Russia
| | - Nikita Zernov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
| | - Sergey Okovityi
- Department of Pharmacology and Clinical Pharmacology, Saint Petersburg State Chemical Pharmaceutical University, 197022 St. Petersburg, Russia;
- N.P. Bechtereva Institute of the Human Brain of the Russian Academy of Sciences, 197376 St. Petersburg, Russia
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia; (V.P.); (D.C.); (Y.S.); (N.Z.)
- Correspondence:
| |
Collapse
|
27
|
He C, Gao P, Cui Y, Li Q, Li Y, Lu Z, Ma H, Zhao Y, Li L, Sun F, Chen X, Jia H, Liu D, Yang G, Zheng H, Zhu Z. Low-glucose-sensitive TRPC6 dysfunction drives hypoglycemia-induced cognitive impairment in diabetes. Clin Transl Med 2020; 10:e205. [PMID: 33135341 PMCID: PMC7568851 DOI: 10.1002/ctm2.205] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background Recurrent moderate hypoglycemia (RH), a major adverse effect of hypoglycemic therapy in diabetic patients, is one of the main risk factors for cognitive impairment and dementia. Transient receptor potential canonical channel 6 (TRPC6) is a potential therapeutic target for Alzheimer's disease (AD) and its expression is highly regulated by glucose concentration. Objective To investigate whether RH regulates the expression of TRPC6 in brain and whether TRPC6 dysfunction can drive hypoglycemia‐associated cognitive impairment in diabetes, and reveal the underlying mechanism. Methods Histological staining, in vivo two‐photon Ca2+ imaging, and behavioral tests were used to measure neuronal death, brain network activity, and cognitive function in mice, respectively. High‐resolution respirometry and transmission electron microscope were used to assess mitochondrial structure and function. Intracellular calcium measurement and molecular biology techniques were conducted to uncover the underlying mechanism. Results Here, we report that the expression of TRPC6 in hippocampus was specifically repressed by RH in streptozocin‐induced type 1 diabetic mice, but not in nondiabetic mice. TRPC6 knockout directly leads to neuron loss, neuronal activity, and cognitive function impairment under diabetic condition, the degree of which is similar to that of RH. Activation of TRPC6 with hyperforin substantially improved RH‐induced cognitive impairment. Mechanistically, TRPC6 inhibited mitochondrial fission in the hippocampus of diabetic mice undergoing RH episodes by activating adenosine 5‘‐monophosphate‐activated protein kinase, and TRPC6‐mediated cytosolic calcium influx was required for this process. Clinically, dysfunction of TRPC6 was closely associated with cognitive impairment in type 2 diabetic patients with RH. Conclusions Our results indicate that TRPC6 is a critical sensitive cation channel to hypoglycemia and is a promising target to prevent RH‐induced cognitive impairment by properly orchestrating the mitochondrial dynamics in diabetic patients.
Collapse
Affiliation(s)
- Chengkang He
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Yuanting Cui
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Qiang Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Yingsha Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Huan Ma
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Yu Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Xiaowei Chen
- Brain Research Center, Army Medical University, Chongqing, China
| | - Hongbo Jia
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Gangyi Yang
- Endocrine Department, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| |
Collapse
|
28
|
Tao R, Lu R, Wang J, Zeng S, Zhang T, Guo W, Zhang X, Cheng Q, Yue C, Wang Y, Jing N. Probing the therapeutic potential of TRPC6 for Alzheimer's disease in live neurons from patient-specific iPSCs. J Mol Cell Biol 2020; 12:807-816. [PMID: 32492143 PMCID: PMC7816687 DOI: 10.1093/jmcb/mjaa027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/14/2020] [Accepted: 04/27/2020] [Indexed: 01/16/2023] Open
Abstract
The induced pluripotent stem cells (iPSCs) offer an unprecedented opportunity to model and study Alzheimer's disease (AD) under patient-specific genetic background. The lower expression of transient receptor potential canonical 6 (TRPC6) was associated with AD patients, which might be involved in AD pathogenesis. However, the role of TRPC6 that played in AD process still needs more investigation in patient-relevant neurons. In this study, the iPSCs were generated from peripheral blood cells of sporadic AD patients and efficiently differentiated into mature cortical neurons. These sporadic AD-bearing neurons displayed higher levels of AD pathological markers Aβ and phospho-tau, but lower levels of TRPC6, than those of control neurons. Treatment of AD neurons with TRPC6 protein fragment or agonist inhibited the elevation of Aβ and phospho-tau. Our results in live AD neurons manifest that the compromised expression of TRPC6 substantially contributed to Aβ pathology of sporadic AD, suggesting that targeting TRPC6 could help to develop novel therapeutic strategies for the treatments of AD.
Collapse
Affiliation(s)
- Ran Tao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rui Lu
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junfeng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shujun Zeng
- Department of Neurology, Ruijin Hospital affiliated with the School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.,School of Public Health, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Ting Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenke Guo
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaobing Zhang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, CA 92350, USA
| | - Qi Cheng
- Department of Neurology, Ruijin Hospital affiliated with the School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.,School of Public Health, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Chunmei Yue
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yizheng Wang
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
29
|
Adipose-derived stem cell transplantation improves learning and memory via releasing neurotrophins in rat model of temporal lobe epilepsy. Brain Res 2020; 1750:147121. [PMID: 32919982 DOI: 10.1016/j.brainres.2020.147121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
Epilepsy is a common neurological disease and its most common type is temporal lobe epilepsy (TLE). Novel therapeutics is needed as many TLE patients are resistant to treatments like anticonvulsants or temporal lobectomy. Stem cell therapy has great promise in regeneration medicine. In the current study, we tried to investigate the potential protective effects of adipose-derived stem cell (ADSC) transplantation in epileptic rats. Epilepsy model was established by intra-hippocampal injection of kainic acid (KA) in rats. ADSCs were isolated, differentiated and transplanted into hippocampus of KA rats. There were three groups of rats: normal control group receiving saline injection and no transplantation, KA + sham group receiving KA injection and sham transplantation surgery and KA + transplantation group receiving KA injection and ADSC transplantation. We found that ADSCs were highly positive for CD44, CD90, CD29 and CD105, and neural differentiation induced the expression of neuronal markers like Tuj1, MAP2, NeuN and PSD-95. EEG recording showed that KA induced seizure activity while ADSC transplantation inhibited seizure activity. In training session of Morris water maze task, KA injection impaired the learning capacity of rats while ADSC transplantation restored the learning capacity at 2-week or 2-month post transplantation. In probe session of Morris water maze task, KA injection impaired the memory of rats while ADSC transplantation restored the memory at 2-week or 2-month post transplantation. Transplanted ADSCs released BDNF, NT3 and NT4. Pro-apoptotic BAX was reduced while anti-apoptotic BCL-2 and BCL-xL were increased in hippocampus post ADSC transplantation. Our study suggests that ADSC transplantation inhibits KA-induced seizures and improves the learning and memory function of epileptic rats.
Collapse
|
30
|
Therapeutic potential of pharmacological agents targeting TRP channels in CNS disorders. Pharmacol Res 2020; 159:105026. [PMID: 32562815 DOI: 10.1016/j.phrs.2020.105026] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/21/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
Central nervous system (CNS) disorders like Alzheimer's disease (AD), Parkinson disease (PD), stroke, epilepsy, depression, and bipolar disorder have a high impact on both medical and social problems due to the surge in their prevalence. All of these neuronal disorders share some common etiologies including disruption of Ca2+ homeostasis and accumulation of misfolded proteins. These misfolded proteins further disrupt the intracellular Ca2+ homeostasis by disrupting the activity of several ion channels including transient receptor potential (TRP) channels. TRP channel families include non-selective Ca2+ permeable channels, which act as cellular sensors activated by various physio-chemical stimuli, exogenous, and endogenous ligands responsible for maintaining the intracellular Ca2+ homeostasis. TRP channels are abundantly expressed in the neuronal cells and disturbance in their activity leads to various neuronal diseases. Under the pathological conditions when the activity of TRP channels is perturbed, there is a disruption of the neuronal homeostasis through increased inflammatory response, generation of reactive oxygen species, and mitochondrial dysfunction. Therefore, there is a potential of pharmacological interventions targeting TRP channels in CNS disorders. This review focuses on the role of TRP channels in neurological diseases; also, we have highlighted the current insights into the pharmacological modulators targeting TRP channels.
Collapse
|
31
|
Ma N, Tie C, Yu B, Zhang W, Wan J. Circular RNAs regulate its parental genes transcription in the AD mouse model using two methods of library construction. FASEB J 2020; 34:10342-10356. [PMID: 32530085 DOI: 10.1096/fj.201903157r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/07/2020] [Accepted: 05/20/2020] [Indexed: 01/22/2023]
Abstract
Circular RNA (circRNA) is an important class of noncoding RNA. Current, protocols to detected circRNAs have utilized nonpolyadenylated RNAs, ribosomal RNA-depleted RNA samples (rRNA- library), and rRNA-depleted RNA that has been treated with RNase R to digest linear RNA (rRNA- RNase R+ library). Accumulating evidence suggests the participation of circRNAs in Alzheimer's disease (AD)-associated pathophysiology, but the details remain largely unknown. Here, we elucidated the brain circRNAs profiles of AD-model and WT mice using two methods of library construction (rRNA- RNase R+ libraries and rRNA- libraries). We focused on the construction of libraries that best allow the identification of circRNAs from next-generation RNA sequencing data. We obtained a significantly higher abundance of circRNAs in the rRNA- RNase R+ libraries than in the rRNA- libraries. Additionally, the rRNA- RNase R+ libraries more clearly revealed differentially expressed circRNAs. We performed a correlation analysis between differentially expressed circRNAs and their parental genes and performed KEGG analysis of the parental genes to explore the role of circRNA in AD. Our results identified significantly dysregulated circRNAs and KEGG analysis revealed that the identified circRNAs are involved in regulating AD development from distinct origins, including cAMP signaling, MAPK signaling, insulin secretion, Axon guidance, Long-term potentiation, dopaminergic synapse, and ErBb signaling pathways. Following rigorous selection, we identified several important circRNAs and mRNAs and propose the circRNAs regulate parental gene transcription or affect variable splicing, which were discovered to be mainly involved in Aβ production and cognitive performance (TRPC6) and neuronal differentiation and development (NME7). These newly identified circRNAs should serve as a valuable resource for the future development of potential biomarkers and therapeutic targets for AD.
Collapse
Affiliation(s)
- Nana Ma
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Changrui Tie
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Bo Yu
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wei Zhang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
32
|
Hong C, Jeong B, Park HJ, Chung JY, Lee JE, Kim J, Shin YC, So I. TRP Channels as Emerging Therapeutic Targets for Neurodegenerative Diseases. Front Physiol 2020; 11:238. [PMID: 32351395 PMCID: PMC7174697 DOI: 10.3389/fphys.2020.00238] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
The development of treatment for neurodegenerative diseases (NDs) such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis is facing medical challenges due to the increasingly aging population. However, some pharmaceutical companies have ceased the development of therapeutics for NDs, and no new treatments for NDs have been established during the last decade. The relationship between ND pathogenesis and risk factors has not been completely elucidated. Herein, we review the potential involvement of transient receptor potential (TRP) channels in NDs, where oxidative stress and disrupted Ca2+ homeostasis consequently lead to neuronal apoptosis. Reactive oxygen species (ROS) -sensitive TRP channels can be key risk factors as polymodal sensors, since progressive late onset with secondary pathological damage after initial toxic insult is one of the typical characteristics of NDs. Recent evidence indicates that the dysregulation of TRP channels is a missing link between disruption of Ca2+ homeostasis and neuronal loss in NDs. In this review, we discuss the latest findings regarding TRP channels to provide insights into the research and quests for alternative therapeutic candidates for NDs. As the structures of TRP channels have recently been revealed by cryo-electron microscopy, it is necessary to develop new TRP channel antagonists and reevaluate existing drugs.
Collapse
Affiliation(s)
- Chansik Hong
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Byeongseok Jeong
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Hyung Joon Park
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Ji Yeon Chung
- Department of Neurology, Chosun University School of Medicine, Gwangju, South Korea
| | - Jung Eun Lee
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jinsung Kim
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Young-Cheul Shin
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
| | - Insuk So
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
33
|
Liu L, Gu L, Chen M, Zheng Y, Xiong X, Zhu S. Novel Targets for Stroke Therapy: Special Focus on TRPC Channels and TRPC6. Front Aging Neurosci 2020; 12:70. [PMID: 32256338 PMCID: PMC7093711 DOI: 10.3389/fnagi.2020.00070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke remains a leading cause of death, disability, and medical care burden worldwide. However, transformation from laboratory findings toward effective pharmacological interventions for clinical stroke has been unsatisfactory. Novel evidence has been gained on the underlying mechanisms and therapeutic potential related to the transient receptor potential (TRP) channels in several disorders. The TRP superfamily consists of a diverse group of Ca2+ permeable non-selective cation channels. In particular, the members of TRP subfamilies, TRP canonical (TRPC) channels and TRPC6, have been found in different cell types in the whole body and have high levels of expression in the central nervous system (CNS). Notably, the TRPCs and TRPC6 channel have been implicated in neurite outgrowth and neuronal survival during normal development and in a range of CNS pathological conditions. Recent studies have shown that suppression of TRPC6 channel degradation prevents ischemic neuronal cell death in experimental stroke. Accumulating evidence supports the important functions of TRPC6 in brain ischemia. We have highlighted some crucial advancement that points toward an important involvement of TRPCs and TRPC6 in ischemic stroke. This review will make an overview of the TRP and TRPC channels due to their roles as targets for clinical trials and CNS disorders. Besides, the primary goal is to discuss and update the critical role of TRPC6 channels in stroke and provide a promising target for stroke prevention and therapy.
Collapse
Affiliation(s)
- Lu Liu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Manli Chen
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoxing Xiong
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
34
|
Popugaeva E, Bezprozvanny I, Chernyuk D. Reversal of Calcium Dysregulation as Potential Approach for Treating Alzheimer's Disease. Curr Alzheimer Res 2020; 17:344-354. [PMID: 32469698 PMCID: PMC8210816 DOI: 10.2174/1567205017666200528162046] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 02/25/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Despite decades of research and effort, there is still no effective disease-modifying treatment for Alzheimer's Disease (AD). Most of the recent AD clinical trials were targeting amyloid pathway, but all these trials failed. Although amyloid pathology is a hallmark and defining feature of AD, targeting the amyloid pathway has been very challenging due to low efficacy and serious side effects. Alternative approaches or mechanisms for our understanding of the major cause of memory loss in AD need to be considered as potential therapeutic targets. Increasing studies suggest that Ca2+ dysregulation in AD plays an important role in AD pathology and is associated with other AD abnormalities, such as excessive inflammation, increased ROS, impaired autophagy, neurodegeneration, synapse, and cognitive dysfunction. Ca2+ dysregulation in cytosolic space, Endoplasmic Reticulum (ER) and mitochondria have been reported in the context of various AD models. Drugs or strategies, to correct the Ca2+ dysregulation in AD, have been demonstrated to be promising as an approach for the treatment of AD in preclinical models. This review will discuss the mechanisms of Ca2+ dysregulation in AD and associated pathology and discuss potential approaches or strategies to develop novel drugs for the treatment of AD by targeting Ca2+ dysregulation.
Collapse
Affiliation(s)
- Elena Popugaeva
- Department of Medical Physics, Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg Polytechnic University, St Petersburg, Russia
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, USA
| | - Daria Chernyuk
- Department of Medical Physics, Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg Polytechnic University, St Petersburg, Russia
| |
Collapse
|
35
|
Chen JM, Li QW, Liu JS, Jiang GX, Liu JR, Chen SD, Cheng Q. TRPC6 mRNA levels in peripheral leucocytes of patients with Alzheimer's disease and mild cognitive impairment: A case-control study. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:279-284. [PMID: 30684527 DOI: 10.1016/j.pnpbp.2019.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/14/2019] [Accepted: 01/23/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Transient receptor potential canonical (TRPC) 6 inhibits Aβ in Alzheimer's disease (AD) mouse brain and improves the behavioral performance. AIMS To evaluate the association of TRPC6 expression in peripheral leucocytes from AD and mild cognitive impairment (MCI) patients and to explore its potential value in early diagnosis of AD. METHODS TRPC6 mRNA levels in peripheral leucocytes were detected by quantitative real-time PCR. The Spearman correlation test was used to ascertain the associations between TRPC6 and the scores of MMSE, ADL, CSDD, CDR. The Receiver Operating Characteristic (ROC) curve was drawn to evaluate the diagnostic potential of TRPC6 for AD and MCI. RESULTS There were 108 CE, 136 MCI, 164 Con and 60 PD in the study. The expression of TRPC6 mRNA level in peripheral leucocytes was significantly lower: 1) in patients with AD and MCI compared to Con; 2) in AD compared to MCI; 3) in hospitalized AD compared to AD from communities. There was a significantly positive correlation between TRPC6 mRNA and MMSE score (p = .001, R = 0.327). Significantly inverse correlations were found between TRPC6 and CDR score (p < 0.001, R = -0.303) as well as between TRPC6 and ADL score (p = .001, R = -0.342) for all AD. The area under curve of ROC was 0.881 for the classification of AD, and 0.706 for the classification of MCI, respectively. CONCLUSION TRPC6 expression is inversely correlated with cognitive performance of AD. TRPC6 in peripheral leucocytes may be a potential biomarker for the diagnosis of AD.
Collapse
Affiliation(s)
- Jin-Mei Chen
- Department of Neurology, Ruijin Hospital affiliated with the School of Medicine, Shanghai Jiao Tong University, 197 Ruijin No.2 Road, Shanghai 200025, China; Department of Neurology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Discipline Construction Research Center of China Hospital Development Institute, Shanghai Jiao Tong University, 280 Mohe Road, Shanghai 201999, China
| | - Qing-Wei Li
- Department of Psychiatry, Tongji Hospital, Tongji University School of Medicine, 389 Xin Cun Road, Shanghai 200065, China; Shanghai Mental Health Central, Shanghai Jiao Tong University School of Medicine, 600 Wanping Nan Road, Shanghai 200013, China
| | - Jian-Sheng Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Discipline Construction Research Center of China Hospital Development Institute, Shanghai Jiao Tong University, 280 Mohe Road, Shanghai 201999, China
| | - Guo-Xin Jiang
- Department of Learning, Informatics, Management and Ethics, Karolinska Institute, Stockholm 17177, Sweden
| | - Jian-Ren Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Discipline Construction Research Center of China Hospital Development Institute, Shanghai Jiao Tong University, 280 Mohe Road, Shanghai 201999, China.
| | - Sheng-Di Chen
- Department of Neurology, Ruijin Hospital affiliated with the School of Medicine, Shanghai Jiao Tong University, 197 Ruijin No.2 Road, Shanghai 200025, China.
| | - Qi Cheng
- Department of Neurology, Ruijin Hospital affiliated with the School of Medicine, Shanghai Jiao Tong University, 197 Ruijin No.2 Road, Shanghai 200025, China; School of Public Health, Shanghai Jiao Tong University, 227 Chong Qing Nan Road, Shanghai 200025, China.
| |
Collapse
|
36
|
Mustaly-Kalimi S, Littlefield AM, Stutzmann GE. Calcium Signaling Deficits in Glia and Autophagic Pathways Contributing to Neurodegenerative Disease. Antioxid Redox Signal 2018; 29:1158-1175. [PMID: 29634342 DOI: 10.1089/ars.2017.7266] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Numerous cellular processes and signaling mechanisms have been identified that contribute to Alzheimer's disease (AD) pathology; however, a comprehensive or unifying pathway that binds together the major disease features remains elusive. As an upstream mechanism, altered calcium (Ca2+) signaling is a common driving force for many pathophysiological events that emerge during normal aging and development of neurodegenerative disease. Recent Advances: Over the previous three decades, accumulated evidence has validated the concept that intracellular Ca2+ dysregulation is centrally involved in AD pathogenesis, including the aggregation of pathogenic β-amyloid (Aβ) and phospho-τ species, synapse loss and dysfunction, cognitive impairment, and neurotoxicity. CRITICAL ISSUES Although neuronal Ca2+ signaling within the cytosol and endoplasmic reticulum (ER) has been well studied, other critical central nervous system-resident cell types affected by aberrant Ca2+ signaling, such as astrocytes and microglia, have not been considered as thoroughly. In addition, certain intracellular Ca2+-harboring organelles have been well studied, such as the ER and mitochondria; however other critical Ca2+-regulated organelles, such as lysosomes and autophagosomes, have only more recently been investigated. In this review, we examine Ca2+ dysregulation in microglia and astrocytes, as well as key intracellular organelles important for cellular maintenance and protein handling. Ca2+ dysregulation within these non-neuronal cells and organelles is hypothesized to disrupt the effective clearance of misaggregated proteins and cellular signaling pathways needed for memory networks. FUTURE DIRECTIONS Overall, we aim to explore how these disrupted mechanisms could be involved in AD pathology and consider their role as potential therapeutic targets. Antioxid. Redox Signal. 29, 1158-1175.
Collapse
Affiliation(s)
- Sarah Mustaly-Kalimi
- 1 Department of Neuroscience, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| | - Alyssa M Littlefield
- 1 Department of Neuroscience, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| | - Grace E Stutzmann
- 2 Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| |
Collapse
|
37
|
Ding M, Wang H, Qu C, Xu F, Zhu Y, Lv G, Lu Y, Zhou Q, Zhou H, Zeng X, Zhang J, Yan C, Lin J, Luo HR, Deng Z, Xiao Y, Tian J, Zhu MX, Hong X. Pyrazolo[1,5-a]pyrimidine TRPC6 antagonists for the treatment of gastric cancer. Cancer Lett 2018; 432:47-55. [PMID: 29859875 PMCID: PMC6345172 DOI: 10.1016/j.canlet.2018.05.041] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/09/2018] [Accepted: 05/24/2018] [Indexed: 01/16/2023]
Abstract
Transient receptor potential canonical 6 (TRPC6) proteins form receptor-operated Ca2+-permeable channels, which have been thought to bring benefit to the treatment of diseases, including cancer. However, selective antagonists for TRPC channels are rare and none of them has been tested against gastric cancer. Compound 14a and analogs were synthesized by chemical elaboration of previously reported TRPC3/6/7 agonist 4o. 14a had very weak agonist activity at TRPC6 expressed in HEK293 cells but exhibited strong inhibition on both 4o-mediated and receptor-operated activation of TRPC6 with an IC50 of about 1 μM. When applied to the culture media, 14a suppressed proliferation of AGS and MKN45 cells with IC50 values of 17.1 ± 0.3 and 18.5 ± 1.0 μM, respectively, and inhibited tube formation and migration of cultured human endothelial cells. This anti-tumor effect on gastric cancer was further verified in xenograft models using nude mice. This study has found a new tool compound which shows excellent therapeutic potential against human gastric cancer most likely through targeting TRPC6 channels.
Collapse
Affiliation(s)
- Mingmin Ding
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chunrong Qu
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Fuchun Xu
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China
| | - Yingmin Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Guangyao Lv
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yungang Lu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Qingjun Zhou
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Hui Zhou
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Xiaodong Zeng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Jingwen Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chunhong Yan
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jiacheng Lin
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Huai-Rong Luo
- Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Zixing Deng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Yuling Xiao
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Medical College, Tibet University, Lasa, China; Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, China.
| |
Collapse
|
38
|
Huang M, Qi W, Fang S, Jiang P, Yang C, Mo Y, Dong C, Li Y, Zhong J, Cai W, Yang Z, Zhou T, Wang Q, Yang X, Gao G. Pigment Epithelium-Derived Factor Plays a Role in Alzheimer's Disease by Negatively Regulating Aβ42. Neurotherapeutics 2018; 15:728-741. [PMID: 29736859 PMCID: PMC6095778 DOI: 10.1007/s13311-018-0628-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Pigment epithelium-derived factor (PEDF), a unique neurotrophic protein, decreases with aging. Previous reports have conflicted regarding whether the PEDF concentration is altered in AD patients. In addition, the effect of PEDF on AD has not been documented. Here, we tested serum samples of 31 AD patients and 271 normal controls. We found that compared to PEDF levels in young and middle-aged control subjects, PEDF levels were reduced in old-aged controls and even more so in AD patients. Furthermore, we verified that PEDF expression was much lower and amyloid β-protein (Aβ)42 expression was much higher in senescence-accelerated mouse prone 8 (SAMP8) strain mice than in senescence-accelerated mouse resistant 1 (SAMR1) control strain mice. Accordingly, high levels of Aβ42 were also observed in PEDF knockout (KO) mice. PEDF notably reduced cognitive impairment in the Morris water maze (MWM) and significantly downregulated Aβ42 in SAMP8 mice. Mechanistically, PEDF downregulated presenilin-1 (PS1) expression by inhibiting the c-Jun N-terminal kinase (JNK) pathway. Taken together, our findings demonstrate for the first time that PEDF negatively regulates Aβ42 and that PEDF deficiency with aging might play a crucial role in the development of AD.
Collapse
Affiliation(s)
- Mao Huang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Weiwei Qi
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shuhuan Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping Jiang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Cong Yang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yousheng Mo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang Dong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yan Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Jun Zhong
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weibin Cai
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zhonghan Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ti Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xia Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
- China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Guoquan Gao
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
39
|
Wang Q, He Q, Chen Y, Shao W, Yuan C, Wang Y. JNK-mediated microglial DICER degradation potentiates inflammatory responses to induce dopaminergic neuron loss. J Neuroinflammation 2018; 15:184. [PMID: 29907159 PMCID: PMC6003208 DOI: 10.1186/s12974-018-1218-1] [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: 01/24/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Background Amplified inflammation is important for the progression of Parkinson’s disease (PD). However, how this enhanced inflammation is regulated remains largely unknown. Deletion of DICER leads to progressive dopamine neuronal loss and induces gliosis. We hypothesized that the homeostasis of microglial DICER would be responsible for the amplified inflammation in the mouse model of PD. Methods The microglia or C57BL/6 mice were treated or injected with l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium (MPP+), respectively, for the model establishment. Microglia and astrocytes sorted by fluorescence-activated cell sorter (FACS) were assayed by quantitative real-time PCR, Western blotting, immunoprecipitation, enzyme-linked immunosorbent assay (ELISA), immunohistofluorescence, and mass spectrometry. Results Microglial DICER was phosphorylated at serine 1456 by c-jun N-terminal kinase (JNK) and downregulated in response to 1-methyl-4-phenylpyridinium (MPP+), a causative agent in PD. Inhibition of JNK phosphorylation of DICER at serine 1456 rescued the MPP+-induced DICER degradation, suppressed microglial inflammatory process, and prevented the loss of tyrosine hydroxylase-expressing neurons in the mouse MPTP model. Conclusions JNK-mediated microglial DICER degradation potentiates inflammation to induce dopaminergic neuronal loss. Thus, preventing microglial DICER degradation could be a novel strategy for controlling neuroinflammation in PD. Electronic supplementary material The online version of this article (10.1186/s12974-018-1218-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Wang
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Qian He
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Yifei Chen
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Wei Shao
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Chao Yuan
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, 100000, People's Republic of China
| | - Yizheng Wang
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
40
|
Liu Q, Wang SC, Ding K. Research advances in the treatment of Alzheimer's disease with polysaccharides from traditional Chinese medicine. Chin J Nat Med 2018; 15:641-652. [PMID: 28991525 DOI: 10.1016/s1875-5364(17)30093-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the loss of patients' memory and their cognitive abilities and the mechanism is not completely clear. Although a variety of drugs have been approved for the AD treatment, substances which can prevent and cure AD are still in great need. The effect of polysaccharides from traditional Chinese medicine (TCM) on anti-AD has gained great progress and attained more and more attention in recent years. In this review, research advances in TCM-polysaccharides on AD made in this decade are summarized.
Collapse
Affiliation(s)
- Qin Liu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Glycochemistry and Glycobiology Lab, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shun-Chun Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Kan Ding
- Glycochemistry and Glycobiology Lab, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
41
|
Li M, Liu E, Zhou Q, Li S, Wang X, Liu Y, Wang L, Sun D, Ye J, Gao Y, Yang X, Liu J, Yang Y, Wang JZ. TRPC1 Null Exacerbates Memory Deficit and Apoptosis Induced by Amyloid-β. J Alzheimers Dis 2018; 63:761-772. [DOI: 10.3233/jad-180077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Mengzhu Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Enjie Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuzhi Zhou
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shihong Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanchao Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongsheng Sun
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinwang Ye
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, and Center for Disease Control and Prevention, Shenzhen, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, and Center for Disease Control and Prevention, Shenzhen, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Key Laboratory of Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
42
|
Secondo A, Bagetta G, Amantea D. On the Role of Store-Operated Calcium Entry in Acute and Chronic Neurodegenerative Diseases. Front Mol Neurosci 2018; 11:87. [PMID: 29623030 PMCID: PMC5874322 DOI: 10.3389/fnmol.2018.00087] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022] Open
Abstract
In both excitable and non-excitable cells, calcium (Ca2+) signals are maintained by a highly integrated process involving store-operated Ca2+ entry (SOCE), namely the opening of plasma membrane (PM) Ca2+ channels following the release of Ca2+ from intracellular stores. Upon depletion of Ca2+ store, the stromal interaction molecule (STIM) senses Ca2+ level reduction and migrates from endoplasmic reticulum (ER)-like sites to the PM where it activates the channel proteins Orai and/or the transient receptor potential channels (TRPC) prompting Ca2+ refilling. Accumulating evidence suggests that SOCE dysregulation may trigger perturbation of intracellular Ca2+ signaling in neurons, glia or hematopoietic cells, thus participating to the pathogenesis of diverse neurodegenerative diseases. Under acute conditions, such as ischemic stroke, neuronal SOCE can either re-establish Ca2+ homeostasis or mediate Ca2+ overload, thus providing a non-excitotoxic mechanism of ischemic neuronal death. The dualistic role of SOCE in brain ischemia is further underscored by the evidence that it also participates to endothelial restoration and to the stabilization of intravascular thrombi. In Parkinson's disease (PD) models, loss of SOCE triggers ER stress and dysfunction/degeneration of dopaminergic neurons. Disruption of neuronal SOCE also underlies Alzheimer's disease (AD) pathogenesis, since both in genetic mouse models and in human sporadic AD brain samples, reduced SOCE contributes to synaptic loss and cognitive decline. Unlike the AD setting, in the striatum from Huntington's disease (HD) transgenic mice, an increased STIM2 expression causes elevated synaptic SOCE that was suggested to underlie synaptic loss in medium spiny neurons. Thus, pharmacological inhibition of SOCE is beneficial to synapse maintenance in HD models, whereas the same approach may be anticipated to be detrimental to cortical and hippocampal pyramidal neurons. On the other hand, up-regulation of SOCE may be beneficial during AD. These intriguing findings highlight the importance of further mechanistic studies to dissect the molecular pathways, and their corresponding targets, involved in synaptic dysfunction and neuronal loss during aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Napoli, Italy
| | - Giacinto Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, Section of Preclinical and Translational Pharmacology, University of Calabria, Cosenza, Italy
| | - Diana Amantea
- Department of Pharmacy, Health and Nutritional Sciences, Section of Preclinical and Translational Pharmacology, University of Calabria, Cosenza, Italy
| |
Collapse
|
43
|
Lu R, Wang J, Tao R, Wang J, Zhu T, Guo W, Sun Y, Li H, Gao Y, Zhang W, Fowler CJ, Li Q, Chen S, Wu Z, Masters CL, Zhong C, Jing N, Wang Y, Wang Y. Reduced TRPC6 mRNA levels in the blood cells of patients with Alzheimer's disease and mild cognitive impairment. Mol Psychiatry 2018; 23:767-776. [PMID: 28696436 DOI: 10.1038/mp.2017.136] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 04/24/2017] [Accepted: 05/10/2017] [Indexed: 02/01/2023]
Abstract
Transient receptor potential canonical 6 (TRPC6) inhibits β-amyloid (Aβ) production. Hyperforin, the TRPC6 agonist, reduces Aβ levels and improves cognitive performance in Alzheimer's disease (AD) models. However, it's unknown whether TRPC6 expression is changed in AD patients. In this case-control study, we measured TRPC6 expression levels in the peripheral blood cells of four independent AD sets from five hospitals and one mild cognitive impairment (MCI) set from a local community (229 AD, 70 MCI, 40 Parkinson disease and 359 controls from China, total n=698) using quantitative real-time PCR assay. We found a specific reduction of TRPC6 mRNA levels in four AD sets and one MCI set. The median TRPC6 mRNA levels were lower in the following: (1) combined AD patients than in age-matched controls (0.78 vs 1.73, P<0.001); (2) mild-to-moderate AD patients than in age-matched controls (0.81 vs 1.73, P<0.001); and (3) MCI patients than in age-matched controls (0.76 vs 1.72, P<0.001). In the receiver-operating characteristic curve analysis, the area under curve was 0.85 for combined AD, 0.84 for mild-to-moderate AD and 0.79 for MCI. In a subgroup of AD patients with brain Aβ examination, TRPC6 was associated with standardized uptake value ratio of Pittsburgh Compound B (Spearman's r=-0.49, P=0.04) and cerebrospinal fluid Aβ42 (Spearman's r=0.43, P=0.04). The TRPC6 reduction in AD patients was further confirmed in blood RNA samples from The Australian Imaging, Biomarkers and Lifestyle Flagship Study of Aging, in post-mortem brain tissues from The Netherlands Brain Bank and in induced pluripotent stem cells-derived neurons from Chinese donors. We conclude that TRPC6 mRNA levels in the blood cells are specifically reduced in AD and MCI patients, and TRPC6 might be a biomarker for the early diagnosis of AD.
Collapse
Affiliation(s)
- R Lu
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Graduate School of Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,Beijing Institute of Medical Sciences, Beijing, China
| | - J Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - R Tao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - J Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - T Zhu
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - W Guo
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Y Sun
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - H Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Y Gao
- Department of Neurology and Institute of Neurology, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - W Zhang
- Department of Geriatrics, Tiantan Hospital, Capital Medical University, Beijing, China
| | - C J Fowler
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Q Li
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - S Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Z Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - C L Masters
- The Florey Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - C Zhong
- Department of Neurology, Zhongshan Hospital and Shanghai Medical College, State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Fudan University, Shanghai, China
| | - N Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Y Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Beijing Institute of Medical Sciences, Beijing, China
| |
Collapse
|
44
|
Reboreda A, Theissen FM, Valero-Aracama MJ, Arboit A, Corbu MA, Yoshida M. Do TRPC channels support working memory? Comparing modulations of TRPC channels and working memory through G-protein coupled receptors and neuromodulators. Behav Brain Res 2018; 354:64-83. [PMID: 29501506 DOI: 10.1016/j.bbr.2018.02.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 02/27/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Working memory is a crucial ability we use in daily life. However, the cellular mechanisms supporting working memory still remain largely unclear. A key component of working memory is persistent neural firing which is believed to serve short-term (hundreds of milliseconds up to tens of seconds) maintenance of necessary information. In this review, we will focus on the role of transient receptor potential canonical (TRPC) channels as a mechanism underlying persistent firing. Many years of in vitro work have been suggesting a crucial role of TRPC channels in working memory and temporal association tasks. If TRPC channels are indeed a central mechanism for working memory, manipulations which impair or facilitate working memory should have a similar effect on TRPC channel modulation. However, modulations of working memory and TRPC channels were never systematically compared, and it remains unanswered whether TRPC channels indeed contribute to working memory in vivo or not. In this article, we review the effects of G-protein coupled receptors (GPCR) and neuromodulators, including acetylcholine, noradrenalin, serotonin and dopamine, on working memory and TRPC channels. Based on comparisons, we argue that GPCR and downstream signaling pathways that activate TRPC, generally support working memory, while those that suppress TRPC channels impair it. However, depending on the channel types, areas, and systems tested, this is not the case in all studies. Further work to clarify involvement of specific TRPC channels in working memory tasks and how they are affected by neuromodulators is still necessary in the future.
Collapse
Affiliation(s)
- Antonio Reboreda
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany.
| | - Frederik M Theissen
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Maria J Valero-Aracama
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 17, 91054 Erlangen, Germany
| | - Alberto Arboit
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany
| | - Mihaela A Corbu
- Ruhr University Bochum (RUB), Universitätsstraße 150, 44801, Bochum, Germany
| | - Motoharu Yoshida
- Leibniz Institute for Neurobiology (LIN) Magdeburg, Brenneckestraße 6, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str. 44/Haus 64, 39120, Magdeburg, Germany; Center for Behavioral Brain Sciences, 39106, Magdeburg, Germany.
| |
Collapse
|
45
|
Qu Z, Wang Y, Li X, Wu L, Wang Y. TRPC6 expression in neurons is differentially regulated by NR2A- and NR2B-containing NMDA receptors. J Neurochem 2017; 143:282-293. [PMID: 28902407 DOI: 10.1111/jnc.14215] [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: 06/07/2017] [Revised: 08/30/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
The expression of transient receptor potential canonical 6 (TRPC6) in central nervous system (CNS) is important for neuronal functions and certain neural disorders. However, the regulatory mechanism of TRPC6 expression in neurons is still obscure. In this study, we show that TRPC6 expression in the primary cultured cortical neurons is bidirectionally regulated by glutamate. Activation of NR2A-containing NMDARs induces TRPC6 transcription through a calcineurin-dependent pathway. In contrast, activation of NR2B-containing NMDARs causes TRPC6 degradation through calpain. Thus, TRPC6 expression in neurons is regulated by glutamate in a bidirectional manner that is dependent on NR2A and NR2B.
Collapse
Affiliation(s)
- Zhongwei Qu
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,The Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuqing Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,The Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Xia Li
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| | - Lin Wu
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| |
Collapse
|
46
|
Store-Operated Calcium Channel Complex in Postsynaptic Spines: A New Therapeutic Target for Alzheimer's Disease Treatment. J Neurosci 2017; 36:11837-11850. [PMID: 27881772 DOI: 10.1523/jneurosci.1188-16.2016] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/08/2016] [Accepted: 09/11/2016] [Indexed: 12/11/2022] Open
Abstract
Mushroom dendritic spine structures are essential for memory storage and the loss of mushroom spines may explain memory defects in aging and Alzheimer's disease (AD). The stability of mushroom spines depends on stromal interaction molecule 2 (STIM2)-mediated neuronal-store-operated Ca2+ influx (nSOC) pathway, which is compromised in AD mouse models, in aging neurons, and in sporadic AD patients. Here, we demonstrate that the Transient Receptor Potential Canonical 6 (TRPC6) and Orai2 channels form a STIM2-regulated nSOC Ca2+ channel complex in hippocampal mushroom spines. We further demonstrate that a known TRPC6 activator, hyperforin, and a novel nSOC positive modulator, NSN21778 (NSN), can stimulate activity of nSOC pathway in the spines and rescue mushroom spine loss in both presenilin and APP knock-in mouse models of AD. We further show that NSN rescues hippocampal long-term potentiation impairment in APP knock-in mouse model. We conclude that the STIM2-regulated TRPC6/Orai2 nSOC channel complex in dendritic mushroom spines is a new therapeutic target for the treatment of memory loss in aging and AD and that NSN is a potential candidate molecule for therapeutic intervention in brain aging and AD. SIGNIFICANCE STATEMENT Mushroom dendritic spine structures are essential for memory storage and the loss of mushroom spines may explain memory defects in Alzheimer's disease (AD). This study demonstrated that Transient Receptor Potential Canonical 6 (TRPC6) and Orai2 form stromal interaction molecule 2 (STIM2)-regulated neuronal-store-operated Ca2+ influx (nSOC) channel complex in hippocampal synapse and the resulting Ca2+ influx is critical for long-term maintenance of mushroom spines in hippocampal neurons. A novel nSOC-positive modulator, NSN21778 (NSN), rescues mushroom spine loss and synaptic plasticity impairment in AD mice models. The TRPC6/Orai2 nSOC channel complex is a new therapeutic target and NSN is a potential candidate molecule for therapeutic intervention in brain aging and AD.
Collapse
|
47
|
Qu C, Ding M, Zhu Y, Lu Y, Du J, Miller M, Tian J, Zhu J, Xu J, Wen M, Er-Bu AGA, Wang J, Xiao Y, Wu M, McManus OB, Li M, Wu J, Luo HR, Cao Z, Shen B, Wang H, Zhu MX, Hong X. Pyrazolopyrimidines as Potent Stimulators for Transient Receptor Potential Canonical 3/6/7 Channels. J Med Chem 2017; 60:4680-4692. [PMID: 28395140 PMCID: PMC5720685 DOI: 10.1021/acs.jmedchem.7b00304] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Transient receptor potential canonical 3/6/7 (TRPC3/6/7) are highly homologous receptor-operated nonselective cation channels. Despite their physiological significance, very few selective and potent agonists are available for functional examination of these channels. Using a cell-based high throughput screening approach, a lead compound with the pyrazolopyrimidine skeleton was identified as a TRPC6 agonist. Synthetic schemes for the lead and its analogues were established, and structural-activity relationship studies were carried out. A series of potent and direct agonists of TRPC3/6/7 channels were identified, and among them, 4m-4p have a potency order of TRPC3 > C7 > C6, with 4n being the most potent with an EC50 of <20 nM on TRPC3. Importantly, these compounds exhibited no stimulatory activity on related TRP channels. The potent and selective compounds described here should be suitable for evaluation of the roles of TRPC channels in the physiology and pathogenesis of diseases, including glomerulosclerosis and cancer.
Collapse
Affiliation(s)
- Chunrong Qu
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei Province 430071, China
| | - Mingmin Ding
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei Province 430071, China
| | - Yingmin Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Yungang Lu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Juan Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui Province 230032, China
| | - Melissa Miller
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Jinmei Zhu
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei Province 430071, China
| | - Jian Xu
- State Key Laboratory of Natural Medicines, Jiangsu Provincial Key laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing, Jiangsu Province 211198, China
- The International Scientist Working Station of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Meng Wen
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei Province 430071, China
| | - AGA Er-Bu
- Medical College, Tibet University, Lasa, Tibet 850000, China
| | - Jule Wang
- Medical College, Tibet University, Lasa, Tibet 850000, China
| | - Yuling Xiao
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei Province 430071, China
| | - Meng Wu
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Owen B. McManus
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Min Li
- Department of Neuroscience, High Throughput Biology Center and Johns Hopkins Ion Channel Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Jilin Wu
- The International Scientist Working Station of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huai-Rong Luo
- Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan Province 650201, China
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines, Jiangsu Provincial Key laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing, Jiangsu Province 211198, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui Province 230032, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, Shangdong Province 264005, China
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas 77030, United States
- The International Scientist Working Station of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei Province 430071, China
- Medical College, Tibet University, Lasa, Tibet 850000, China
| |
Collapse
|
48
|
Liu N, Zhuang Y, Zhou Z, Zhao J, Chen Q, Zheng J. NF-κB dependent up-regulation of TRPC6 by Aβ in BV-2 microglia cells increases COX-2 expression and contributes to hippocampus neuron damage. Neurosci Lett 2017; 651:1-8. [PMID: 28458019 DOI: 10.1016/j.neulet.2017.04.056] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/15/2017] [Accepted: 04/27/2017] [Indexed: 01/26/2023]
Abstract
The deposition of amyloid β-protein (Aβ) has been involved in neurodegeneration of Alzheimer's disease (AD). Besides Aβ plaques and neuronal loss, microglia activation is also common in AD patient brains, suggesting its important role in the pathogenesis of AD. Although activation of microglia by Aβ plaques has been demonstrated, the mechanism underlying it is still largely unclear. Here, we found that TRPC6 has a crucial role in microglia activation by Aβ. Aβ up-regulates the level of TRPC6 via NF-κB in BV-2 microglia and increases the expression of pro-inflammatory factors and oxidative enzyme, COX-2. Knock-down of TRPC6 reduces the Aβ-induced expression of pro-inflammatory factors and COX-2 and the damage of hippocampus neurons. Furthermore, inhibition of COX-2 also protects hippocampus neurons from Aβ-induced inflammatory damage. Collectively, our studies suggest that Aβ increase the expression of TRPC6 via NF-κB in BV-2 microglia and promotes the production of COX-2, which induces hippocampus neuron damage.
Collapse
Affiliation(s)
- Na Liu
- Department of Neurology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Yuansu Zhuang
- Department of Neurology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Zhikui Zhou
- Department of Neurology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Jinhua Zhao
- Department of Neurology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Qiaoyun Chen
- Department of Central Laboratory, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, Jiangsu, China
| | - Jinxu Zheng
- Department of Respiratory, Affiliated Hospital of Jiangsu University, 81 Jiangbin Road, Zhenjiang 212001, Jiangsu, China.
| |
Collapse
|
49
|
Downregulation of RBO-PI4KIIIα Facilitates Aβ 42 Secretion and Ameliorates Neural Deficits in Aβ 42-Expressing Drosophila. J Neurosci 2017; 37:4928-4941. [PMID: 28424219 DOI: 10.1523/jneurosci.3567-16.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/12/2017] [Accepted: 03/31/2017] [Indexed: 12/25/2022] Open
Abstract
Phosphoinositides and their metabolizing enzymes are involved in Aβ42 metabolism and Alzheimer's disease pathogenesis. In yeast and mammals, Eighty-five requiring 3 (EFR3), whose Drosophila homolog is Rolling Blackout (RBO), forms a plasma membrane-localized protein complex with phosphatidylinositol-4-kinase Type IIIα (PI4KIIIα) and a scaffold protein to tightly control the level of plasmalemmal phosphatidylinositol-4-phosphate (PI4P). Here, we report that RBO binds to Drosophila PI4KIIIα, and that in an Aβ42-expressing Drosophila model, separate genetic reduction of PI4KIIIα and RBO, or pharmacological inhibition of PI4KIIIα ameliorated synaptic transmission deficit, climbing ability decline, premature death, and reduced neuronal accumulation of Aβ42 Moreover, we found that RBO-PI4KIIIa downregulation increased neuronal Aβ42 release and that PI4P facilitated the assembly or oligomerization of Aβ42 in/on liposomes. These results indicate that RBO-PI4KIIIa downregulation facilitates neuronal Aβ42 release and consequently reduces neuronal Aβ42 accumulation likely via decreasing Aβ42 assembly in/on plasma membrane. This study suggests the RBO-PI4KIIIα complex as a potential therapeutic target and PI4KIIIα inhibitors as drug candidates for Alzheimer's disease treatment.SIGNIFICANCE STATEMENT Phosphoinositides and their metabolizing enzymes are involved in Aβ42 metabolism and Alzheimer's disease pathogenesis. Here, in an Aβ42-expressing Drosophila model, we discovered and studied the beneficial role of downregulating RBO or its interacting protein PI4KIIIα-a protein that tightly controls the plasmalemmal level of PI4P-against the defects caused by Aβ42 expression. Mechanistically, RBO-PI4KIIIα downregulation reduced neuronal Aβ42 accumulation, and interestingly increased neuronal Aβ42 release. This study suggests the RBO-PI4KIIIα complex as a novel therapeutic target, and PI4KIIIα inhibitors as new drug candidates.
Collapse
|
50
|
Abstract
Alzheimer's disease (AD) is the primary cause of dementia in the elderly. It remains incurable and poses a huge socio-economic challenge for developed countries with an aging population. AD manifests by progressive decline in cognitive functions and alterations in behaviour, which are the result of the extensive degeneration of brain neurons. The AD pathogenic mechanism involves the accumulation of amyloid beta peptide (Aβ), an aggregating protein fragment that self-associates to form neurotoxic fibrils that trigger a cascade of cellular events leading to neuronal injury and death. Researchers from academia and the pharmaceutical industry have pursued a rational approach to AD drug discovery and targeted the amyloid cascade. Schemes have been devised to prevent the overproduction and accumulation of Aβ in the brain. The extensive efforts of the past 20 years have been translated into bringing new drugs to advanced clinical trials. The most progressed mechanism-based therapies to date consist of immunological interventions to clear Aβ oligomers, and pharmacological drugs to inhibit the secretase enzymes that produce Aβ, namely β-site amyloid precursor-cleaving enzyme (BACE) and γ-secretase. After giving an update on the development and current status of new AD therapeutics, this review will focus on BACE inhibitors and, in particular, will discuss the prospects of verubecestat (MK-8931), which has reached phase III clinical trials.
Collapse
Affiliation(s)
- Genevieve Evin
- Florey Institute of Neuroscience and Mental Health, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|