51
|
Phosphodiesterase 7(PDE7): A unique drug target for central nervous system diseases. Neuropharmacology 2021; 196:108694. [PMID: 34245775 DOI: 10.1016/j.neuropharm.2021.108694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/11/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022]
Abstract
Phosphodiesterase 7 (PDE7), one of the 11 phosphodiesterase (PDE) families, specifically hydrolyzes cyclic 3', 5'-adenosine monophosphate (cAMP). PDE7 is involved in many important functional processes in physiology and pathology by regulating intracellular cAMP signaling. Studies have demonstrated that PDE7 is widely expressed in the central nervous system (CNS) and potentially related to pathogenesis of many CNS diseases. Here, we summarized the classification and distribution of PDE7 in the brain and its functional roles in the mediation of CNS diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and schizophrenia. It is expected that the findings collected here will not only lead to a better understanding of the mechanisms by which PDE7 mediates CNS function and diseases, but also aid in the development of novel drugs targeting PDE7 for treatment of CNS diseases.
Collapse
|
52
|
Price BR, Johnson LA, Norris CM. Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease. Ageing Res Rev 2021; 68:101335. [PMID: 33812051 PMCID: PMC8168445 DOI: 10.1016/j.arr.2021.101335] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/21/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023]
Abstract
Astrocyte reactivity is a hallmark of neuroinflammation that arises with Alzheimer’s disease (AD) and nearly every other neurodegenerative condition. While astrocytes certainly contribute to classic inflammatory processes (e.g. cytokine release, waste clearance, and tissue repair), newly emerging technologies for measuring and targeting cell specific activities in the brain have uncovered essential roles for astrocytes in synapse function, brain metabolism, neurovascular coupling, and sleep/wake patterns. In this review, we use a holistic approach to incorporate, and expand upon, classic neuroinflammatory concepts to consider how astrocyte dysfunction/reactivity modulates multiple pathological and clinical hallmarks of AD. Our ever-evolving understanding of astrocyte signaling in neurodegeneration is not only revealing new drug targets and treatments for dementia but is suggesting we reimagine AD pathophysiological mechanisms.
Collapse
Affiliation(s)
- Brittani R Price
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Lance A Johnson
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Physiology, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA.
| |
Collapse
|
53
|
Qiao O, Zhang X, Zhang Y, Ji H, Li Z, Han X, Wang W, Li X, Wang J, Liu C, Gao W. Cerebralcare Granule® enhances memantine hydrochloride efficacy in APP/PS1 mice by ameliorating amyloid pathology and cognitive functions. Chin Med 2021; 16:47. [PMID: 34183022 PMCID: PMC8240384 DOI: 10.1186/s13020-021-00456-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/08/2021] [Indexed: 01/17/2023] Open
Abstract
Background Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by memory deficits and cognitive decline. Current drugs can only relieve symptoms, but cannot really cure AD. Cerebralcare Granule® (CG) is a Traditional Chinese medicine (TCM) containing a variety of biologically active compounds. In our previous studies, CG has shown a beneficial effect against memory impairment in mice caused by d-galactose. However, whether CG can be used as a complementary medicine for the treatment of AD remains unexplored. Here, we use a combination of CG and memantine hydrochloride (Mm) to treat Alzheimer-like pathology and investigate the effects and mechanisms in vivo. Methods The histology of brain was examined with Hematoxylin–eosin (HE) staining, Golgi staining and Thioflavin S staining. ELISA was applied to assess the expression levels or activities of CAT, SOD, GSH-Px, MDA, alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), total bilirubin (TBIL) in serum, as well as the levels of IL-6, IL-1β, and TNF-α in the mice brain. Western blotting was used to assess the expression of β-secretase (BACE1), amyloid precursor protein (APP), APPβ, APPα, synaptophysin (SYN), growth-associated protein 43 (GAP43), and postsynaptic density 95 (PSD95). Results In the present study, the combination group (CG + Mm) significantly attenuated Alzheimer-like behavior without adverse effects in APP/PS1 mice, indicating its high degree of safety and efficacy after long-term treatment. CG + Mm reduced AD pathological biomarker Aβ plaque accumulation by inhibiting BACE1 and APP expression (P < 0.05 or P < 0.001). Besides, the combination group markedly inhibited the levels of IL-1β, IL-6, and TNF-α in hippocampus (P < 0.001), as well as activities of SOD, CAT, and GSH-Px in serum (P < 0.001). By contrast, the combination group improved synaptic plasticity by enhancing SYN, PSD95, and GAP43 expression. Conclusions Taken together, these data supported the notion that CG combined with Mm might ameliorate the cognitive impairment through multiple pathways, suggesting that CG could play a role as complementary medicine to increase anti-AD effect of chemical drugs by reducing Aβ deposition, neuroinflammation, oxidative damage, and improving synaptic plasticity.
Collapse
Affiliation(s)
- Ou Qiao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Xinyu Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Yi Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Haixia Ji
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Zhi Li
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Xiaoying Han
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Wenzhe Wang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Xia Li
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Juan Wang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China
| | - Changxiao Liu
- The State Key Laboratories of Pharmacodynamics and Pharmacokinetics, Tianjin, 300193, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin, 300072, China.
| |
Collapse
|
54
|
Sadasivam P, Fang XT, Toyonaga T, Lee S, Xu Y, Zheng MQ, Spurrier J, Huang Y, Strittmatter SM, Carson RE, Cai Z. Quantification of SV2A Binding in Rodent Brain Using [ 18F]SynVesT-1 and PET Imaging. Mol Imaging Biol 2021; 23:372-381. [PMID: 33258040 PMCID: PMC8105262 DOI: 10.1007/s11307-020-01567-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Synapse loss is a hallmark of Alzheimer's disease (AD) and correlates with cognitive decline. The validation of a noninvasive in vivo imaging approach to quantify synapse would greatly facilitate our understanding of AD pathogenesis and assist drug developments for AD. As animal models of neurodegenerative and neuropsychiatric disorders play a critical role in the drug discovery and development process, a robust, objective, and translational method for quantifying therapeutic drug efficacy in animal models will facilitate the drug development process. In this study, we tested the quantification reliability of the SV2A PET tracer, [18F]SynVesT-1, in a mouse model of AD (APP/PS1) and wild-type controls, and developed a simplified quantification method to facilitate large cohort preclinical imaging studies. PROCEDURES We generated nondisplaceable binding potential (BPND) and distribution volume ratio (DVR) values using the simplified reference tissue model (SRTM) on the 90-min dynamic PET imaging data, with brain stem and cerebellum as the reference region, respectively. Then, we correlated the standardized uptake value ratio (SUVR)-1 and SUVR averaged from different imaging windows with BPND and DVR, using brain stem and cerebellum as the reference region, respectively. We performed homologous competitive binding assay and autoradiographic saturation binding assay using [18F]SynVesT-1 to calculate the Bmax and Kd. RESULTS Using brain stem as the reference region, the averaged SUVR-1 from 30 to 60 min postinjection correlated well with the BPND calculated using SRTM. Using cerebellum as the reference region, the averaged SUVR from 30 to 60 min postinjection correlated well with the SRTM DVR. From the homologous competitive binding assay and autoradiographic saturation binding assay, the calculated the Bmax and Kd were 4.5-18 pmol/mg protein and 9.8-19.6 nM, respectively, for rodent brain tissue. CONCLUSIONS This simplified SUVR method provides reasonable SV2A measures in APP/PS1 mice and their littermate controls. Our data indicate that, in lieu of a full 90-min dynamic scan, a 30-min static PET scan (from 30 to 60 min postinjection) would be sufficient to provide quantification data on SV2A expression, equivalent to the data generated from kinetic modeling. The methods developed here are readily applicable to the evaluation of therapeutic effects of novel drugs in this rodent model using [18F]SynVesT-1 and small animal PET.
Collapse
Affiliation(s)
- Pragalath Sadasivam
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Xiaotian T. Fang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Takuya Toyonaga
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Supum Lee
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Yuping Xu
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Ming-Qiang Zheng
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Joshua Spurrier
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Cell Biology, Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Yiyun Huang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Stephen M. Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Cell Biology, Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Richard E. Carson
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT
| | - Zhengxin Cai
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
55
|
Bréchet L, Yu W, Biagi MC, Ruffini G, Gagnon M, Manor B, Pascual-Leone A. Patient-Tailored, Home-Based Non-invasive Brain Stimulation for Memory Deficits in Dementia Due to Alzheimer's Disease. Front Neurol 2021; 12:598135. [PMID: 34093384 PMCID: PMC8173168 DOI: 10.3389/fneur.2021.598135] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 04/20/2021] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is an irreversible, progressive brain disorder that can cause dementia (Alzheimer's disease-related dementia, ADRD) with growing cognitive disability and vast physical, emotional, and financial pressures not only on the patients but also on caregivers and families. Loss of memory is an early and very debilitating symptom in AD patients and a relevant predictor of disease progression. Data from rodents, as well as human studies, suggest that dysregulation of specific brain oscillations, particularly in the hippocampus, is linked to memory deficits. Animal and human studies demonstrate that non-invasive brain stimulation (NIBS) in the form of transcranial alternating current stimulation (tACS) allows to reliably and safely interact with ongoing oscillatory patterns in the brain in specific frequencies. We developed a protocol for patient-tailored home-based tACS with an instruction program to train a caregiver to deliver daily sessions of tACS that can be remotely monitored by the study team. We provide a discussion of the neurobiological rationale to modulate oscillations and a description of the study protocol. Data of two patients with ADRD who have completed this protocol illustrate the feasibility of the approach and provide pilot evidence on the safety of the remotely-monitored, caregiver-administered, home-based tACS intervention. These findings encourage the pursuit of a large, adequately powered, randomized controlled trial of home-based tACS for memory dysfunction in ADRD.
Collapse
Affiliation(s)
- Lucie Bréchet
- Hinda and Arthur Marcus Institute for Aging Research and Center for Memory Health, Hebrew SeniorLife, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Wanting Yu
- Hinda and Arthur Marcus Institute for Aging Research and Center for Memory Health, Hebrew SeniorLife, Boston, MA, United States
| | | | - Giulio Ruffini
- Neuroelectrics Barcelona, Barcelona, Spain
- Neuroelectrics Corp., Cambridge, MA, United States
| | - Margaret Gagnon
- Hinda and Arthur Marcus Institute for Aging Research and Center for Memory Health, Hebrew SeniorLife, Boston, MA, United States
| | - Brad Manor
- Hinda and Arthur Marcus Institute for Aging Research and Center for Memory Health, Hebrew SeniorLife, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Alvaro Pascual-Leone
- Hinda and Arthur Marcus Institute for Aging Research and Center for Memory Health, Hebrew SeniorLife, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Guttmann Brain Health Institute, Institut Guttman de Neurorehabilitació, Barcelona, Spain
| |
Collapse
|
56
|
Pei YA, Davies J, Zhang M, Zhang HT. The Role of Synaptic Dysfunction in Alzheimer's Disease. J Alzheimers Dis 2021; 76:49-62. [PMID: 32417776 DOI: 10.3233/jad-191334] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Deemed as incurable, Alzheimer's disease (AD) research is becoming less convoluted as our understanding of its pathology increases. With current treatments focusing on merely mitigating the symptoms of AD, there have been many attempts to find a molecular culprit to serve as the single underlying cause and therapeutic target for clinical applications to approach the disease from its roots. Indeed, over the course of decades, the endless search for a singular target culprit in AD has uncovered a cascade of pathological defects, adding on to each other throughout the progression of the disease. The developmental patterns of amyloid-β (Aβ) oligomers have been studied as a means to discover the complex molecular interplay between various immune responses, genetic mutations, pathway disturbances, and regulating factors that disturb synapse homeostasis before disease manifestation. This new understanding has shifted the underlying goal of the research community from merely removing Aβ oligomers to finding methods that can predict high risk individuals and resorting to cocktail-drug treatments in an attempt to regulate multiple pathways that cumulatively result in the debilitating symptoms of the disease. By utilizing various assays from immuno-targeting to molecular biomarkers, we then interfere in the molecular cascades in an endeavor to avoid synapse dysfunction before disease maturity. Here, we review the current literature supporting the importance of synapses in AD, our current understanding of the molecular interactions leading up to clinical diagnoses, and the techniques used in targeted therapies.
Collapse
Affiliation(s)
- Yixuan Amy Pei
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Julie Davies
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK
| | - Melanie Zhang
- Department of Neurobiology, Northwestern University Feinberg School of Medicine, Evanston, IL, USA
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, USA
| |
Collapse
|
57
|
Jangampalli Adi P, Reddy PH. Phosphorylated tau targeted small-molecule PROTACs for the treatment of Alzheimer's disease and tauopathies. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166162. [PMID: 33940164 DOI: 10.1016/j.bbadis.2021.166162] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Tau is a microtubule-stabilizing protein that plays an important role in the formation of axonal microtubules in neurons. Phosphorylated tau (p-Tau) has received great attention in the field of Alzheimer's disease (AD) as a potential therapeutic target due to its involvement with synaptic damage and neuronal dysfunction. Mounting evidence suggests that amyloid beta (Aβ)-targeted clinical trials continuously failed; therefore, it is important to consider alternative therapeutic strategies such as p-tau-PROTACs targeted small molecules for AD and other tauopathies. The present article describes the characteristics of tau biology, structure, and function in both healthy and pathological states in AD. It also explains data from studies that have identified the involvement of p-tau in neuronal damage and synaptic and cognitive functions in AD. Current article also covers several aspects, including small molecule inhibitors, and the development of p-tau-PROTACs targeted drug molecules to treat patients with AD and other tauopathies.
Collapse
Affiliation(s)
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
58
|
Ikram M, Jo MG, Park TJ, Kim MW, Khan I, Jo MH, Kim MO. Oral Administration of Gintonin Protects the Brains of Mice against A β-Induced Alzheimer Disease Pathology: Antioxidant and Anti-Inflammatory Effects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6635552. [PMID: 33953832 PMCID: PMC8068536 DOI: 10.1155/2021/6635552] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/10/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022]
Abstract
The study was aimed at analyzing the protective effects of gintonin in an amyloid beta- (Aβ-) induced Alzheimer's disease (AD) mouse model. For the development of the Aβ-induced AD mouse model, the amyloid-β (Aβ 1-42) peptide was stereotaxically injected into the brains of mice. Subsequently, gintonin was administered at a dose of 100 mg/kg/day/per oral (p.o) for four weeks daily, and its effects were evaluated by using western blotting, fluorescence analysis of brain sections, biochemical tests, and memory-related behavioral evaluations. To elucidate the effects of gintonin at the mechanistic level, the activation of endogenous antioxidant mechanisms, as well as the activation of astrocytes, microglia, and proinflammatory mediators such as nuclear factor erythroid 2-related factor 2 (NRF-2) and heme oxygenase-1 (HO-1), was evaluated. In addition, microglial cells (BV-2 cells) were used to analyze the effects of gintonin on microglial activation and signaling mechanisms. Collectively, the results suggested that gintonin reduced elevated oxidative stress by improving the expression of NRF-2 and HO-1 and thereby reducing the generation of reactive oxygen species (ROS) and lipid peroxidation (LPO). Moreover, gintonin significantly suppressed activated microglial cells and inflammatory mediators in the brains of Aβ-injected mice. Our findings also indicated improved synaptic and memory functions in the brains of Aβ-injected mice after treatment with gintonin. These results suggest that gintonin may be effective for relieving AD symptoms by regulating oxidative stress and inflammatory processes in a mouse model of AD. Collectively, the findings of this preclinical study highlight and endorse the potential, multitargeted protective effects of gintonin against AD-associated oxidative damage, neuroinflammation, cognitive impairment, and neurodegeneration.
Collapse
Affiliation(s)
- Muhammad Ikram
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Min Gi Jo
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, MVLS, University of Glasgow, UK
| | - Min Woo Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Ibrahim Khan
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Myeung Hoon Jo
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
59
|
Brabec JL, Lara MK, Tyler AL, Mahoney JM. System-Level Analysis of Alzheimer's Disease Prioritizes Candidate Genes for Neurodegeneration. Front Genet 2021; 12:625246. [PMID: 33889174 PMCID: PMC8056044 DOI: 10.3389/fgene.2021.625246] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disorder. Since the advent of the genome-wide association study (GWAS) we have come to understand much about the genes involved in AD heritability and pathophysiology. Large case-control meta-GWAS studies have increased our ability to prioritize weaker effect alleles, while the recent development of network-based functional prediction has provided a mechanism by which we can use machine learning to reprioritize GWAS hits in the functional context of relevant brain tissues like the hippocampus and amygdala. In parallel with these developments, groups like the Alzheimer’s Disease Neuroimaging Initiative (ADNI) have compiled rich compendia of AD patient data including genotype and biomarker information, including derived volume measures for relevant structures like the hippocampus and the amygdala. In this study we wanted to identify genes involved in AD-related atrophy of these two structures, which are often critically impaired over the course of the disease. To do this we developed a combined score prioritization method which uses the cumulative distribution function of a gene’s functional and positional score, to prioritize top genes that not only segregate with disease status, but also with hippocampal and amygdalar atrophy. Our method identified a mix of genes that had previously been identified in AD GWAS including APOE, TOMM40, and NECTIN2(PVRL2) and several others that have not been identified in AD genetic studies, but play integral roles in AD-effected functional pathways including IQSEC1, PFN1, and PAK2. Our findings support the viability of our novel combined score as a method for prioritizing region- and even cell-specific AD risk genes.
Collapse
Affiliation(s)
- Jeffrey L Brabec
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States
| | - Montana Kay Lara
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States
| | - Anna L Tyler
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - J Matthew Mahoney
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States.,The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
60
|
Nagy D, Ennis KA, Wei R, Su SC, Hinckley CA, Gu RF, Gao B, Massol RH, Ehrenfels C, Jandreski L, Thomas AM, Nelson A, Gyoneva S, Hajós M, Burkly LC. Developmental synaptic regulator, TWEAK/Fn14 signaling, is a determinant of synaptic function in models of stroke and neurodegeneration. Proc Natl Acad Sci U S A 2021; 118:e2001679118. [PMID: 33526652 PMCID: PMC8017933 DOI: 10.1073/pnas.2001679118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Identifying molecular mediators of neural circuit development and/or function that contribute to circuit dysfunction when aberrantly reengaged in neurological disorders is of high importance. The role of the TWEAK/Fn14 pathway, which was recently reported to be a microglial/neuronal axis mediating synaptic refinement in experience-dependent visual development, has not been explored in synaptic function within the mature central nervous system. By combining electrophysiological and phosphoproteomic approaches, we show that TWEAK acutely dampens basal synaptic transmission and plasticity through neuronal Fn14 and impacts the phosphorylation state of pre- and postsynaptic proteins in adult mouse hippocampal slices. Importantly, this is relevant in two models featuring synaptic deficits. Blocking TWEAK/Fn14 signaling augments synaptic function in hippocampal slices from amyloid-beta-overexpressing mice. After stroke, genetic or pharmacological inhibition of TWEAK/Fn14 signaling augments basal synaptic transmission and normalizes plasticity. Our data support a glial/neuronal axis that critically modifies synaptic physiology and pathophysiology in different contexts in the mature brain and may be a therapeutic target for improving neurophysiological outcomes.
Collapse
Affiliation(s)
- Dávid Nagy
- Clinical Sciences, Biogen, Cambridge, MA 02142
- Biogen Postdoctoral Scientist Program, Cellular Physiology, Biogen, Cambridge, MA 02142
| | - Katelin A Ennis
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Ru Wei
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Susan C Su
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | | | - Rong-Fang Gu
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Benbo Gao
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Ramiro H Massol
- Translational Cellular Sciences, Biogen, Cambridge, MA 02142
| | - Chris Ehrenfels
- Translational Cellular Sciences, Biogen, Cambridge, MA 02142
| | | | - Ankur M Thomas
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Ashley Nelson
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Stefka Gyoneva
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Mihály Hajós
- Clinical Sciences, Biogen, Cambridge, MA 02142
- Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Linda C Burkly
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142;
| |
Collapse
|
61
|
Evolution of the Human Diet and Its Impact on Gut Microbiota, Immune Responses, and Brain Health. Nutrients 2021; 13:nu13010196. [PMID: 33435203 PMCID: PMC7826636 DOI: 10.3390/nu13010196] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
The relatively rapid shift from consuming preagricultural wild foods for thousands of years, to consuming postindustrial semi-processed and ultra-processed foods endemic of the Western world less than 200 years ago did not allow for evolutionary adaptation of the commensal microbial species that inhabit the human gastrointestinal (GI) tract, and this has significantly impacted gut health. The human gut microbiota, the diverse and dynamic population of microbes, has been demonstrated to have extensive and important interactions with the digestive, immune, and nervous systems. Western diet-induced dysbiosis of the gut microbiota has been shown to negatively impact human digestive physiology, to have pathogenic effects on the immune system, and, in turn, cause exaggerated neuroinflammation. Given the tremendous amount of evidence linking neuroinflammation with neural dysfunction, it is no surprise that the Western diet has been implicated in the development of many diseases and disorders of the brain, including memory impairments, neurodegenerative disorders, and depression. In this review, we discuss each of these concepts to understand how what we eat can lead to cognitive and psychiatric diseases.
Collapse
|
62
|
Maldonado-Moreles A, Cordova-Fraga T, Bonilla-Jaime H, Lopez-Camacho PY, Basurto-Islas G. Low frequency vortex magnetic field reduces amyloid β aggregation, increase cell viability and protect from amyloid β toxicity. Electromagn Biol Med 2021; 40:191-200. [PMID: 33043710 DOI: 10.1080/15368378.2020.1830288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/18/2020] [Indexed: 01/05/2023]
Abstract
Plaques formed by abnormal accumulation of amyloid β-peptide (Aβ) lead to onset of Alzheimer's disease (AD). Pharmacological treatments do not reduce Aβ aggregation neither restore learning and memory. Noninvasive techniques have emerged as an alternative to treat AD, such as stimulation with electromagnetic fields (EMF) that decrease Aβ deposition and reverses cognitive impairment in AD mice, even though some studies showed side effects on parallel magnetic fields stimulation. As a new approach of magnetic field (MF) stimulation, vortex magnetic fields (VMF) have been tested inducing a random movement of charged biomolecules in cells, promoting cell viability and apparently safer than parallel magnetic fields. In this study we demonstrate the effect of VMF on Aβ aggregation. The experimental strategy includes, i) design and construction of a coil capable to induce VMF, ii) evaluation of VMF stimulation on Aβ peptide induced-fibrils-formation, iii) evaluation of VMF stimulation on SH-SY5Y neuroblastoma cell line in the presence of Aβ peptide. We demonstrated for the first time that Aβ aggregation exposed to VMF during 24 h decreased ~ 86% of Aβ fibril formation compared to control. Likewise, VMF stimulation reduced Aβ fibrils-cytotoxicity and increase SH-SY5Y cell viability. These data establish the basis for future investigation that involve VMF as inhibitor of Aβ-pathology and indicate the therapeutic potential of VMF for AD treatment.
Collapse
Affiliation(s)
- Alejandro Maldonado-Moreles
- Doctorado en Ciencias Biologicas y de la Salud, Universidad Autonoma Metropolitana , Ciudad de México, México
| | | | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Lab de Psicobiología, Universidad Autónoma Metropolitana Iztapalapa , Ciudad de México, México
| | - Perla Y Lopez-Camacho
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana Cuajimalpa , Ciudad de México, México
| | | |
Collapse
|
63
|
Engin AB, Engin A. Alzheimer's Disease and Protein Kinases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:285-321. [PMID: 33539020 DOI: 10.1007/978-3-030-49844-3_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and accounts for more than 60-80% of all cases of dementia. Loss of pyramidal neurons, extracellular amyloid beta (Abeta) accumulated senile plaques, and neurofibrillary tangles that contain hyperphosphorylated tau constitute the main pathological alterations in AD.Synaptic dysfunction and extrasynaptic N-methyl-D-aspartate receptor (NMDAR) hyperactivation contributes to excitotoxicity in patients with AD. Amyloid precursor protein (APP) and Abeta promoted neurodegeneration develop through the activation of protein kinase signaling cascade in AD. Furthermore, ultimate neuronal death in AD is under control of protein kinases-related signaling pathways. In this chapter, critical check-points within the cross-talk between neuron and protein kinases have been defined regarding the initiation and progression of AD. In this context, amyloid cascade hypothesis, neuroinflammation, oxidative stress, granulovacuolar degeneration, loss of Wnt signaling, Abeta-related synaptic alterations, prolonged calcium ions overload and NMDAR-related synaptotoxicity, damage signals hypothesis and type-3 diabetes are discussed briefly.In addition to clinical perspective of AD pathology, recommendations that might be effective in the treatment of AD patients have been reviewed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
64
|
John A, Reddy PH. Synaptic basis of Alzheimer's disease: Focus on synaptic amyloid beta, P-tau and mitochondria. Ageing Res Rev 2021; 65:101208. [PMID: 33157321 PMCID: PMC7770124 DOI: 10.1016/j.arr.2020.101208] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is a progressive and synaptic failure disease. Despite the many years of research, AD still harbors many secrets. As more of the world's population grows older, researchers are striving to find greater information on disease progression and pathogenesis. Identifying and treating the markers of this disease, or better yet, preventing it all together, are the hopes of those investing in this field of study. Several years of research revealed that synaptic pathology and mitochondrial oxidative damage are early events in disease progression. Loss of synapses and synaptic damage are the best correlates of cognitive deficits found in AD patients. As the disease progresses, there are significant changes at the synapse. These changes can both shed greater light onto the progression of the disease and serve as markers and therapeutic targets. This article addresses the mechanisms of synaptic action, mitochondrial regulation/dysregulation, resulting synaptic changes caused by amyloid beta and phosphorylated tau in AD progression. This article also highlights recent developments of risk factors, genetics and ApoE4 involvement, factors related to synaptic damage and loss, mislocalization of amyloid beta and phosphorylated tau, mitophagy, microglial activation and synapse-based therapies in AD. Furthermore, impairments in LTD and reactivation of microglia are discussed.
Collapse
Affiliation(s)
- Albin John
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
65
|
Subramanian J, Savage JC, Tremblay MÈ. Synaptic Loss in Alzheimer's Disease: Mechanistic Insights Provided by Two-Photon in vivo Imaging of Transgenic Mouse Models. Front Cell Neurosci 2020; 14:592607. [PMID: 33408613 PMCID: PMC7780885 DOI: 10.3389/fncel.2020.592607] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/25/2020] [Indexed: 01/05/2023] Open
Abstract
Synapse loss is the strongest correlate for cognitive decline in Alzheimer's disease. The mechanisms underlying synapse loss have been extensively investigated using mouse models expressing genes with human familial Alzheimer's disease mutations. In this review, we summarize how multiphoton in vivo imaging has improved our understanding of synapse loss mechanisms associated with excessive amyloid in the living animal brain. We also discuss evidence obtained from these imaging studies for the role of cell-intrinsic calcium dyshomeostasis and cell-extrinsic activities of microglia, which are the immune cells of the brain, in mediating synapse loss.
Collapse
Affiliation(s)
- Jaichandar Subramanian
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, United States
| | - Julie C Savage
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
66
|
Yang X, Du W, Zhang Y, Wang H, He M. Neuroprotective Effects of Higenamine Against the Alzheimer's Disease Via Amelioration of Cognitive Impairment, A β Burden, Apoptosis and Regulation of Akt/GSK3β Signaling Pathway. Dose Response 2020; 18:1559325820972205. [PMID: 33354171 PMCID: PMC7734528 DOI: 10.1177/1559325820972205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 01/01/2023] Open
Abstract
The present investigation was envisaged to elucidate the neuroprotective effect of Higenamine (HGN) against aluminum chloride (AlCl3) triggered experimental Alzheimer's disease (AD) rat model. Thirty-six male albino Wister rats were randomized and divided in 6 groups and subjected to experimentation for 6 weeks. Control group, AlCl3 (100 mg/kg orally), HGN (50 mg/kg orally), HGN25, HGN50, HGN75 (HGN 25, 50 and 75 mg/kg respectively and AlCl3 100 mg/kg orally). After completion of 42 days protocol, the animals were subjected to passive avoidance test. The animals were then anesthetized by intramuscularly injecting ketamine hydrochloride (24 mg/kg body weight) and euthanized by cervical amputation. Cortical and hippocampal tissues were carefully removed and were employed for quantification of aluminum and acetylcholinesterase. The tissues were quantified using Western blotting and detection kits for APP, Aβ1-42, β and γ secretases, Bax, Bad, caspases-9, cyto-c, pAkt and pGSK-3β, and oxidative markers. HGN significantly protected AlCl3 induced memory and learning impairments, Al overload, AChE hyperactivity, amyloid β (Aβ) burden and apoptosis in brain tissues via activating Akt/GSK3β pathway. HGN attenuated oxidative damage induced by Al by modulation of oxidative markers. Our findings advocate the neuroprotective effect of HGN in AlCl3 induced AD rat model.
Collapse
Affiliation(s)
- Xiaona Yang
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wanliang Du
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yun Zhang
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hui Wang
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Maolin He
- Department of Neurology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
67
|
Yang T, Tran KC, Zeng AY, Massa SM, Longo FM. Small molecule modulation of the p75 neurotrophin receptor inhibits multiple amyloid beta-induced tau pathologies. Sci Rep 2020; 10:20322. [PMID: 33230162 PMCID: PMC7683564 DOI: 10.1038/s41598-020-77210-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
Longitudinal preclinical and clinical studies suggest that Aβ drives neurite and synapse degeneration through an array of tau-dependent and independent mechanisms. The intracellular signaling networks regulated by the p75 neurotrophin receptor (p75NTR) substantially overlap with those linked to Aβ and to tau. Here we examine the hypothesis that modulation of p75NTR will suppress the generation of multiple potentially pathogenic tau species and related signaling to protect dendritic spines and processes from Aβ-induced injury. In neurons exposed to oligomeric Aβ in vitro and APP mutant mouse models, modulation of p75NTR signaling using the small-molecule LM11A-31 was found to inhibit Aβ-associated degeneration of neurites and spines; and tau phosphorylation, cleavage, oligomerization and missorting. In line with these effects on tau, LM11A-31 inhibited excess activation of Fyn kinase and its targets, tau and NMDA-NR2B, and decreased Rho kinase signaling changes and downstream aberrant cofilin phosphorylation. In vitro studies with pseudohyperphosphorylated tau and constitutively active RhoA revealed that LM11A-31 likely acts principally upstream of tau phosphorylation, and has effects preventing spine loss both up and downstream of RhoA activation. These findings support the hypothesis that modulation of p75NTR signaling inhibits a broad spectrum of Aβ-triggered, tau-related molecular pathology thereby contributing to synaptic resilience.
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA
| | - Kevin C Tran
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA
| | - Anne Y Zeng
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA
| | - Stephen M Massa
- Department of Neurology, San Francisco Veterans Affairs Health Care System, University of California, San Francisco, 4150 Clement St., San Francisco, CA, 94121, USA.
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA.
| |
Collapse
|
68
|
Shaposhnikov MV, Zemskaya NV, Koval LА, Minnikhanova NR, Kechko OI, Mitkevich VA, Makarov AA, Moskalev AА. Amyloid-β peptides slightly affect lifespan or antimicrobial peptide gene expression in Drosophila melanogaster. BMC Genet 2020; 21:65. [PMID: 33092519 PMCID: PMC7583308 DOI: 10.1186/s12863-020-00866-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 11/23/2022] Open
Abstract
Background Beta-amyloid peptide (Aβ) is the key protein in the pathogenesis of Alzheimer’s disease, the most common age-related neurodegenerative disorder in humans. Aβ peptide induced pathological phenotypes in different model organisms include neurodegeneration and lifespan decrease. However, recent experimental evidence suggests that Aβ may utilize oligomerization and fibrillization to function as an antimicrobial peptide (AMP), and protect the host from infections. We used the power of Drosophila model to study mechanisms underlying a dual role for Aβ peptides. Results We investigated the effects of Drosophila treatment with three Aβ42 peptide isoforms, which differ in their ability to form oligomers and aggregates on the lifespan, locomotor activity and AMP genes expression. Aβ42 slightly decreased female’s median lifespan (by 4.5%), but the effect was not related to the toxicity of peptide isoform. The lifespan and relative levels of AMP gene expression in male flies as well as locomotor activity in both sexes were largely unaffected by Aβ42 peptide treatment. Regardless of the effects on lifespan, Aβ42 peptide treatment induced decrease in AMP genes expression in females, but the effects were not robust. Conclusions The results demonstrate that chronic treatment with Aβ42 peptides does not drastically affect fly aging or immunity.
Collapse
Affiliation(s)
- Mikhail V Shaposhnikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.,Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Nadezhda V Zemskaya
- Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Lyubov А Koval
- Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Natalya R Minnikhanova
- Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia
| | - Olga I Kechko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Alexey А Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia. .,Institute of Biology of Komi Science Center of Ural Branch of RAS, 167982, Syktyvkar, Russia.
| |
Collapse
|
69
|
Tan X, Liang Z, Li Y, Zhi Y, Yi L, Bai S, Forest KH, Nichols RA, Dong Y, Li QX. Isoorientin, a GSK-3β inhibitor, rescues synaptic dysfunction, spatial memory deficits and attenuates pathological progression in APP/PS1 model mice. Behav Brain Res 2020; 398:112968. [PMID: 33069740 DOI: 10.1016/j.bbr.2020.112968] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/13/2020] [Accepted: 10/10/2020] [Indexed: 02/07/2023]
Abstract
β-Amyloid (Aβ) elevation, tau hyperphosphorylation, and neuroinflammation are major hallmarks of Alzheimer's disease (AD). Glycogen synthase kinase-3β (GSK-3β) is a key protein kinase implicated in the pathogenesis of AD. Blockade of GSK-3β is an attractive therapeutic strategy for AD. Isoorientin, a 6-C-glycosylflavone, was previously shown to be a highly selective inhibitor of GSK-3β, while exerting neuroprotective effects in neuronal models of AD. In the present study, we evaluated the in vivo effects of isoorientin on GSK-3β, tau phosphorylation, Aβ deposition, neuroinflammatory response, long-term potentiation, and spatial memory in amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice using biochemical, electrophysiological, and behavioral tests. Chronic oral administration of isoorientin to APP/PS1 mice at 8 months of age attenuated multiple AD pathogenic hallmarks in the brains, including GSK-3β overactivation, tau hyperphosphorylation, Aβ deposition, and neuroinflammation. For neuroinflammation, isoorientin treatment reduced the number of activated microglia associated with Aβ-positive plaques, and in parallel reduced the levels of pro-inflammatory factors in the brains of APP/PS1 mice. Strikingly, isoorientin reversed deficits in synaptic long-term potentiation and spatial memory relevant to cognitive functions. Together, the findings suggest that isoorientin is a brain neuroprotector and may be a promising drug lead for treatment of AD and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Xiaoqin Tan
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, 1955 East-West Road, Honolulu, HI 96822, United States
| | - Zhibin Liang
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, 1955 East-West Road, Honolulu, HI 96822, United States
| | - Yingui Li
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yingkun Zhi
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lang Yi
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shasha Bai
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Kelly H Forest
- Department of Cell and Molecular Biology, John A. Burn School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI 96813, United States
| | - Robert A Nichols
- Department of Cell and Molecular Biology, John A. Burn School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street, Honolulu, HI 96813, United States
| | - Yan Dong
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, 1955 East-West Road, Honolulu, HI 96822, United States.
| |
Collapse
|
70
|
Sadeghmousavi S, Eskian M, Rahmani F, Rezaei N. The effect of insomnia on development of Alzheimer's disease. J Neuroinflammation 2020; 17:289. [PMID: 33023629 PMCID: PMC7542374 DOI: 10.1186/s12974-020-01960-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and a neurodegenerative disorder characterized by memory deficits especially forgetting recent information, recall ability impairment, and loss of time tracking, problem-solving, language, and recognition difficulties. AD is also a globally important health issue but despite all scientific efforts, the treatment of AD is still a challenge. Sleep has important roles in learning and memory consolidation. Studies have shown that sleep deprivation (SD) and insomnia are associated with the pathogenesis of Alzheimer's disease and may have an impact on the symptoms and development. Thus, sleep disorders have decisive effects on AD; this association deserves more attention in research, diagnostics, and treatment, and knowing this relation also can help to prevent AD through screening and proper management of sleep disorders. This study aimed to show the potential role of SD and insomnia in the pathogenesis and progression of AD.
Collapse
Affiliation(s)
- Shaghayegh Sadeghmousavi
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Eskian
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Rahmani
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nima Rezaei
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
71
|
Huang C, Wen C, Yang M, Li A, Fan C, Gan D, Li Q, Zhao J, Zhu L, Lu D. Astaxanthin Improved the Cognitive Deficits in APP/PS1 Transgenic Mice Via Selective Activation of mTOR. J Neuroimmune Pharmacol 2020; 16:609-619. [PMID: 32944864 DOI: 10.1007/s11481-020-09953-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/26/2020] [Indexed: 01/20/2023]
Abstract
Astaxanthin (Ast) is an effective neuroprotective and antioxidant compound used to treat Alzheimer's disease (AD); however, the underlying in vivo molecular mechanisms remain unknown. In this study, we report that Ast can activate the mammalian target of rapamycin (mTOR) pathway in the 8-month-old APP/PS1 transgenic mouse model of AD. Our results suggest that Ast could ameliorate the cognitive defects in APP/PS1 mice by activating the mTOR pathway. Moreover, mTOR activation perturbed the mitochondrial dynamics, increased the synaptic plasticity after 21 days of treatment with Ast (10 mg/kg/day), and increased the expression of Aβ-degrading enzymes, mitochondrial fusion, and synapse-associated proteins and decreased the expression of mitochondrial fission proteins. Intraperitoneal injection of the mTOR inhibitor, rapamycin, abolished the effects of Ast. In conclusion, Ast activates the mTOR pathway, which is necessary for mitochondrial dynamics and synaptic plasticity, leading to improved learning and memory. Our results support the use of Ast for the treatment of cognitive deficits. Graphical abstract In summary, Ast ameliorates cognitive deficits via facilitating the mTOR-dependent mitochondrial dynamics and synaptic damage, and reducing Aβ accumulation. This model supports the use of Ast for the treatment of cognitive deficits.
Collapse
Affiliation(s)
- Cuiqin Huang
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Caiyan Wen
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Mei Yang
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - An Li
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Chongzhu Fan
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Danhui Gan
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Qin Li
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Jiayi Zhao
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Lihong Zhu
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Daxiang Lu
- Department of Pathophysiology, Institute of Brain Science Research, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, 510632, Guangzhou, Guangdong, China.
| |
Collapse
|
72
|
Koller EJ, Chakrabarty P. Tau-Mediated Dysregulation of Neuroplasticity and Glial Plasticity. Front Mol Neurosci 2020; 13:151. [PMID: 32973446 PMCID: PMC7472665 DOI: 10.3389/fnmol.2020.00151] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/20/2020] [Indexed: 01/14/2023] Open
Abstract
The inability of individual neurons to compensate for aging-related damage leads to a gradual loss of functional plasticity in the brain accompanied by progressive impairment in learning and memory. Whereas this loss in neuroplasticity is gradual during normal aging, in neurodegenerative diseases such as Alzheimer’s disease (AD), this loss is accelerated dramatically, leading to the incapacitation of patients within a decade of onset of cognitive symptoms. The mechanisms that underlie this accelerated loss of neuroplasticity in AD are still not completely understood. While the progressively increasing proteinopathy burden, such as amyloid β (Aβ) plaques and tau tangles, definitely contribute directly to a neuron’s functional demise, the role of non-neuronal cells in controlling neuroplasticity is slowly being recognized as another major factor. These non-neuronal cells include astrocytes, microglia, and oligodendrocytes, which through regulating brain homeostasis, structural stability, and trophic support, play a key role in maintaining normal functioning and resilience of the neuronal network. It is believed that chronic signaling from these cells affects the homeostatic network of neuronal and non-neuronal cells to an extent to destabilize this harmonious milieu in neurodegenerative diseases like AD. Here, we will examine the experimental evidence regarding the direct and indirect pathways through which astrocytes and microglia can alter brain plasticity in AD, specifically as they relate to the development and progression of tauopathy. In this review article, we describe the concepts of neuroplasticity and glial plasticity in healthy aging, delineate possible mechanisms underlying tau-induced plasticity dysfunction, and discuss current clinical trials as well as future disease-modifying approaches.
Collapse
Affiliation(s)
- Emily J Koller
- Department of Neuroscience, University of Florida, Gainesville, FL, United States.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL, United States.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States.,McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
73
|
Najm R, Zalocusky KA, Zilberter M, Yoon SY, Hao Y, Koutsodendris N, Nelson M, Rao A, Taubes A, Jones EA, Huang Y. In Vivo Chimeric Alzheimer's Disease Modeling of Apolipoprotein E4 Toxicity in Human Neurons. Cell Rep 2020; 32:107962. [PMID: 32726626 PMCID: PMC7430173 DOI: 10.1016/j.celrep.2020.107962] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/15/2020] [Accepted: 07/03/2020] [Indexed: 02/08/2023] Open
Abstract
Despite its clear impact on Alzheimer's disease (AD) risk, apolipoprotein (apo) E4's contributions to AD etiology remain poorly understood. Progress in answering this and other questions in AD research has been limited by an inability to model human-specific phenotypes in an in vivo environment. Here we transplant human induced pluripotent stem cell (hiPSC)-derived neurons carrying normal apoE3 or pathogenic apoE4 into human apoE3 or apoE4 knockin mouse hippocampi, enabling us to disentangle the effects of apoE4 produced in human neurons and in the brain environment. Using single-nucleus RNA sequencing (snRNA-seq), we identify key transcriptional changes specific to human neuron subtypes in response to endogenous or exogenous apoE4. We also find that Aβ from transplanted human neurons forms plaque-like aggregates, with differences in localization and interaction with microglia depending on the transplant and host apoE genotype. These findings highlight the power of in vivo chimeric disease modeling for studying AD.
Collapse
Affiliation(s)
- Ramsey Najm
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelly A Zalocusky
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Gladstone Center for Translational Advancement, San Francisco, CA 94158, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Gladstone Center for Translational Advancement, San Francisco, CA 94158, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maxine Nelson
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antara Rao
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alice Taubes
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Emily A Jones
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone Center for Translational Advancement, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Departments of Neurology and Pathology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
74
|
Advantages and Pitfalls in Fluid Biomarkers for Diagnosis of Alzheimer's Disease. J Pers Med 2020; 10:jpm10030063. [PMID: 32708853 PMCID: PMC7563364 DOI: 10.3390/jpm10030063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023] Open
Abstract
Alzheimer’s disease (AD) is a commonly occurring neurodegenerative disease in the advanced-age population, with a doubling of prevalence for each 5 years of age above 60 years. In the past two decades, there has been a sustained effort to find suitable biomarkers that may not only aide with the diagnosis of AD early in the disease process but also predict the onset of the disease in asymptomatic individuals. Current diagnostic evidence is supportive of some biomarker candidates isolated from cerebrospinal fluid (CSF), including amyloid beta peptide (Aβ), total tau (t-tau), and phosphorylated tau (p-tau) as being involved in the pathophysiology of AD. However, there are a few biomarkers that have been shown to be helpful, such as proteomic, inflammatory, oral, ocular and olfactory in the early detection of AD, especially in the individuals with mild cognitive impairment (MCI). To date, biomarkers are collected through invasive techniques, especially CSF from lumbar puncture; however, non-invasive (radio imaging) methods are used in practice to diagnose AD. In order to reduce invasive testing on the patients, present literature has highlighted the potential importance of biomarkers in blood to assist with diagnosing AD.
Collapse
|
75
|
Yeast-Based Aβ1-15 Vaccine Elicits Strong Immunogenicity and Attenuates Neuropathology and Cognitive Deficits in Alzheimer's Disease Transgenic Mice. Vaccines (Basel) 2020; 8:vaccines8030351. [PMID: 32630299 PMCID: PMC7563250 DOI: 10.3390/vaccines8030351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy focusing on reducing the amyloid-beta (Aβ) burden is a promising treatment strategy for Alzheimer's disease (AD). Many clinical studies on AD immunotherapies have failed due to low safety and efficacy, calling for a highly potent AD vaccine which induces sufficient antibody titer while avoiding side effects. Here, we designed a yeast-based vaccine Y-5A15 comprising five copies of Aβ1-15 displayed on the surface of yeast cell wall, and we subcutaneously immunized APP/PS1 mice three times. Our results demonstrated that the Y-5A15 remarkably enhanced the Aβ epitope immunogenicity and elicited high antibody titers against Aβ in AD mice. Importantly, Y-5A15 vaccination successfully reduced Aβ levels, plaque burden and glial activation, rescued synaptic deficits and significantly ameliorated memory and cognitive decline in APP/PS1 transgenic mice, suggesting that the yeast-based Aβ epitope vaccine has a promising potency for the treatment of AD.
Collapse
|
76
|
Caccavano A, Bozzelli PL, Forcelli PA, Pak DTS, Wu JY, Conant K, Vicini S. Inhibitory Parvalbumin Basket Cell Activity is Selectively Reduced during Hippocampal Sharp Wave Ripples in a Mouse Model of Familial Alzheimer's Disease. J Neurosci 2020; 40:5116-5136. [PMID: 32439703 PMCID: PMC7314414 DOI: 10.1523/jneurosci.0425-20.2020] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/07/2020] [Accepted: 05/18/2020] [Indexed: 12/29/2022] Open
Abstract
Memory disruption in mild cognitive impairment (MCI) and Alzheimer's disease (AD) is poorly understood, particularly at early stages preceding neurodegeneration. In mouse models of AD, there are disruptions to sharp wave ripples (SWRs), hippocampal population events with a critical role in memory consolidation. However, the microcircuitry underlying these disruptions is under-explored. We tested whether a selective reduction in parvalbumin-expressing (PV) inhibitory interneuron activity underlies hyperactivity and SWR disruption. We employed the 5xFAD model of familial AD crossed with mouse lines labeling excitatory pyramidal cells (PCs) and inhibitory PV cells. We observed a 33% increase in frequency, 58% increase in amplitude, and 8% decrease in duration of SWRs in ex vivo slices from male and female three-month 5xFAD mice versus littermate controls. 5xFAD mice of the same age were impaired in a hippocampal-dependent memory task. Concurrent with SWR recordings, we performed calcium imaging, cell-attached, and whole-cell recordings of PC and PV cells within the CA1 region. PCs in 5xFAD mice participated in enlarged ensembles, with superficial PCs (sPCs) having a higher probability of spiking during SWRs. Both deep PCs (dPCs) and sPCs displayed an increased synaptic E/I ratio, suggesting a disinhibitory mechanism. In contrast, we observed a 46% spike rate reduction during SWRs in PV basket cells (PVBCs), while PV bistratified and axo-axonic cells were unimpaired. Excitatory synaptic drive to PVBCs was selectively reduced by 50%, resulting in decreased E/I ratio. Considering prior studies of intrinsic PV cell dysfunction in AD, these findings suggest alterations to the PC-PVBC microcircuit also contribute to impairment.SIGNIFICANCE STATEMENT We demonstrate that a specific subtype of inhibitory neuron, parvalbumin-expressing (PV) basket cells, have selectively reduced activity in a model of Alzheimer's disease (AD) during activity critical for the consolidation of memory. These results identify a potential cellular target for therapeutic intervention to restore aberrant network activity in early amyloid pathology. While PV cells have previously been identified as a potential therapeutic target, this study for the first time recognizes that other PV neuronal subtypes, including bistratified and axo-axonic cells, are spared. These experiments are the first to record synaptic and spiking activity during sharp wave ripple (SWR) events in early amyloid pathology and reveal that a selective decrease in excitatory synaptic drive to PV basket cells (PVBCs) likely underlies reduced function.
Collapse
Affiliation(s)
- Adam Caccavano
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - P Lorenzo Bozzelli
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Patrick A Forcelli
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057
| | - Daniel T S Pak
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057
| | - Jian-Young Wu
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Katherine Conant
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057
| |
Collapse
|
77
|
Albert-Gascó H, Ros-Bernal F, Castillo-Gómez E, Olucha-Bordonau FE. MAP/ERK Signaling in Developing Cognitive and Emotional Function and Its Effect on Pathological and Neurodegenerative Processes. Int J Mol Sci 2020; 21:E4471. [PMID: 32586047 PMCID: PMC7352860 DOI: 10.3390/ijms21124471] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
The signaling pathway of the microtubule-associated protein kinase or extracellular regulated kinase (MAPK/ERK) is a common mechanism of extracellular information transduction from extracellular stimuli to the intracellular space. The transduction of information leads to changes in the ongoing metabolic pathways and the modification of gene expression patterns. In the central nervous system, ERK is expressed ubiquitously, both temporally and spatially. As for the temporal ubiquity, this signaling system participates in three key moments: (i) Embryonic development; (ii) the early postnatal period; and iii) adulthood. During embryonic development, the system is partly responsible for the patterning of segmentation in the encephalic vesicle through the FGF8-ERK pathway. In addition, during this period, ERK directs neurogenesis migration and the final fate of neural progenitors. During the early postnatal period, ERK participates in the maturation process of dendritic trees and synaptogenesis. During adulthood, ERK participates in social and emotional behavior and memory processes, including long-term potentiation. Alterations in mechanisms related to ERK are associated with different pathological outcomes. Genetic alterations in any component of the ERK pathway result in pathologies associated with neural crest derivatives and mental dysfunctions associated with autism spectrum disorders. The MAP-ERK pathway is a key element of the neuroinflammatory pathway triggered by glial cells during the development of neurodegenerative diseases, such as Parkinson's and Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, as well as prionic diseases. The process triggered by MAPK/ERK activation depends on the stage of development (mature or senescence), the type of cellular element in which the pathway is activated, and the anatomic neural structure. However, extensive gaps exist with regards to the targets of the phosphorylated ERK in many of these processes.
Collapse
Affiliation(s)
- Héctor Albert-Gascó
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Hills Road, Cambridge CB2 0AH, UK;
| | - Francisco Ros-Bernal
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
| | - Esther Castillo-Gómez
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| | - Francisco E. Olucha-Bordonau
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| |
Collapse
|
78
|
Antia, a Natural Antioxidant Product, Attenuates Cognitive Dysfunction in Streptozotocin-Induced Mouse Model of Sporadic Alzheimer's Disease by Targeting the Amyloidogenic, Inflammatory, Autophagy, and Oxidative Stress Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4386562. [PMID: 32655767 PMCID: PMC7320293 DOI: 10.1155/2020/4386562] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/07/2020] [Accepted: 04/11/2020] [Indexed: 12/13/2022]
Abstract
Background Many neurodegenerative diseases such as Alzheimer's disease are associated with oxidative stress. Therefore, antioxidant therapy has been suggested for the prevention and treatment of neurodegenerative diseases. Objective We investigated the ability of the antioxidant Antia to exert a protective effect against sporadic Alzheimer's disease (SAD) induced in mice. Antia is a natural product that is extracted from the edible yamabushitake mushroom, the gotsukora and kothala himbutu plants, diosgenin (an extract from wild yam tubers), and amla (Indian gooseberry) after treatment with MRN-100. Methods Single intracerebroventricular (ICV) injection of streptozotocin (STZ) (3 mg/kg) was used for induction of SAD in mice. Antia was injected intraperitoneally (i.p.) in 3 doses (25, 50, and 100 mg/kg/day) for 21 days. Neurobehavioral tests were conducted within 24 h after the last day of injection. Afterwards, mice were sacrificed and their hippocampi were rapidly excised, weighed, and homogenized to be used for measuring biochemical parameters. Results Treatment with Antia significantly improved mice performance in the Morris water maze. In addition, biochemical analysis showed that Antia exerted a protective effect for several compounds, including GSH, MDA, NF-κB, IL-6, TNF-α, and amyloid β. Further studies with western blot showed the protective effect of Antia for the JAK2/STAT3 pathway. Conclusions Antia exerts a significant protection against cognitive dysfunction induced by ICV-STZ injection. This effect is achieved through targeting of the amyloidogenic, inflammatory, and oxidative stress pathways. The JAK2/STAT3 pathway plays a protective role for neuroinflammatory and neurodegenerative diseases such as SAD.
Collapse
|
79
|
Arora S, Sharma D, Singh J. GLUT-1: An Effective Target To Deliver Brain-Derived Neurotrophic Factor Gene Across the Blood Brain Barrier. ACS Chem Neurosci 2020; 11:1620-1633. [PMID: 32352752 DOI: 10.1021/acschemneuro.0c00076] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, inflicts enormous suffering to patients and their family members. It is the third deadliest disease, affecting 46.8 million people worldwide. Brain-derived neurotrophic factor (BDNF) is involved in the development, maintenance, and plasticity of the central nervous system. This crucial protein is significantly reduced in AD patients leading to reduced plasticity and neuronal death. In this study, we demonstrate the targeted delivery of the BDNF gene to the brain using liposome nanoparticles. These liposomes were surface modified with glucose transporter-1 targeting ligand (mannose) and cell penetrating peptides (penetratin or rabies virus glycoprotein) to promote selective and enhanced delivery to the brain. Surface modified liposomes showed significantly higher transfection of BDNF in primary astrocytes and neurons, compared to unmodified (plain) liposomes. BDNF transfection via dual modified liposomes resulted in an increase in presynaptic marker synaptophysin protein in primary neuronal cells, which is usually found to be reduced in AD patients. Liposomes surface modified with mannose and cell penetrating peptides demonstrated ∼50% higher transport across the in vitro blood brain barrier (BBB) model and showed significantly higher transfection efficiency in primary neuronal cells compared to plain liposomes. These results were correlated with significantly higher transport of surface modified liposomes (∼7% of injected dose/gram of tissue) and BDNF transfection (∼1.7 times higher than baseline level) across BBB following single intravenous administration in C57BL/6 mice without any signs of inflammation or toxicity. Overall, this study suggests a safe and targeted strategy to increase BDNF protein in the brain, which has the potential to reverse AD pathophysiology.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
80
|
Uddin MS, Rahman MA, Kabir MT, Behl T, Mathew B, Perveen A, Barreto GE, Bin-Jumah MN, Abdel-Daim MM, Ashraf GM. Multifarious roles of mTOR signaling in cognitive aging and cerebrovascular dysfunction of Alzheimer's disease. IUBMB Life 2020; 72:1843-1855. [PMID: 32472959 DOI: 10.1002/iub.2324] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/10/2020] [Accepted: 05/13/2020] [Indexed: 12/19/2022]
Abstract
Age-related cognitive failure is a main devastating incident affecting even healthy people. Alzheimer's disease (AD) is the utmost common form of dementia among the geriatric community. In the pathogenesis of AD, cerebrovascular dysfunction is revealed before the beginning of the cognitive decline. Mounting proof shows a precarious impact of cerebrovascular dysregulation in the development of AD pathology. Recent studies document that the mammalian target of rapamycin (mTOR) acts as a crucial effector of cerebrovascular dysregulation in AD. The mTOR contributes to brain vascular dysfunction and subsequence cerebral blood flow deficits as well as cognitive impairment. Furthermore, mTOR causes the blood-brain barrier (BBB) breakdown in AD models. Inhibition of mTOR hyperactivity protects the BBB integrity in AD. Furthermore, mTOR drives cognitive defect and cerebrovascular dysfunction, which are greatly prevalent in AD, but the central molecular mechanisms underlying these alterations are obscure. This review represents the crucial and current research findings regarding the role of mTOR signaling in cognitive aging and cerebrovascular dysfunction in the pathogenesis of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Md Ataur Rahman
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Patiala, India
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
81
|
Dufort-Gervais J, Provost C, Charbonneau L, Norris CM, Calon F, Mongrain V, Brouillette J. Neuroligin-1 is altered in the hippocampus of Alzheimer's disease patients and mouse models, and modulates the toxicity of amyloid-beta oligomers. Sci Rep 2020; 10:6956. [PMID: 32332783 PMCID: PMC7181681 DOI: 10.1038/s41598-020-63255-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
Synapse loss occurs early and correlates with cognitive decline in Alzheimer's disease (AD). Synaptotoxicity is driven, at least in part, by amyloid-beta oligomers (Aβo), but the exact synaptic components targeted by Aβo remain to be identified. We here tested the hypotheses that the post-synaptic protein Neuroligin-1 (NLGN1) is affected early in the process of neurodegeneration in the hippocampus, and specifically by Aβo, and that it can modulate Aβo toxicity. We found that hippocampal NLGN1 was decreased in patients with AD in comparison to patients with mild cognitive impairment and control subjects. Female 3xTg-AD mice also showed a decreased NLGN1 level in the hippocampus at an early age (i.e., 4 months). We observed that chronic hippocampal Aβo injections initially increased the expression of one specific Nlgn1 transcript, which was followed by a clear decrease. Lastly, the absence of NLGN1 decreased neuronal counts in the dentate gyrus, which was not the case in wild-type animals, and worsens impairment in spatial learning following chronic hippocampal Aβo injections. Our findings support that NLGN1 is impacted early during neurodegenerative processes, and that Aβo contributes to this effect. Moreover, our results suggest that the presence of NLGN1 favors the cognitive prognosis during Aβo-driven neurodegeneration.
Collapse
Affiliation(s)
- Julien Dufort-Gervais
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal (Recherche CIUSSS-NIM), Montréal, Québec, Canada
| | - Chloé Provost
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal (Recherche CIUSSS-NIM), Montréal, Québec, Canada
| | | | - Christopher M Norris
- Department of Molecular and Biomedical Pharmacology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Frédéric Calon
- Neuroscience Unit, Research Center - CHU de Québec, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - Valérie Mongrain
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal (Recherche CIUSSS-NIM), Montréal, Québec, Canada.
- Department of Neuroscience, Université de Montréal, Montréal, Québec, Canada.
| | - Jonathan Brouillette
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada.
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal (Recherche CIUSSS-NIM), Montréal, Québec, Canada.
| |
Collapse
|
82
|
Rodrigues BDS, Kanekiyo T, Singh J. Nerve Growth Factor Gene Delivery across the Blood–Brain Barrier to Reduce Beta Amyloid Accumulation in AD Mice. Mol Pharm 2020; 17:2054-2063. [DOI: 10.1021/acs.molpharmaceut.0c00218] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Bruna dos Santos Rodrigues
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
83
|
Li S, Selkoe DJ. A mechanistic hypothesis for the impairment of synaptic plasticity by soluble Aβ oligomers from Alzheimer's brain. J Neurochem 2020; 154:583-597. [PMID: 32180217 DOI: 10.1111/jnc.15007] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 12/18/2022]
Abstract
It is increasingly accepted that early cognitive impairment in Alzheimer's disease results in considerable part from synaptic dysfunction caused by the accumulation of a range of oligomeric assemblies of amyloid β-protein (Aβ). Most studies have used synthetic Aβ peptides to explore the mechanisms of memory deficits in rodent models, but recent work suggests that Aβ assemblies isolated from human (AD) brain tissue are far more potent and disease-relevant. Although reductionist experiments show Aβ oligomers to impair synaptic plasticity and neuronal viability, the responsible mechanisms are only partly understood. Glutamatergic receptors, GABAergic receptors, nicotinic receptors, insulin receptors, the cellular prion protein, inflammatory mediators, and diverse signaling pathways have all been suggested. Studies using AD brain-derived soluble Aβ oligomers suggest that only certain bioactive forms (principally small, diffusible oligomers) can disrupt synaptic plasticity, including by binding to plasma membranes and changing excitatory-inhibitory balance, perturbing mGluR, PrP, and other neuronal surface proteins, down-regulating glutamate transporters, causing glutamate spillover, and activating extrasynaptic GluN2B-containing NMDA receptors. We synthesize these emerging data into a mechanistic hypothesis for synaptic failure in Alzheimer's disease that can be modified as new knowledge is added and specific therapeutics are developed.
Collapse
Affiliation(s)
- Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
84
|
Kanyo R, Leighton PLA, Neil GJ, Locskai LF, Allison WT. Amyloid-β precursor protein mutant zebrafish exhibit seizure susceptibility that depends on prion protein. Exp Neurol 2020; 328:113283. [PMID: 32165257 DOI: 10.1016/j.expneurol.2020.113283] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 02/03/2020] [Accepted: 03/08/2020] [Indexed: 12/11/2022]
Abstract
It has been proposed that Amyloid β Precursor Protein (APP) might act as a rheostat controlling neuronal excitability, but mechanisms have remained untested. APP and its catabolite Aβ are known to impact upon synapse function and dysfunction via their interaction with the prion protein (PrPC), suggesting a candidate pathway. Here we test if PrPC is required for this APP function in vivo, perhaps via modulating mGluR5 ion channels. We engineered zebrafish to lack homologs of PrPC and APP, allowing us to assess their purported genetic and physiological interactions in CNS development. We generated four appa null alleles as well as prp1-/-;appa-/- double mutants (engineering of prp1 mutant alleles is described elsewhere). Unexpectedly, appa-/- and compound prp1-/-;appa-/- mutants are viable and lacked overt phenotypes (except being slightly smaller than wildtype fish at some developmental stages). Zebrafish prp1-/- mutants were substantially more sensitive to appa knockdown than wildtype fish, and both zebrafish prp1 and mammalian Prnp mRNA were significantly able to partially rescue this effect. Further, appa-/- mutants exhibited increased seizures upon exposure to low doses of convulsant. The mechanism of this seizure susceptibility requires prp1 insomuch that seizures were significantly dampened to wildtype levels in prp1-/-;appa-/- mutants. Inhibiting mGluR5 channels, which may be downstream of PrPC, increased seizure intensity only in prp1-/- mutants, and this seizure mechanism required intact appa. Taken together, these results support an intriguing genetic interaction between prp1 and appa with their shared roles impacting upon neuron hyperexcitability, thus complementing and extending past works detailing their biochemical interaction(s).
Collapse
Affiliation(s)
- Richard Kanyo
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Patricia L A Leighton
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Gavin J Neil
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Laszlo F Locskai
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - W Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada; Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
85
|
Jiang H, Esparza TJ, Kummer TT, Zhong H, Rettig J, Brody DL. Live Neuron High-Content Screening Reveals Synaptotoxic Activity in Alzheimer Mouse Model Homogenates. Sci Rep 2020; 10:3412. [PMID: 32098978 PMCID: PMC7042280 DOI: 10.1038/s41598-020-60118-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/05/2020] [Indexed: 12/28/2022] Open
Abstract
Accurate quantification of synaptic changes is essential for understanding the molecular mechanisms of synaptogenesis, synaptic plasticity, and synaptic toxicity. Here we demonstrate a robust high-content imaging method for the assessment of synaptic changes and apply the method to brain homogenates from an Alzheimer's disease mouse model. Our method uses serial imaging of endogenous fluorescent labeled presynaptic VAMP2 and postsynaptic PSD95 in long-term cultured live primary neurons in 96 well microplates, and uses automatic image analysis to quantify the number of colocalized mature synaptic puncta for the assessment of synaptic changes in live neurons. As a control, we demonstrated that our synaptic puncta assay is at least 10-fold more sensitive to the toxic effects of glutamate than the MTT assay. Using our assay, we have compared synaptotoxic activities in size-exclusion chromatography fractioned protein samples from 3xTg-AD mouse model brain homogenates. Multiple synaptotoxic activities were found in high and low molecular weight fractions. Amyloid-beta immunodepletion alleviated some but not all of the synaptotoxic activities. Although the biochemical entities responsible for the synaptotoxic activities have yet to be determined, these proof-of-concept results demonstrate that this novel assay may have many potential mechanistic and therapeutic applications.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
| | - Thomas J Esparza
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, 20817, USA
- National Institute of Neurological Disorders and Stroke, 10 Center Drive, Bethesda, Maryland, 20892, USA
| | - Terrance T Kummer
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, Oregon, 97239, USA
| | - Jens Rettig
- Department of Physiology, Saarland University, Center for Integrative Physiology and Molecular Medicine (CIPMM), Building 48, Homburg, 66421, Germany
| | - David L Brody
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA.
- National Institute of Neurological Disorders and Stroke, 10 Center Drive, Bethesda, Maryland, 20892, USA.
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 20814, USA.
| |
Collapse
|
86
|
Pan Y, Monje M. Activity Shapes Neural Circuit Form and Function: A Historical Perspective. J Neurosci 2020; 40:944-954. [PMID: 31996470 PMCID: PMC6988998 DOI: 10.1523/jneurosci.0740-19.2019] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/30/2022] Open
Abstract
The brilliant and often prescient hypotheses of Ramon y Cajal have proven foundational for modern neuroscience, but his statement that "In adult centers the nerve paths are something fixed, ended, immutable … " is an exception that did not stand the test of empirical study. Mechanisms of cellular and circuit-level plasticity continue to shape and reshape many regions of the adult nervous system long after the neurodevelopmental period. Initially focused on neurons alone, the field has followed a meteoric trajectory in understanding of activity-regulated neurodevelopment and ongoing neuroplasticity with an arc toward appreciating neuron-glial interactions and the role that each neural cell type plays in shaping adaptable neural circuity. In this review, as part of a celebration of the 50th anniversary of Society for Neuroscience, we provide a historical perspective, following this arc of inquiry from neuronal to neuron-glial mechanisms by which activity and experience modulate circuit structure and function. The scope of this consideration is broad, and it will not be possible to cover the wealth of knowledge about all aspects of activity-dependent circuit development and plasticity in depth.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305
| |
Collapse
|
87
|
Zeng B, Zhao G, Liu HL. The Differential Effect of Treadmill Exercise Intensity on Hippocampal Soluble Aβ and Lipid Metabolism in APP/PS1 Mice. Neuroscience 2020; 430:73-81. [PMID: 31954827 DOI: 10.1016/j.neuroscience.2020.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is characterized clinically by progressive impairments in learning and memory. Accumulating evidence suggests that regular exercise plays a neuroprotective role in aging-associated memory loss. Our previous study has confirmed that long-term treadmill exercise initiated either before or during the onset of β-amyloid (Aβ) pathology, was beneficial for reducing the levels of soluble Aβ and further improved cognition. In this study, in APP/PS1 mice, we assessed changes in soluble Aβ, and various blood biochemistry and molecular biological indices to assess whether exercise modulated lipid metabolism and thereby decelerated AD progression. Our results show that long-term treadmill exercise reduced the total cholesterol, triglyceride, and low-density lipoprotein cholesterol levels, and increased the level of high-density lipoprotein cholesterol. Exercise also decreased the levels of soluble Aβ1-40 and Aβ1-42, down-regulated retinoid X receptor expression, and up-regulated liver X receptor, Apolipoprotein E, Low density lipoprotein receptor, Low density lipoprotein receptor-related protein 1, and ATP-binding cassette transporter A1 expression. This indicates that long-term treadmill exercise alters the lipoprotein content, increases lipid metabolism and cholesterol transportation, reduces the soluble Aβ, and therein plays an important neuroprotective role and delays AD progression. We further show that medium exercise intensity (60%-70% of maximal oxygen uptake) was more efficacious in increasing lipid metabolism and reducing blood lipid levels and soluble Aβ levels, than low-intensity exercise (45-55% of maximal oxygen uptake). This research has broad prospects and implications, and offers a theoretical basis for the prevention of AD.
Collapse
Affiliation(s)
- B Zeng
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China
| | - G Zhao
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China
| | - H L Liu
- Department of Sports Medicine, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
88
|
Hippocampal Neurogenesis Is Enhanced in Adult Tau Deficient Mice. Cells 2020; 9:cells9010210. [PMID: 31947657 PMCID: PMC7016791 DOI: 10.3390/cells9010210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 12/22/2022] Open
Abstract
Tau dysfunction is common in several neurodegenerative diseases including Alzheimer’s disease (AD) and frontotemporal dementia (FTD). Affective symptoms have often been associated with aberrant tau pathology and are commonly comorbid in patients with tauopathies, indicating a connection between tau functioning and mechanisms of depression. The current study investigated depression-like behavior in Mapt−/− mice, which contain a targeted deletion of the gene coding for tau. We show that 6-month Mapt−/− mice are resistant to depressive behaviors, as evidenced by decreased immobility time in the forced swim and tail suspension tests, as well as increased escape behavior in a learned helplessness task. Since depression has also been linked to deficient adult neurogenesis, we measured neurogenesis in the hippocampal dentate gyrus and subventricular zone using 5-bromo-2-deoxyuridine (BrdU) labeling. We found that neurogenesis is increased in the dentate gyrus of 14-month-old Mapt−/− brains compared to wild type, providing a potential mechanism for their behavioral phenotypes. In addition to the hippocampus, an upregulation of proteins involved in neurogenesis was observed in the frontal cortex and amygdala of the Mapt−/− mice using proteomic mass spectrometry. All together, these findings suggest that tau may have a role in the depressive symptoms observed in many neurodegenerative diseases and identify tau as a potential molecular target for treating depression.
Collapse
|
89
|
Abstract
The Amyloid Precursor Protein (APP) is infamous for its proposed pivotal role in the pathogenesis of Alzheimer’s disease (AD). Much research on APP focusses on potential contributions to neurodegeneration, mostly based on mouse models with altered expression or mutated forms of APP. However, cumulative evidence from recent years indicates the indispensability of APP and its metabolites for normal brain physiology. APP contributes to the regulation of synaptic transmission, plasticity, and calcium homeostasis. It plays an important role during development and it exerts neuroprotective effects. Of particular importance is the soluble secreted fragment APPsα which mediates many of its physiological actions, often counteracting the effects of the small APP-derived peptide Aβ. Understanding the contribution of APP for normal functions of the nervous system is of high importance, both from a basic science perspective and also as a basis for generating new pathophysiological concepts and therapeutic approaches in AD. In this article, we review the physiological functions of APP and its metabolites, focusing on synaptic transmission, plasticity, calcium signaling, and neuronal network activity.
Collapse
Affiliation(s)
- Dimitri Hefter
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany.,RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Susann Ludewig
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany.,Cellular Neurobiology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Martin Korte
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany.,Cellular Neurobiology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany
| |
Collapse
|
90
|
Aβ modulates actin cytoskeleton via SHIP2-mediated phosphoinositide metabolism. Sci Rep 2019; 9:15557. [PMID: 31664099 PMCID: PMC6820556 DOI: 10.1038/s41598-019-51914-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/02/2019] [Indexed: 12/22/2022] Open
Abstract
Emerging evidences suggest that phospholipid metabolism is altered in Alzheimer’s disease (AD), but molecular mechanisms on how this affects neurodegeneration in AD is poorly understood. SHIP2 is a phosphoinositide-metabolizing enzyme, which dephosphorylates PI(3,4,5)P3 resulting to PI(3,4)P2, and it has been recently shown that Aβ directly increases the activity of SHIP2. Here we monitored, utilizing fluorescent SHIP2 biosensor, real-time increase of PI(3,4)P2-containing vesicles in HT22 cells treated with Aβ. Interestingly, PI(3,4)P2 is accumulated at late endosomes and lysosomal vesicles. We further discovered that ARAP3 can be attracted to PI(3,4)P2-positive mature endosomes via its PH domain and this facilitates the degradation of ARAP3. The reduced level of ARAP3 then causes RhoA hyperactivation and filamentous actin, which are critical for neurodegeneration in AD. These results provide a novel molecular link between Aβ and actin disruption through dysregulated phosphoinositide metabolism, and the SHIP2-PI(3,4)P2-ARAP3-RhoA signaling pathway can be considered as new therapeutic targets for synaptic dysfunctions in Alzheimer’s disease.
Collapse
|
91
|
Amadoro G, Latina V, Corsetti V, Calissano P. N-terminal tau truncation in the pathogenesis of Alzheimer's disease (AD): Developing a novel diagnostic and therapeutic approach. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165584. [PMID: 31676377 DOI: 10.1016/j.bbadis.2019.165584] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/04/2023]
Abstract
Tau truncation occurs at early stages during the development of human Alzheimer's disease (AD) and other tauopathy dementias. Tau cleavage, particularly in its N-terminal projection domain, is able to drive per se neurodegeneration, regardless of its pro-aggregative pathway(s) and in fragment(s)-dependent way. In this short review, we highlight the pathological relevance of the 20-22 kDa NH2-truncated tau fragment which is endowed with potent neurotoxic "gain-of-function" action(s), both in vitro and in vivo. An extensive comment on its clinical value as novel progression/diagnostic biomarker and potential therapeutic target in the context of tau-mediated neurodegeneration is also provided.
Collapse
Affiliation(s)
- G Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy; Institute of Translational Pharmacology (IFT)-CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| | - V Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - V Corsetti
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - P Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| |
Collapse
|
92
|
Morrissey JA, Bigus E, Necarsulmer JC, Srinivasan V, Peppercorn K, O'Leary DJ, Mockett BG, Tate WP, Hughes SM, Parfitt KD, Abraham WC. The Tripeptide RER Mimics Secreted Amyloid Precursor Protein-Alpha in Upregulating LTP. Front Cell Neurosci 2019; 13:459. [PMID: 31680870 PMCID: PMC6813913 DOI: 10.3389/fncel.2019.00459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022] Open
Abstract
Secreted amyloid precursor protein-alpha (sAPPα), generated by enzymatic processing of the APP, possesses a range of neurotrophic and neuroprotective properties and plays a critical role in the molecular mechanisms of memory and learning. One of the key active regions of sAPPα is the central APP domain (E2) that contains within it the tripeptide sequence, RER. This sequence is exposed on the surface of a coiled coil substructure of E2. RER has by itself displayed memory-enhancing properties, and can protect newly formed engrams from interference in a manner similar to that displayed by sAPPα itself. In order to determine whether RER mimics other properties of sAPPα, we investigated the electrophysiological effects of the N-terminal protected acetylated RER (Ac-RER) and an isoform containing a chiral switch in the first amino acid from an l- to a d-orientation (Ac-rER), on synaptic plasticity. We found that, like sAPPα, exogenous perfusion with nanomolar concentrations of Ac-RER or Ac-rER enhanced the induction and stability of long-term potentiation (LTP) in area CA1 of rat and mouse hippocampal slices, in a protein synthesis- and trafficking-dependent manner. This effect did not occur with a control Ac-AAA or Ac-IFR tripeptide, nor with a full-length sAPPα protein where RER was substituted with AAA. Ac-rER also protected LTP against amyloid-beta (Aβ25–35)-induced LTP impairment. Our findings provide further evidence that the RER-containing region of sAPPα is functionally significant and by itself can produce effects similar to those displayed by full length sAPPα, suggesting that this tripeptide, like sAPPα, may have therapeutic potential.
Collapse
Affiliation(s)
- Jodi A Morrissey
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand.,Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Erin Bigus
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | | | - Vinay Srinivasan
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | - Katie Peppercorn
- Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Daniel J O'Leary
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | - Bruce G Mockett
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Warren P Tate
- Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Stephanie M Hughes
- Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Karen D Parfitt
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| |
Collapse
|
93
|
KCa3.1 deficiency attenuates neuroinflammation by regulating an astrocyte phenotype switch involving the PI3K/AKT/GSK3β pathway. Neurobiol Dis 2019; 132:104588. [PMID: 31470105 DOI: 10.1016/j.nbd.2019.104588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/22/2019] [Accepted: 08/23/2019] [Indexed: 12/26/2022] Open
Abstract
Neuroinflammation may induce a phenotype switch to reactive astrogliosis in neurodegenerative disorders. The calcium-activated potassium channel (KCa3.1) is active in the phenotypic switch that occurs during astrogliosis in Alzheimer's disease and ischemic stroke. Here, transcriptome sequencing (RNA-Seq), immunohistochemistry, western blotting, pharmacological blockade, and calcium imaging were used to investigate astrocyte KCa3.1 activity in neuroinflammation, Tau accumulation, and insulin signaling deficits in male wild-type C57BL/6 and KCa3.1-/- knockout (KO) mice, and in primary astrocyte cultures. KCa3.1 deficiency in KO mice decreased lipopolysaccharide (LPS)-induced memory deficits, neuronal loss, glial activation, Tau phosphorylation, and insulin signaling deficits in vivo. KCa3.1 expression in astrocytes was associated with LPS-induced upregulation of the Orai1 store-operated Ca2+ channel protein. The KCa3.1 channel was found to regulate store-operated Ca2+ overload through an interaction with Orai1 in LPS-induced reactive astrocytes. The LPS-induced effects on KCa3.1 and Orai1 indirectly promoted astrogliosis-related changes via the PI3K/AKT/GSK3β and NF-κB signaling pathways in vitro. Unbiased evaluation of RNA-Seq results for actively translated RNAs confirmed that substantial astrocyte diversity was associated with KCa3.1 deficiency. Our results suggest that KCa3.1 regulated astrogliosis-mediated neuroinflammation, Tau accumulation, and insulin signaling deficiency via PI3K/AKT/GSK3β and NF-κB signaling pathways, and contributing to neuronal loss and memory deficits in this neuroinflammation mouse model.
Collapse
|
94
|
Taniguchi K, Yamamoto F, Arai T, Yang J, Sakai Y, Itoh M, Mamada N, Sekiguchi M, Yamada D, Saitoh A, Kametani F, Tamaoka A, Araki YM, Wada K, Mizusawa H, Araki W. Tyrosol Reduces Amyloid-β Oligomer Neurotoxicity and Alleviates Synaptic, Oxidative, and Cognitive Disturbances in Alzheimer’s Disease Model Mice. J Alzheimers Dis 2019; 70:937-952. [DOI: 10.3233/jad-190098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Kaori Taniguchi
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Fumiko Yamamoto
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takuya Arai
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Jinwei Yang
- Tokiwa Phytochemical Co., Ltd, Sakura, Chiba, Japan
| | - Yusuke Sakai
- Tokiwa Phytochemical Co., Ltd, Sakura, Chiba, Japan
| | - Masayuki Itoh
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Naomi Mamada
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masayuki Sekiguchi
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Daisuke Yamada
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Akiyoshi Saitoh
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yumiko M. Araki
- Department of Psychiatry and Behavioral Science, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Keiji Wada
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Hidehiro Mizusawa
- National Center Hospital, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| | - Wataru Araki
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo, Japan
| |
Collapse
|
95
|
3D Electron Microscopy Study of Synaptic Organization of the Normal Human Transentorhinal Cortex and Its Possible Alterations in Alzheimer's Disease. eNeuro 2019; 6:ENEURO.0140-19.2019. [PMID: 31217195 PMCID: PMC6620390 DOI: 10.1523/eneuro.0140-19.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 01/10/2023] Open
Abstract
The transentorhinal cortex (TEC) is an obliquely oriented cortex located in the medial temporal lobe and, together with the entorhinal cortex, is one of the first affected areas in Alzheimer’s disease (AD). One of the most widely accepted hypotheses is that synaptopathy (synaptic alterations and loss) represents the major structural correlate of the cognitive decline observed in AD. However, very few electron microscope (EM) studies are available; the most common method to estimate synaptic density indirectly is by counting, at the light microscopic level, immunoreactive puncta using synaptic markers. To investigate synaptic morphology and possible alterations related to AD, a detailed three-dimensional (3D) ultrastructural analysis using focused ion beam/scanning EM (FIB/SEM) was performed in the neuropil of Layer II of the TEC in human brain samples from non-demented subjects and AD patients. Evaluation of the proportion and shape of asymmetric synapses (AS) and symmetric synapses (SS) targeting spines or dendritic shafts was performed using 3D reconstructions of every synapse. The 3D analysis of 4722 synapses revealed that the preferable targets were spine heads for AS and dendritic shafts for SS, both in control and AD cases. However, in AD patients, we observed a reduction in the percentage of synapses targeting spine heads. Regarding the shape of synapses, in both control cases and AD samples, the vast majority of synapses had a macular shape, followed by perforated or horseshoe-shaped synapses, with fragmented synapses being the least frequent type. Moreover, comparisons showed an increased number of fragmented AS in AD patients.
Collapse
|
96
|
Miao Y, Wang N, Shao W, Xu Z, Yang Z, Wang L, Ju C, Zhang R, Zhang F. Overexpression of TIPE2, a Negative Regulator of Innate and Adaptive Immunity, Attenuates Cognitive Deficits in APP/PS1 Mice. J Neuroimmune Pharmacol 2019; 14:519-529. [DOI: 10.1007/s11481-019-09861-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/30/2019] [Indexed: 12/29/2022]
|
97
|
Stockburger C, Eckert S, Eckert GP, Friedland K, Müller WE. Mitochondrial Function, Dynamics, and Permeability Transition: A Complex Love Triangle as A Possible Target for the Treatment of Brain Aging and Alzheimer's Disease. J Alzheimers Dis 2019; 64:S455-S467. [PMID: 29504539 DOI: 10.3233/jad-179915] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Because of the failure of all amyloid-β directed treatment strategies for Alzheimer's disease (AD), the concept of mitochondrial dysfunction as a major pathomechanism of the cognitive decline in aging and AD has received substantial support. Accordingly, improving mitochondrial function as an alternative strategy for new drug development became of increasing interest and many different compounds have been identified which improve mitochondrial function in preclinical in vitro and in vivo experiments. However, very few if any have been investigated in clinical trials, representing a major drawback of the mitochondria directed drug development. To overcome these problems, we used a top-down approach by investigating several older antidementia drugs with clinical evidence of therapeutic efficacy. These include EGb761® (standardized ginkgo biloba extract), piracetam, and Dimebon. All improve experimentally many aspects of mitochondrial dysfunction including mitochondrial dynamics and also improve cognition and impaired neuronal plasticity, the functionally most relevant consequences of mitochondrial dysfunction. All partially inhibit opening events of the mitochondrial permeability transition pore (mPTP) which previously has mainly been discussed as a mechanism relevant for the induction of apoptosis. However, as more recent work suggests the mPTP as a master regulator of many mitochondrial functions, our data suggest the mPTP as a possible relevant drug target within the love triangle between mPTP regulation, mitochondrial dynamics, and mitochondrial function including regulation of neuronal plasticity. Drugs interfering with mPTP function will improve not only mitochondrial impairment in aging and AD but also will have beneficial effects on impaired neuronal plasticity, the pathomechanism which correlates best with functional deficits (cognition, behavior) in aging and AD.
Collapse
Affiliation(s)
- Carola Stockburger
- Department of Pharmacology, University of Frankfurt/M, Biocenter, Frankfurt/Main, Germany
| | - Schamim Eckert
- Department of Pharmacology, University of Frankfurt/M, Biocenter, Frankfurt/Main, Germany
| | - Gunter P Eckert
- Department of Nutritional Sciences, University of Giessen, Giessen, Germany
| | - Kristina Friedland
- Department of Molecular and Clinical Pharmacy, University of Erlangen, Erlangen, Germany
| | - Walter E Müller
- Department of Pharmacology, University of Frankfurt/M, Biocenter, Frankfurt/Main, Germany
| |
Collapse
|
98
|
Meng G, Mei H. Transcriptional Dysregulation Study Reveals a Core Network Involving the Progression of Alzheimer's Disease. Front Aging Neurosci 2019; 11:101. [PMID: 31133844 PMCID: PMC6513962 DOI: 10.3389/fnagi.2019.00101] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/15/2019] [Indexed: 12/26/2022] Open
Abstract
Background: The pathogenesis of Alzheimer's disease is associated with dysregulation at different levels from transcriptome to cellular functioning. Such complexity necessitates investigations of disease etiology to be carried out considering multiple aspects of the disease and the use of independent strategies. The established works more emphasized on the structural organization of gene regulatory network while neglecting the internal regulation changes. Methods: Applying a strategy different from popularly used co-expression network analysis, this study investigated the transcriptional dysregulations during the transition from normal to disease states. Results: Ninety- seven genes were predicted as dysregulated genes, which were also associated with clinical outcomes of Alzheimer's disease. Both the co-expression and differential co-expression analysis suggested these genes to be interconnected as a core network and that their regulations were strengthened during the transition to disease states. Functional studies suggested the dysregulated genes to be associated with aging and synaptic function. Further, we checked the conservation of the gene co-expression and found that human and mouse brain might have divergent transcriptional co-regulation even when they had conserved gene expression profiles. Conclusion: Overall, our study reveals a core network of transcriptional dysregulation associated with the progression of Alzheimer's disease by affecting the aging and synaptic functions related genes; the gene regulation is not conserved in the human and mouse brains.
Collapse
Affiliation(s)
- Guofeng Meng
- Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Computational and Modeling Science, PTS China, GSK R&D, Shanghai, China
| | - Hongkang Mei
- Computational and Modeling Science, PTS China, GSK R&D, Shanghai, China
| |
Collapse
|
99
|
Berberine Alleviates Amyloid β-Induced Mitochondrial Dysfunction and Synaptic Loss. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7593608. [PMID: 31191803 PMCID: PMC6525905 DOI: 10.1155/2019/7593608] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/12/2019] [Indexed: 11/18/2022]
Abstract
Synaptic structural and functional damage is a typical pathological feature of Alzheimer's disease (AD). Normal axonal mitochondrial function and transportation are vital to synaptic function and plasticity because they are necessary for maintaining cellular energy supply and regulating calcium and redox signalling as well as synaptic transmission and vesicle release. Amyloid-β (Aβ) accumulation is another pathological hallmark of AD that mediates synaptic loss and dysfunction by targeting mitochondria. Therefore, it is important to develop strategies to protect against synaptic mitochondrial damage induced by Aβ. The present study examined the beneficial effects of berberine, a natural isoquinoline alkaloid extracted from the traditional medicinal plant Coptis chinensis, on Aβ-induced mitochondrial and synaptic damage in primary cultured hippocampal neurons. We demonstrate that berberine alleviates axonal mitochondrial abnormalities by preserving the mitochondrial membrane potential and preventing decreases in ATP, increasing axonal mitochondrial density and length, and improving mitochondrial motility and trafficking in cultured hippocampal neurons. Although the underlying protective mechanism remains to be elucidated, the data suggest that the effects of berberine were in part related to its potent antioxidant activity. These findings highlight the neuroprotective and specifically mitoprotective effects of berberine treatment under conditions of Aβ enrichment.
Collapse
|
100
|
Ansari Dezfouli M, Zahmatkesh M, Farahmandfar M, Khodagholi F. Melatonin protective effect against amyloid β-induced neurotoxicity mediated by mitochondrial biogenesis; involvement of hippocampal Sirtuin-1 signaling pathway. Physiol Behav 2019; 204:65-75. [DOI: 10.1016/j.physbeh.2019.02.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 02/03/2019] [Accepted: 02/11/2019] [Indexed: 11/30/2022]
|