1
|
Salem S, Alpaugh M, Saint-Pierre M, Alves-Martins-Borba FN, Cerquera-Cleves C, Lemieux M, Ngonza-Nito SB, De Koninck P, Melki R, Cicchetti F. Treatment with Tau fibrils impact Huntington's disease-related phenotypes in cell and mouse models. Neurobiol Dis 2024; 202:106696. [PMID: 39389154 DOI: 10.1016/j.nbd.2024.106696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/13/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
There is now compelling evidence for the presence of pathological forms of Tau in tissues of both patients and animal models of Huntington's disease (HD). While the root cause of this illness is a mutation within the huntingtin gene, a number of studies now suggest that HD could also be considered a secondary tauopathy. However, the contributory role of Tau in the pathogenesis and pathophysiology of this condition, as well as its implications in cellular toxicity and consequent behavioral impairments are largely unknown. We therefore performed intracerebral stereotaxic injections of recombinant human Tau monomers and fibrils into the knock-in zQ175 mouse model of HD. Tau fibrils induced cognitive and anxiety-like phenotypes predominantly in zQ175 mice and increased the number and size of insoluble mutant huntingtin (mHTT) aggregates in the brains of treated animals. To better understand the putative mechanisms through which Tau could initiate and/or contribute to pathology, we incubated StHdh striatal cells, an in vitro model of HD, with the different Tau forms and evaluated the effects on cell functionality and heat shock proteins Hsp70 and Hsp90. Calcium imaging experiments showed functional impairments of HD StHdh cells following treatment with Tau fibrils, as well as significant changes to the levels of both heat shock proteins which were found trapped within mHTT aggregates. The accumulation of Hsp70 and 90 within aggregates was also present in mouse tissue which suggests that alteration of molecular chaperone-dependent protein quality control may influence aggregation, implicating proteostasis in the mHTT-Tau interplay.
Collapse
Affiliation(s)
- Shireen Salem
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| | - Melanie Alpaugh
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Martine Saint-Pierre
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Flavia Natale Alves-Martins-Borba
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Catalina Cerquera-Cleves
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Mado Lemieux
- CERVO Brain Research Center, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Soki Bradel Ngonza-Nito
- Labortory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, Fontenay-aux-Roses, France
| | - Paul De Koninck
- CERVO Brain Research Center, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Ronald Melki
- Labortory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, Fontenay-aux-Roses, France
| | - Francesca Cicchetti
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
2
|
Chu D, Yang X, Wang J, Zhou Y, Gu JH, Miao J, Wu F, Liu F. Tau truncation in the pathogenesis of Alzheimer's disease: a narrative review. Neural Regen Res 2024; 19:1221-1232. [PMID: 37905868 PMCID: PMC11467920 DOI: 10.4103/1673-5374.385853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/07/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Alzheimer's disease is characterized by two major neuropathological hallmarks-the extracellular β-amyloid plaques and intracellular neurofibrillary tangles consisting of aggregated and hyperphosphorylated Tau protein. Recent studies suggest that dysregulation of the microtubule-associated protein Tau, especially specific proteolysis, could be a driving force for Alzheimer's disease neurodegeneration. Tau physiologically promotes the assembly and stabilization of microtubules, whereas specific truncated fragments are sufficient to induce abnormal hyperphosphorylation and aggregate into toxic oligomers, resulting in them gaining prion-like characteristics. In addition, Tau truncations cause extensive impairments to neural and glial cell functions and animal cognition and behavior in a fragment-dependent manner. This review summarizes over 60 proteolytic cleavage sites and their corresponding truncated fragments, investigates the role of specific truncations in physiological and pathological states of Alzheimer's disease, and summarizes the latest applications of strategies targeting Tau fragments in the diagnosis and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xingyue Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Jin Miao
- Laboratory of Animal Center, Nantong University, Nantong, Jiangsu Province, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
3
|
Shim YJ, Jung WH, Billig AJ, Sedley W, Song JJ. Hippocampal atrophy is associated with hearing loss in cognitively normal adults. Front Neurosci 2023; 17:1276883. [PMID: 37942139 PMCID: PMC10628109 DOI: 10.3389/fnins.2023.1276883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Objectives A growing body of evidence suggests that age-related hearing loss (HL) is associated with morphological changes of the cerebral cortex, but the results have been drawn from a small amount of data in most studies. The aim of this study is to investigate the correlation between HL and gray matter volume (GMV) in a large number of subjects, strictly controlling for an extensive set of possible biases. Methods Medical records of 576 subjects who underwent pure tone audiometry, brain magnetic resonance imaging (MRI), and the Korean Mini-Mental State Exam (K-MMSE) were reviewed. Among them, subjects with normal cognitive function and free of central nervous system disorders or coronary artery disease were included. Outliers were excluded after a sample homogeneity check. In the end, 405 subjects were enrolled. Pure tone hearing thresholds were determined at 0.5, 1, 2, and 4 kHz in the better ear. Enrolled subjects were divided into 3 groups according to pure tone average: normal hearing (NH), mild HL (MHL), and moderate-to-severe HL (MSHL) groups. Using voxel-based morphometry, we evaluated GMV changes that may be associated with HL. Sex, age, total intracranial volume, type of MRI scanner, education level, K-MMSE score, smoking status, and presence of hypertension, diabetes mellitus and dyslipidemia were used as covariates. Results A statistically significant negative correlation between the hearing thresholds and GMV of the hippocampus was elucidated. Additionally, in group comparisons, the left hippocampal GMV of the MSHL group was significantly smaller than that of the NH and MHL groups. Conclusion Based on the negative correlation between hearing thresholds and hippocampal GMV in cognitively normal old adults, the current study indicates that peripheral deafferentation could be a potential contributing factor to hippocampal atrophy.
Collapse
Affiliation(s)
- Ye Ji Shim
- Department of Otorhinolaryngology-Head and Neck Surgery, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Republic of Korea
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Wi Hoon Jung
- Department of Psychology, Gachon University, Seongnam, Republic of Korea
| | | | - William Sedley
- Translational and Clinical Research Institute, Newcastle University Medical School, Newcastle upon Tyne, United Kingdom
| | - Jae-Jin Song
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
4
|
Baazaoui N, Iqbal K. Alzheimer's Disease: Challenges and a Therapeutic Opportunity to Treat It with a Neurotrophic Compound. Biomolecules 2022; 12:biom12101409. [PMID: 36291618 PMCID: PMC9599095 DOI: 10.3390/biom12101409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with an insidious onset and multifactorial nature. A deficit in neurogenesis and synaptic plasticity are considered the early pathological features associated with neurofibrillary tau and amyloid β pathologies and neuroinflammation. The imbalance of neurotrophic factors with an increase in FGF-2 level and a decrease in brain derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) in the hippocampus, frontal cortex and parietal cortex and disruption of the brain micro-environment are other characteristics of AD. Neurotrophic factors are crucial in neuronal differentiation, maturation, and survival. Several attempts to use neurotrophic factors to treat AD were made, but these trials were halted due to their blood-brain barrier (BBB) impermeability, short-half-life, and severe side effects. In the present review we mainly focus on the major etiopathology features of AD and the use of a small neurotrophic and neurogenic peptide mimetic compound; P021 that was discovered in our laboratory and was found to overcome the difficulties faced in the administration of the whole neurotrophic factor proteins. We describe pre-clinical studies on P021 and its potential as a therapeutic drug for AD and related neurodegenerative disorders. Our study is limited because it focuses only on P021 and the relevant literature; a more thorough investigation is required to review studies on various therapeutic approaches and potential drugs that are emerging in the AD field.
Collapse
Affiliation(s)
- Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
- Correspondence: ; Tel.: +1-718-494-5259; Fax: +1-718-494-1080
| |
Collapse
|
5
|
Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev 2022; 78:101636. [PMID: 35490966 PMCID: PMC9168971 DOI: 10.1016/j.arr.2022.101636] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 12/11/2022]
Abstract
Adult neurogenesis, the process by which neurons are generated in certain areas of the adult brain, declines in an age-dependent manner and is one potential target for extending cognitive healthspan. Aging is a major risk factor for neurodegenerative diseases and, as lifespans are increasing, these health challenges are becoming more prevalent. An age-associated loss in neural stem cell number and/or activity could cause this decline in brain function, so interventions that reverse aging in stem cells might increase the human cognitive healthspan. In this review, we describe the involvement of adult neurogenesis in neurodegenerative diseases and address the molecular mechanistic aspects of neurogenesis that involve some of the key aggregation-prone proteins in the brain (i.e., tau, Aβ, α-synuclein, …). We summarize the research pertaining to interventions that increase neurogenesis and regulate known targets in aging research, such as mTOR and sirtuins. Lastly, we share our outlook on restoring the levels of neurogenesis to physiological levels in elderly individuals and those with neurodegeneration. We suggest that modulating neurogenesis represents a potential target for interventions that could help in the fight against neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Luka Culig
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xixia Chu
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
6
|
Rajkumar M, Sakthivel M, Senthilkumar K, Thangaraj R, Kannan S. Galantamine tethered hydrogel as a novel therapeutic target for streptozotocin-induced Alzheimer's disease in Wistar rats. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100100. [PMID: 35510084 PMCID: PMC9058960 DOI: 10.1016/j.crphar.2022.100100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 01/20/2023] Open
Abstract
Amyloid-β (Aβ) plaque formation, neuronal cell death, and cognitive impairment are the unique symptoms of Alzheimer's disease (AD). No single step remedy is available to treat AD, so the present study aimed to improve the drugability and minimize the abnormal behavioral and biochemical activities in streptozotocin (STZ) induced AD experimental Wistar rats. In particular, we explored the utilization of methacrylated gelatin (GelMA), which is a biopolymeric hydrogel that mimics the natural tissue environment. The synthesized biopolymeric gel contained the drug galantamine (Gal). Investigations were conducted to evaluate the behavioral activities of STZ-induced AD experimental rats under STZ + GelMA + Gal treatment. The experimental groups comprised the control and STZ, STZ + GelMA, STZ + Gal, and STZ + GelMA + Gal (10 mg/kg) treated rats. Intracerebroventricular STZ ensures cognitive decline in terms of an increase in the escape latency period, with a decrease in the spontaneous alteration of behavioral activities. Our results indicated decrease Aβ aggregation in the hydrogel-based drug treatment group and significant decreases in the levels of acetylcholinesterase and lipid peroxidation (p < 0.001). In addition, the glutathione and superoxide dismutase activities appeared to be improved in the STZ + GelMA + Gal group compared with the other treatment groups. Furthermore, histopathological and immunohistochemical experiments showed that the GelMA + Gal treated AD rats exhibited significantly improved behavioral and biochemical activities compared with the STZ treated AD rats. Therefore, STZ + GelMA + Gal administration from the pre-plaque stage may have a potential clinical application in the prevention of AD. Thus, we conclude that hydrogel-based Gal drugs are efficient at decreasing Aβ aggregation and improving the neuroinflammatory process, antioxidant activity, and neuronal growth.
Collapse
Affiliation(s)
- Manickam Rajkumar
- Cancer Nanomedicine Laboratory, Department of Zoology, School of Life Sciences, Periyar University, Salem, 636 011, Tamil Nadu, India
| | - Murugesan Sakthivel
- Cancer Nanomedicine Laboratory, Department of Zoology, School of Life Sciences, Periyar University, Salem, 636 011, Tamil Nadu, India
| | - Kottaisamy Senthilkumar
- National Institute for Research in Tuberculosis/Indian Council for Medical Research, Ward No. 62, Government Rajaji Hospital Madurai, 625 001, Tamil Nadu, India
| | - Ramasundaram Thangaraj
- Vermitechnology and Ecotoxicology Laboratory, Department of Zoology, School of Life Sciences, Periyar University, Salem, 636 011, Tamil Nadu, India
| | - Soundarapandian Kannan
- Cancer Nanomedicine Laboratory, Department of Zoology, School of Life Sciences, Periyar University, Salem, 636 011, Tamil Nadu, India
| |
Collapse
|
7
|
Weerasinghe-Mudiyanselage PDE, Ang MJ, Kang S, Kim JS, Moon C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:3349. [PMID: 35328770 PMCID: PMC8955928 DOI: 10.3390/ijms23063349] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/10/2022] Open
Abstract
Neuroplasticity is the capacity of neural networks in the brain to alter through development and rearrangement. It can be classified as structural and functional plasticity. The hippocampus is more susceptible to neuroplasticity as compared to other brain regions. Structural modifications in the hippocampus underpin several neurodegenerative diseases that exhibit cognitive and emotional dysregulation. This article reviews the findings of several preclinical and clinical studies about the role of structural plasticity in the hippocampus in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In this study, literature was surveyed using Google Scholar, PubMed, Web of Science, and Scopus, to review the mechanisms that underlie the alterations in the structural plasticity of the hippocampus in neurodegenerative diseases. This review summarizes the role of structural plasticity in the hippocampus for the etiopathogenesis of neurodegenerative diseases and identifies the current focus and gaps in knowledge about hippocampal dysfunctions. Ultimately, this information will be useful to propel future mechanistic and therapeutic research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Poornima D. E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
- College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| |
Collapse
|
8
|
Hsu WH, Huang NK, Shiao YJ, Lu CK, Chao YM, Huang YJ, Yeh CH, Lin YL. Gastrodiae rhizoma attenuates brain aging via promoting neuritogenesis and neurodifferentiation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 87:153576. [PMID: 33985879 DOI: 10.1016/j.phymed.2021.153576] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 03/23/2021] [Accepted: 04/15/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Gastrodiae Rhizoma (Tianma), the dried tuber of Gastrodia elata Bl. (Orchidaceae), is listed as a top-grade herbal medicine in Shen-nong Ben-ts'ao Jing and has been used for treating headaches, dizziness, vertigo and convulsion. It has a neuroprotective effect and extends the lifespan in mouse models of Huntington's disease and Niemann-Pick type C disease. However, its effect on senescence remains unknown. PURPOSE This study aimed to investigate the anti-aging effects and the underlying mechanism of Gastrodiae Rhizoma. METHODS D-galactose (D-gal)- and BeSO4-induced cellular senescence and senescence-associated β-galactosidase (SA-β-gal) activity were evaluated in SH-SY5Y and PC12 cells. D-gal-induced aging mice were used as an in vivo model. Animal behaviors including nesting and burrowing and Morris water maze were conducted. Neurogenesis in the hippocampus was assessed by immunohistochemistry and confocal microscopy, and the aging-related proteins were assessed by Western blot analysis. The potential neuritogenesis activity of the partially purified fraction of Gastrodiae Rhizoma (TM-2) and its major ingredients were investigated in PC12 cells. RESULTS TM-2 could improve D-gal-induced learning and memory impairement by inhibiting oxidative stress, increasing hippocampal neurogenesis and regulating the SH2B1-Akt pathway. Moreover, N6-(4-hydroxybenzyl)adenine riboside (T1-11) and parishins A and B, three constituents of TM-2, had anti-aging activity, as did T1-11 and parishin A induced neuritogenesis. CONCLUSION Our data suggested that TM-2 slowed down D-gal-induced cellular and mouse brain aging. These results indicate that Gastrodiae Rhizoma has a beneficial effect on senescence. It may be used for neuroprotection and promoting neurogenesis.
Collapse
Affiliation(s)
- Wei-Hsiang Hsu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan
| | - Nai-Kuei Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan; Institute of Biopharmaceutical Science, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chung-Kuang Lu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11221, Taiwan
| | - Yen-Ming Chao
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan
| | - Yi-Jeng Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan
| | - Chih-Hsin Yeh
- Taoyuan District Agricultural Research and Extension Station, Council of Agriculture, Executive Yuan, Taoyuan 32754, Taiwan
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40402, Taiwan; Department of Pharmacy, National Taiwan University, Taipei 10050, Taiwan.
| |
Collapse
|
9
|
Liu H, Zhang H, Ma Y. Molecular mechanisms of altered adult hippocampal neurogenesis in Alzheimer's disease. Mech Ageing Dev 2021; 195:111452. [PMID: 33556365 DOI: 10.1016/j.mad.2021.111452] [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: 08/17/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia globally. AD is a progressive neurodegenerative disorder, eventually manifesting as severe cognitive impairment. Adult hippocampal neurogenesis (AHN) occurs throughout adulthood and plays an important role in hippocampus-dependent learning and memory. The stages of AHN, predominantly comprising the proliferation, differentiation, survival, and maturation of newborn neurons, are affected to varying degrees in AD. However, the exact molecular mechanisms remain to be elucidated. Recent evidence suggests that the molecules involved in AD pathology contribute to the compromised AHN in AD. Notably, various interventions may have common signaling pathways that, once identified, could be harnessed to enhance adult neurogenesis. This in turn could putatively rescue cognitive deficits associated with impaired neurogenesis as observed in animal models of AD. In this manuscript, we review the current knowledge concerning AHN under normal physiological and AD pathological conditions and highlight the possible role of specific molecules in AHN alteration in AD. In addition, we summarize in vivo experiments with emphasis on the effect of the activation of certain key signalings on AHN in AD rodent models. We propose that these signaling targets and corresponding interventions should be considered when developing novel therapies for AD.
Collapse
Affiliation(s)
- Hang Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Han Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ying Ma
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
10
|
Li X, Jiang M, Cheng J, Ye M, Zhang W, Jaffrezic-Renault N, Guo Z. Signal multi-amplified electrochemical biosensor for voltammetric determination of tau-441 protein in biological samples using carbon nanomaterials and gold nanoparticles to hint dementia. Mikrochim Acta 2020; 187:302. [PMID: 32350619 DOI: 10.1007/s00604-020-04273-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 04/11/2020] [Indexed: 12/22/2022]
Abstract
A signal multi-amplified electrochemical biosensor was fabricated for tau-441 protein, a dementia biomarker. It utilizes a carbon nanocomposite film modified gold electrode. The carbon nanocomposite film was composed of multi-walled carbon nanotubes (MWCNTs), reduced graphene oxide (rGO), and chitosan (CS). For the nanocomposite film, rGO improved the dispersibility of MWCNTs, and the effective surface area of MWCNTs was increased. On the other hand, MWCNTs also increased the interlayer spacing of rGO, resulting in a thinner rGO layer. MWCNTs-rGO had a better conductivity than that of MWCNTs and rGO due to the synergy effect. Biocompatible CS was employed for immobilization of the specific antibody. Tau-441 protein was modified with gold nanoparticles (AuNPs) for signal amplification again. The response of the electrochemical biosensor is linear in the range 0.5-80 fM (0.5, 1.5, 5, 10, 40, 80 fM) with a limit of detection (LOD) of 0.46 fM, using differential pulse voltammetry (DPV) in a potential range of - 100-500 mV. The biosensor was successfully applied to the analysis of serum samples of 14 normal people, 14 mild cognitive impairment (MCI) patients, and 14 dementia patients. Graphical abstract Schematic representation of signal multi-amplified electrochemical biosensor for determination of tau-441 protein in human serum.
Collapse
Affiliation(s)
- Xuanying Li
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
| | - Mingdi Jiang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
| | - Jing Cheng
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
| | - Mengsha Ye
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
- School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China
| | - Weiying Zhang
- Institute for Interdisciplinary Research, Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan, 430056, People's Republic of China
| | - Nicole Jaffrezic-Renault
- Institute of Analytical Sciences, UMR-CNRS 5280, University of Lyon, 5, La Doua Street, 69100, Villeurbanne, France
| | - Zhenzhong Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, People's Republic of China.
| |
Collapse
|
11
|
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
|
12
|
Twendee X Ameliorates Phosphorylated Tau, α-Synuclein and Neurovascular Dysfunction in Alzheimer's Disease Transgenic Mice With Chronic Cerebral Hypoperfusion. J Stroke Cerebrovasc Dis 2019; 28:104310. [DOI: 10.1016/j.jstrokecerebrovasdis.2019.104310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/09/2019] [Accepted: 07/21/2019] [Indexed: 11/23/2022] Open
|
13
|
Sarkar SN, Russell AE, Engler-Chiurazzi EB, Porter KN, Simpkins JW. MicroRNAs and the Genetic Nexus of Brain Aging, Neuroinflammation, Neurodegeneration, and Brain Trauma. Aging Dis 2019; 10:329-352. [PMID: 31011481 PMCID: PMC6457055 DOI: 10.14336/ad.2018.0409] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex and integrated gradual deterioration of cellular activities in specific organs of the body, which is associated with increased mortality. This deterioration is the primary risk factor for major human pathologies, including cancer, diabetes, cardiovascular disorders, neurovascular disorders, and neurodegenerative diseases. There are nine tentative hallmarks of aging. In addition, several of these hallmarks are increasingly being associated with acute brain injury conditions. In this review, we consider the genes and their functional pathways involved in brain aging as a means of developing new strategies for therapies targeted to the neuropathological processes themselves, but also as targets for many age-related brain diseases. A single microRNA (miR), which is a short, non-coding RNA species, has the potential for targeting many genes simultaneously and, like practically all other cellular processes, genes associated with many features of brain aging and injury are regulated by miRs. We highlight how certain miRs can mediate deregulation of genes involved in neuroinflammation, acute neuronal injury and chronic neurodegenerative diseases. Finally, we review the recent progress in the development of effective strategies to block specific miR functions and discuss future approaches with the prediction that anti-miR drugs may soon be used in the clinic.
Collapse
Affiliation(s)
- Saumyendra N Sarkar
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Ashley E Russell
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Elizabeth B Engler-Chiurazzi
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Keyana N Porter
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - James W Simpkins
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
14
|
Free d-aspartate triggers NMDA receptor-dependent cell death in primary cortical neurons and perturbs JNK activation, Tau phosphorylation, and protein SUMOylation in the cerebral cortex of mice lacking d-aspartate oxidase activity. Exp Neurol 2019; 317:51-65. [PMID: 30822420 DOI: 10.1016/j.expneurol.2019.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/19/2019] [Accepted: 02/24/2019] [Indexed: 11/23/2022]
Abstract
In mammals, free d-aspartate (D-Asp) is abundant in the embryonic brain, while levels remain very low during adulthood as a result of the postnatal expression and activity of the catabolizing enzyme d-aspartate oxidase (DDO). Previous studies have shown that long-lasting exposure to nonphysiological, higher D-Asp concentrations in Ddo knockout (Ddo-/-) mice elicits a precocious decay of synaptic plasticity and cognitive functions, along with a dramatic age-dependent expression of active caspase 3, associated with increased cell death in different brain regions, including hippocampus, prefrontal cortex, and substantia nigra pars compacta. Here, we investigate the yet unclear molecular and cellular events associated with the exposure of abnormally high D-Asp concentrations in cortical primary neurons and in the brain of Ddo-/- mice. For the first time, our in vitro findings document that D-Asp induces in a time-, dose-, and NMDA receptor-dependent manner alterations in JNK and Tau phosphorylation levels, associated with pronounced cell death in primary cortical neurons. Moreover, observations obtained in Ddo-/- animals confirmed that high in vivo levels of D-Asp altered cortical JNK signaling, Tau phosphorylation and enhanced protein SUMOylation, indicating a robust indirect role of DDO activity in regulating these biochemical NMDA receptor-related processes. Finally, no gross modifications in D-Asp concentrations and DDO mRNA expression were detected in the cortex of patients with Alzheimer's disease when compared to age-matched healthy controls.
Collapse
|
15
|
D'Souza MS. Brain and Cognition for Addiction Medicine: From Prevention to Recovery Neural Substrates for Treatment of Psychostimulant-Induced Cognitive Deficits. Front Psychiatry 2019; 10:509. [PMID: 31396113 PMCID: PMC6667748 DOI: 10.3389/fpsyt.2019.00509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/28/2019] [Indexed: 01/10/2023] Open
Abstract
Addiction to psychostimulants like cocaine, methamphetamine, and nicotine poses a continuing medical and social challenge both in the United States and all over the world. Despite a desire to quit drug use, return to drug use after a period of abstinence is a common problem among individuals dependent on psychostimulants. Recovery for psychostimulant drug-dependent individuals is particularly challenging because psychostimulant drugs induce significant changes in brain regions associated with cognitive functions leading to cognitive deficits. These cognitive deficits include impairments in learning/memory, poor decision making, and impaired control of behavioral output. Importantly, these drug-induced cognitive deficits often impact adherence to addiction treatment programs and predispose abstinent addicts to drug use relapse. Additionally, these cognitive deficits impact effective social and professional rehabilitation of abstinent addicts. The goal of this paper is to review neural substrates based on animal studies that could be pharmacologically targeted to reverse psychostimulant-induced cognitive deficits such as impulsivity and impairment in learning and memory. Further, the review will discuss neural substrates that could be used to facilitate extinction learning and thus reduce emotional and behavioral responses to drug-associated cues. Moreover, the review will discuss some non-pharmacological approaches that could be used either alone or in combination with pharmacological compounds to treat the above-mentioned cognitive deficits. Psychostimulant addiction treatment, which includes treatment for cognitive deficits, will help promote abstinence and allow for better rehabilitation and integration of abstinent individuals into society.
Collapse
Affiliation(s)
- Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, Ada, OH, United States
| |
Collapse
|
16
|
Moreira-Silva D, Carrettiero DC, Oliveira ASA, Rodrigues S, Dos Santos-Lopes J, Canas PM, Cunha RA, Almeida MC, Ferreira TL. Anandamide Effects in a Streptozotocin-Induced Alzheimer's Disease-Like Sporadic Dementia in Rats. Front Neurosci 2018; 12:653. [PMID: 30333717 PMCID: PMC6176656 DOI: 10.3389/fnins.2018.00653] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by multiple cognitive deficits including memory and sensorimotor gating impairments as a result of neuronal and synaptic loss. The endocannabinoid system plays an important role in these deficits but little is known about its influence on the molecular mechanism regarding phosphorylated tau (p-tau) protein accumulation - one of the hallmarks of AD -, and on the density of synaptic proteins. Thus, the aim of this study was to investigate the preventive effects of anandamide (N-arachidonoylethanolamine, AEA) on multiple cognitive deficits and on the levels of synaptic proteins (syntaxin 1, synaptophysin and synaptosomal-associated protein, SNAP-25), cannabinoid receptor type 1 (CB1) and molecules related to p-tau degradation machinery (heat shock protein 70, HSP70), and Bcl2-associated athanogene (BAG2) in an AD-like sporadic dementia model in rats using intracerebroventricular (icv) injection of streptozotocin (STZ). Our hypothesis is that AEA could interact with HSP70, modulating the level of p-tau and synaptic proteins, preventing STZ-induced cognitive impairments. Thirty days after receiving bilateral icv injections of AEA or STZ or both, the cognitive performance of adult male Wistar rats was evaluated in the object recognition test, by the escape latency in the elevated plus maze (EPM), by the tone and context fear conditioning as well as in prepulse inhibition tests. Subsequently, the animals were euthanized and their brains were removed for histological analysis or for protein quantification by Western Blotting. The behavioral results showed that STZ impaired recognition, plus maze and tone fear memories but did not affect contextual fear memory and prepulse inhibition. Moreover, AEA prevented recognition and non-associative emotional memory impairments induced by STZ, but did not influence tone fear conditioning. STZ increased the brain ventricular area and this enlargement was prevented by AEA. Additionally, STZ reduced the levels of p-tau (Ser199/202) and increased p-tau (Ser396), although AEA did not affect these alterations. HSP70 was found diminished only by STZ, while BAG2 levels were decreased by STZ and AEA. Synaptophysin, syntaxin and CB1 receptor levels were reduced by STZ, but only syntaxin was recovered by AEA. Altogether, albeit AEA failed to modify some AD-like neurochemical alterations, it partially prevented STZ-induced cognitive impairments, changes in synaptic markers and ventricle enlargement. This study showed, for the first time, that the administration of an endocannabinoid can prevent AD-like effects induced by STZ, boosting further investigations about the modulation of endocannabinoid levels as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Daniel Moreira-Silva
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Daniel C Carrettiero
- Center for Natural and Human Sciences, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Adriele S A Oliveira
- Center for Natural and Human Sciences, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Samanta Rodrigues
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Joyce Dos Santos-Lopes
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Paula M Canas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Rodrigo A Cunha
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Maria C Almeida
- Center for Natural and Human Sciences, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | - Tatiana L Ferreira
- Center for Mathematics, Computing and Cognition, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| |
Collapse
|
17
|
Zhang WJ, Cao WY, Huang YQ, Cui YH, Tu BX, Wang LF, Zou GJ, Liu Y, Hu ZL, Hu R, Li CQ, Xing XW, Li F. The Role of miR-150 in Stress-Induced Anxiety-Like Behavior in Mice. Neurotox Res 2018; 35:160-172. [PMID: 30120712 DOI: 10.1007/s12640-018-9943-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/28/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
Abstract
Stress plays a crucial role in several psychiatric disorders, including anxiety. However, the underlying mechanisms remain poorly understood. Here, we used acute stress (AS) and chronic restraint stress (CRS) models to develop anxiety-like behavior and investigate the role of miR-150 in the hippocampi of mice. Corticosterone levels as well as glutamate receptors in the hippocampus were evaluated. We found that anxiety-like behavior was induced after either AS or CRS, as determined by the open-field test (OFT) and elevated plus-maze test (EPM). Increased corticosterone levels were observed in the blood of AS and CRS groups, while the expression of miR-150 mRNA in the hippocampus was significantly decreased. The expressions of GluN2A, GluR1, GluR2, and V-Glut2 in the hippocampus were decreased after either AS or CRS. Hippocampal GAD67 expression was increased by AS but not CRS, and GluN2B expression was decreased by CRS but not AS. Adult miR-150 knockout mice showed anxiety-like behavior, as assessed by the OFT and EPM. The expressions of GluN2A, GluN2B, GluR1, and GluR2 were also downregulated, but the expression of V-Glut2 was upregulated in the hippocampi of miR-150 knockout mice compared with wild-type mice. Interestingly, we found that the miR-150 knockout mice showed decreased dendrite lengths, dendrite branchings, and numbers of dendrite spines in the hippocampus compared with wild-type mice. These results suggest that miR-150 may influence the synaptic plasticity of the hippocampus and play a significant role in stress-induced anxiety-like behavior in adult mice.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Wen-Yu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Yan-Qing Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Yan-Hui Cui
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Bo-Xuan Tu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Lai-Fa Wang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Guang-Jing Zou
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Yu Liu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Zhao-Lan Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Rong Hu
- Department of Pain, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chang-Qi Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China
| | - Xiao-Wei Xing
- Center for Medical Experiments, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Fang Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, Hunan, China.
| |
Collapse
|
18
|
Saraulli D, Costanzi M, Mastrorilli V, Farioli-Vecchioli S. The Long Run: Neuroprotective Effects of Physical Exercise on Adult Neurogenesis from Youth to Old Age. Curr Neuropharmacol 2018; 15:519-533. [PMID: 27000776 PMCID: PMC5543673 DOI: 10.2174/1570159x14666160412150223] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 03/08/2016] [Accepted: 03/16/2016] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The rapid lengthening of life expectancy has raised the problem of providing social programs to counteract the age-related cognitive decline in a growing number of older people. Physical activity stands among the most promising interventions aimed at brain wellbeing, because of its effective neuroprotective action and low social cost. The purpose of this review is to describe the neuroprotective role exerted by physical activity in different life stages. In particular, we focus on adult neurogenesis, a process which has proved being highly responsive to physical exercise and may represent a major factor of brain health over the lifespan. METHODS The most recent literature related to the subject has been reviewed. The text has been divided into three main sections, addressing the effects of physical exercise during childhood/ adolescence, adulthood and aging, respectively. For each one, the most relevant studies, carried out on both human participants and rodent models, have been described. RESULTS The data reviewed converge in indicating that physical activity exerts a positive effect on brain functioning throughout the lifespan. However, uncertainty remains about the magnitude of the effect and its biological underpinnings. Cellular and synaptic plasticity provided by adult neurogenesis are highly probable mediators, but the mechanism for their action has yet to be conclusively established. CONCLUSION Despite alternative mechanisms of action are currently debated, age-appropriate physical activity programs may constitute a large-scale, relatively inexpensive and powerful approach to dampen the individual and social impact of age-related cognitive decline.
Collapse
Affiliation(s)
- Daniele Saraulli
- Institute of Cell Biology and Neurobiology, National Research Council, & Fondazione S. Lucia, Rome. Italy
| | - Marco Costanzi
- Department of Human Sciences, LUMSA University, Rome. Italy
| | - Valentina Mastrorilli
- Institute of Cell Biology and Neurobiology, National Research Council, & Fondazione S. Lucia, Rome. Italy
| | - Stefano Farioli-Vecchioli
- Institute of Cell Biology and Neurobiology, National Research Council, Via del Fosso di Fiorano 64, 00143 Rome. Italy
| |
Collapse
|
19
|
Florenzano F, Veronica C, Ciasca G, Ciotti MT, Pittaluga A, Olivero G, Feligioni M, Iannuzzi F, Latina V, Maria Sciacca MF, Sinopoli A, Milardi D, Pappalardo G, Marco DS, Papi M, Atlante A, Bobba A, Borreca A, Calissano P, Amadoro G. Extracellular truncated tau causes early presynaptic dysfunction associated with Alzheimer's disease and other tauopathies. Oncotarget 2017; 8:64745-64778. [PMID: 29029390 PMCID: PMC5630290 DOI: 10.18632/oncotarget.17371] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/11/2017] [Indexed: 12/14/2022] Open
Abstract
The largest part of tau secreted from AD nerve terminals and released in cerebral spinal fluid (CSF) is C-terminally truncated, soluble and unaggregated supporting potential extracellular role(s) of NH2 -derived fragments of protein on synaptic dysfunction underlying neurodegenerative tauopathies, including Alzheimer's disease (AD). Here we show that sub-toxic doses of extracellular-applied human NH2 tau 26-44 (aka NH 2 htau) -which is the minimal active moiety of neurotoxic 20-22kDa peptide accumulating in vivo at AD synapses and secreted into parenchyma- acutely provokes presynaptic deficit in K+ -evoked glutamate release on hippocampal synaptosomes along with alteration in local Ca2+ dynamics. Neuritic dystrophy, microtubules breakdown, deregulation in presynaptic proteins and loss of mitochondria located at nerve endings are detected in hippocampal cultures only after prolonged exposure to NH 2 htau. The specificity of these biological effects is supported by the lack of any significant change, either on neuronal activity or on cellular integrity, shown by administration of its reverse sequence counterpart which behaves as an inactive control, likely due to a poor conformational flexibility which makes it unable to dynamically perturb biomembrane-like environments. Our results demonstrate that one of the AD-relevant, soluble and secreted N-terminally truncated tau forms can early contribute to pathology outside of neurons causing alterations in synaptic activity at presynaptic level, independently of overt neurodegeneration.
Collapse
Affiliation(s)
| | | | - Gabriele Ciasca
- Institute of Physics, Catholic University of the Sacred Heart, Largo F Vito 1, Rome, Italy
| | - Maria Teresa Ciotti
- Institute of Cellular Biology and Neuroscience, CNR, IRCSS Santa Lucia Foundation, Rome, Italy
| | - Anna Pittaluga
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Viale Cembrano, Italy
| | - Gunedalina Olivero
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Viale Cembrano, Italy
| | - Marco Feligioni
- European Brain Research Institute, Rome, Italy
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | | | | | | | | | - Danilo Milardi
- Institute of Biostructures and Bioimaging, CNR, Catania, Italy
| | | | - De Spirito Marco
- Institute of Physics, Catholic University of the Sacred Heart, Largo F Vito 1, Rome, Italy
| | - Massimiliano Papi
- Institute of Physics, Catholic University of the Sacred Heart, Largo F Vito 1, Rome, Italy
| | - Anna Atlante
- Institute of Biomembranes and Bioenergetics, CNR, Bari, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Viale Benedetto XV, Italy
| | - Antonella Bobba
- Institute of Biomembranes and Bioenergetics, CNR, Bari, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Viale Benedetto XV, Italy
| | - Antonella Borreca
- Institute of Cellular Biology and Neuroscience, CNR, IRCSS Santa Lucia Foundation, Rome, Italy
| | | | - Giuseppina Amadoro
- European Brain Research Institute, Rome, Italy
- Institute of Translational Pharmacology, CNR, Rome, Italy
| |
Collapse
|
20
|
Li X, Wang Z, Tan L, Wang Y, Lu C, Chen R, Zhang S, Gao Y, Liu Y, Yin Y, Liu X, Liu E, Yang Y, Hu Y, Xu Z, Xu F, Wang J, Liu GP, Wang JZ. Correcting miR92a-vGAT-Mediated GABAergic Dysfunctions Rescues Human Tau-Induced Anxiety in Mice. Mol Ther 2017; 25:140-152. [PMID: 28129110 DOI: 10.1016/j.ymthe.2016.10.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 01/30/2023] Open
Abstract
Patients with Alzheimer's disease (AD) commonly show anxiety behaviors, but the molecular mechanisms are not clear and no efficient intervention exists. Here, we found that overexpression of human wild-type, full-length tau (termed htau) in hippocampus significantly decreased the extracellular γ-aminobutyric acid (GABA) level with inhibition of γ oscillation and the evoked inhibitory postsynaptic potential (eIPSP). With tau accumulation, the mice show age-dependent anxiety behaviors. Among the factors responsible for GABA synthesis, release, uptake, and transport, we found that accumulation of htau selectively suppressed expression of the intracellular vesicular GABA transporter (vGAT). Tau accumulation increased miR92a, which targeted vGAT mRNA 3' UTR and inhibited vGAT translation. Importantly, we found that upregulating GABA tones by intraperitoneal injection of midazolam (a GABA agonist), ChR2-mediated photostimulating and overexpressing vGAT, or blocking miR92a by using specific antagomir or inhibitor efficiently rescued the htau-induced GABAergic dysfunctions with attenuation of anxiety. Finally, we also demonstrated that vGAT level decreased while the miR92a increased in the AD brains. These findings demonstrate that the AD-like tau accumulation induces anxiety through disrupting miR92a-vGAT-GABA signaling, which reveals molecular mechanisms underlying the anxiety behavior in AD patients and potentially leads to the development of new therapeutics for tauopathies.
Collapse
Affiliation(s)
- Xiaoguang Li
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhihao Wang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lu Tan
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yali Wang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Physiology Department, Henan Provincial Key Laboratory for Brain Research, Xinxiang Medical University, Xinxiang 453000, China
| | - Chengbiao Lu
- Physiology Department, Henan Provincial Key Laboratory for Brain Research, Xinxiang Medical University, Xinxiang 453000, China
| | - Rongxiang Chen
- State Key Laboratory for Magnet Resonance and Atom and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academia of Science, Wuhan 430071, China
| | - Shujuan Zhang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuan Gao
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanchao Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yaling Yin
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinghua Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Enjie Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Hu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhipeng Xu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fuqiang Xu
- State Key Laboratory for Magnet Resonance and Atom and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academia of Science, Wuhan 430071, China
| | - Jie Wang
- State Key Laboratory for Magnet Resonance and Atom and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academia of Science, Wuhan 430071, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine and Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
21
|
Lam CTW, Gong AGW, Lam KYC, Zhang LM, Chen JP, Dong TTX, Lin HQ, Tsim KWK. Jujube-containing herbal decoctions induce neuronal differentiation and the expression of anti-oxidant enzymes in cultured PC12 cells. JOURNAL OF ETHNOPHARMACOLOGY 2016; 188:275-283. [PMID: 27178633 DOI: 10.1016/j.jep.2016.05.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The fruit of Ziziphus jujuba (Mill.), known as Jujuba Fructus (JF) or jujube, is a well-known Traditional Chinese Medicine (TCM) for blood nourishment and sedation effect. Apart from prescribing as single herb alone, JF is very often being included in multi-herbal decoctions to prolong, enhance and harmonize pharmaceutical effects of decoctions while at the same time reducing toxicity. Here, we aimed to compare the protective and differentiating activities of three chemically standardized jujube-containing decoctions, including Guizhi Tang (GZT), Neibu Dangguijianzhong Tang (NDT) and ZaoTang (ZOT) in cultured PC12 cells. MATERIALS AND METHODS The protein expressions of neurofilaments, including NF68, NF160 and NF200, under the herbal treatment were revealed by western blot. The determination of neurite outgrowth in cultured PC12 cells upon the treatment of herbal extracts was performed by light microscope equipped with a phase-contrast condenser and SPOT imaging software. The protective effect against tBHP-induced cytotoxicity under the herbal treatment was measured by MTT assay. A luciferase reporter construct carrying four repeats of anti-oxidant response element (ARE) and a downstream luciferase reporter gene luc2P was transfected into PC12 cells to study the transcriptional activation of ARE. The mRNA expression of antioxidant enzymes under the herbal treatment was analyzed by quantitative real-time PCR. RESULTS These jujube-containing decoctions processed similar neuro-protective and brain beneficial properties. The herbal treatment induced the protein expression of neurofilaments. Neurite outgrowth was observed under the herbal treatment. In parallel, the pre-treatment of herbal extracts protected PC 12 cells against oxidative stress-induced apoptosis in a dose-dependent manner. Moreover, the herbal treatments triggered the mRNA expressions of relevant anti-oxidation genes, i.e. glutamate-cysteine ligase catalytic subunit (GCLC), glutamate-cysteine ligase modulatory subunit (GCLM), glutathione S-transferase (GST) and NAD(P)H quinone oxidoreductase (NQO1) via the activation of anti-oxidant response element (ARE). CONCLUSION The results therefore demonstrated neuro-protective and differentiating properties of the three closely related decoctions, and which subsequently illustrated the enhancement function of jujube within a multi-herbal decoction.
Collapse
Affiliation(s)
- Candy T W Lam
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Amy G W Gong
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kelly Y C Lam
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Laura M Zhang
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jian-Ping Chen
- Pharmaceutical Department, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Tina T X Dong
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Huang-Quan Lin
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Karl W K Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
22
|
Ryan SM, Kelly ÁM. Exercise as a pro-cognitive, pro-neurogenic and anti-inflammatory intervention in transgenic mouse models of Alzheimer's disease. Ageing Res Rev 2016; 27:77-92. [PMID: 27039886 DOI: 10.1016/j.arr.2016.03.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/22/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022]
Abstract
It is now well established, at least in animal models, that exercise elicits potent pro-cognitive and pro-neurogenic effects. Alzheimer's disease (AD) is one of the leading causes of dementia and represents one of the greatest burdens on healthcare systems worldwide, with no effective treatment for the disease to date. Exercise presents a promising non-pharmacological option to potentially delay the onset of or slow down the progression of AD. Exercise interventions in mouse models of AD have been explored and have been found to reduce amyloid pathology and improve cognitive function. More recent studies have expanded the research question by investigating potential pro-neurogenic and anti-inflammatory effects of exercise. In this review we summarise studies that have examined exercise-mediated effects on AD pathology, cognitive function, hippocampal neurogenesis and neuroinflammation in transgenic mouse models of AD. Furthermore, we attempt to identify the optimum exercise conditions required to elicit the greatest benefits, taking into account age and pathology of the model, as well as type and duration of exercise.
Collapse
|
23
|
Caspase-cleaved tau exhibits rapid memory impairment associated with tau oligomers in a transgenic mouse model. Neurobiol Dis 2016; 87:19-28. [DOI: 10.1016/j.nbd.2015.12.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/30/2015] [Accepted: 12/14/2015] [Indexed: 12/23/2022] Open
|
24
|
Conditional Inhibition of Adult Neurogenesis by Inducible and Targeted Deletion of ERK5 MAP Kinase Is Not Associated with Anxiety/Depression-Like Behaviors. eNeuro 2015; 2:eN-NWR-0014-14. [PMID: 26464972 PMCID: PMC4596085 DOI: 10.1523/eneuro.0014-14.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/31/2015] [Accepted: 03/23/2015] [Indexed: 12/15/2022] Open
Abstract
Although there is evidence that adult neurogenesis contributes to the therapeutic efficacy of chronic antidepressant treatment for anxiety and depression disorders, the role of adult neurogenesis in the onset of depression-related symptoms is still open to question. To address this issue, we utilized a transgenic mouse strain in which adult neurogenesis was specifically and conditionally impaired by Nestin-CreER-driven, inducible knockout (icKO) of erk5 MAP kinase in Nestin-expressing neural progenitors of the adult mouse brain upon tamoxifen administration. Here, we report that inhibition of adult neurogenesis by this mechanism is not associated with an increase of the baseline anxiety or depression in non-stressed animals, nor does it increase the animal's susceptibility to depression after chronic unpredictable stress treatment. Our findings indicate that impaired adult neurogenesis does not lead to anxiety or depression.
Collapse
|
25
|
Luo Y, Zhou J, Li M, Wu P, Hu Z, Ni L, Jin Y, Chen J, Wang F. Reversal of aging-related emotional memory deficits by norepinephrine via regulating the stability of surface AMPA receptors. Aging Cell 2015; 14:170-9. [PMID: 25564942 PMCID: PMC4364829 DOI: 10.1111/acel.12282] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2014] [Indexed: 12/19/2022] Open
Abstract
Aging-related emotional memory deficit is a well-known complication in Alzheimer's disease and normal aging. However, little is known about its molecular mechanism. To address this issue, we examined the role of norepinephrine (NE) and its relevant drug desipramine in the regulation of hippocampal long-term potentiation (LTP), surface expression of AMPA receptor, and associative fear memory in rats. We found that there was a defective regulation of NE content and AMPA receptor trafficking during fear conditioning, which were accompanied by impaired emotional memory and LTP in aged rats. Furthermore, we also found that the exogenous upregulation of NE ameliorated the impairment of LTP and emotional memory via enhancing AMPA receptor trafficking in aged rats, and the downregulation of NE impaired LTP in adult rats. Finally, acute treatment with NE or desipramine rescued the impaired emotional memory in aged rats. These results imply a pivotal role for NE in synaptic plasticity and associative fear memory in aging rats and suggest that desipramine is a potential candidate for treating aging-related emotional memory deficit.
Collapse
Affiliation(s)
- Yi Luo
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
| | - Jun Zhou
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
| | - Ming‐Xing Li
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
| | - Peng‐Fei Wu
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province Wuhan 430030 China
| | - Zhuang‐Li Hu
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province Wuhan 430030 China
| | - Lan Ni
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province Wuhan 430030 China
| | - You Jin
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province Wuhan 430030 China
| | - Jian‐Guo Chen
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province Wuhan 430030 China
- The Institute of Brain Research Huazhong University of Science and Technology Wuhan 430030 China
| | - Fang Wang
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
- Key Laboratory of Neurological Diseases (HUST) Ministry of Education of China Wuhan 430030 China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province Wuhan 430030 China
- The Institute of Brain Research Huazhong University of Science and Technology Wuhan 430030 China
| |
Collapse
|
26
|
Li L, Xu ZP, Liu GP, Xu C, Wang ZH, Li XG, Liu EJ, Zeng J, Chai DM, Yao WL, Wang JZ. Expression of 1N3R-Tau isoform inhibits cell proliferation by inducing S phase arrest in N2a cells. PLoS One 2015; 10:e0119865. [PMID: 25822823 PMCID: PMC4378987 DOI: 10.1371/journal.pone.0119865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/16/2015] [Indexed: 12/17/2022] Open
Abstract
Tau is a microtubule-associated protein implicated in neurodegenerative tauopathies. Six tau isoforms are generated from a single gene through alternative splicing of exons 2, 3 and 10 in human brain. Differential expression of tau isoforms has been detected in different brain areas, during neurodevelopment and in neurodegenerative disorders. However, the biological significance of different tau isoforms is not clear. Here, we investigated the individual effect of six different isoforms of tau on cell proliferation and the possible mechanisms by transient expression of eGFP-labeled tau isoform plasmid in N2a cells. Our study showed the transfection efficiency was comparable between different isoforms of tau by examining GFP expression. Compared with other isoforms, we found expression of 1N3R-tau significantly inhibited cell proliferation by Cell Counting Kit-8 assay and BrdU incorporation. Flow cytometry analysis further showed expression of 1N3R-tau induced S phase arrest. Compared with the longest isoform of tau, expression of 1N3R-tau induced cyclin E translocation from the nuclei to cytoplasm, while it did not change the level of cell cycle checkpoint proteins. These data indicate that 1N3R-tau inhibits cell proliferation through inducing S phase arrest.
Collapse
Affiliation(s)
- Li Li
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Peng Xu
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong-Ping Liu
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Xu
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Hao Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Guang Li
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - En-Jie Liu
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Zeng
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Da-Min Chai
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Long Yao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- * E-mail:
| |
Collapse
|
27
|
Corsetti V, Florenzano F, Atlante A, Bobba A, Ciotti MT, Natale F, Della Valle F, Borreca A, Manca A, Meli G, Ferraina C, Feligioni M, D'Aguanno S, Bussani R, Ammassari-Teule M, Nicolin V, Calissano P, Amadoro G. NH2-truncated human tau induces deregulated mitophagy in neurons by aberrant recruitment of Parkin and UCHL-1: implications in Alzheimer's disease. Hum Mol Genet 2015; 24:3058-81. [PMID: 25687137 DOI: 10.1093/hmg/ddv059] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/10/2015] [Indexed: 01/26/2023] Open
Abstract
Disarrangement in functions and quality control of mitochondria at synapses are early events in Alzheimer's disease (AD) pathobiology. We reported that a 20-22 kDa NH2-tau fragment mapping between 26 and 230 amino acids of the longest human tau isoform (aka NH2htau): (i) is detectable in cellular and animal AD models, as well in synaptic mitochondria and cerebrospinal fluids (CSF) from human AD subjects; (ii) is neurotoxic in primary hippocampal neurons; (iii) compromises the mitochondrial biology both directly, by inhibiting the ANT-1-dependent ADP/ATP exchange, and indirectly, by impairing their selective autophagic clearance (mitophagy). Here, we show that the extensive Parkin-dependent turnover of mitochondria occurring in NH2htau-expressing post-mitotic neurons plays a pro-death role and that UCHL-1, the cytosolic Ubiquitin-C-terminal hydrolase L1 which directs the physiological remodeling of synapses by controlling ubiquitin homeostasis, critically contributes to mitochondrial and synaptic failure in this in vitro AD model. Pharmacological or genetic suppression of improper mitophagy, either by inhibition of mitochondrial targeting to autophagosomes or by shRNA-mediated silencing of Parkin or UCHL-1 gene expression, restores synaptic and mitochondrial content providing partial but significant protection against the NH2htau-induced neuronal death. Moreover, in mitochondria from human AD synapses, the endogenous NH2htau is stably associated with Parkin and with UCHL-1. Taken together, our studies show a causative link between the excessive mitochondrial turnover and the NH2htau-induced in vitro neuronal death, suggesting that pathogenetic tau truncation may contribute to synaptic deterioration in AD by aberrant recruitment of Parkin and UCHL-1 to mitochondria making them more prone to detrimental autophagic clearance.
Collapse
Affiliation(s)
- V Corsetti
- Institute of Translational Pharmacology (IFT) - National Research Council (CNR), Via Fosso del Cavaliere 100-00133, Rome, Italy
| | - F Florenzano
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - A Atlante
- Institute of Biomembranes and Bioenergetics (IBBE)-CNR, Via Amendola 165/A, 70126 Bari, Italy
| | - A Bobba
- Institute of Biomembranes and Bioenergetics (IBBE)-CNR, Via Amendola 165/A, 70126 Bari, Italy
| | - M T Ciotti
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - F Natale
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - F Della Valle
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - A Borreca
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - A Manca
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - G Meli
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - C Ferraina
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - M Feligioni
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - S D'Aguanno
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - R Bussani
- UCO Pathological Anatomy and Histopathology Unit, Cattinara Hospital Strada di Fiume 447, 34149 Trieste, Italy and
| | - M Ammassari-Teule
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - V Nicolin
- Department of Medical, Surgical and Health Science, University of Trieste, Strada di Fiume 449, 34149 Trieste, Italy
| | - P Calissano
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - G Amadoro
- Institute of Translational Pharmacology (IFT) - National Research Council (CNR), Via Fosso del Cavaliere 100-00133, Rome, Italy European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| |
Collapse
|
28
|
Zhao Y, Gong CX. From chronic cerebral hypoperfusion to Alzheimer-like brain pathology and neurodegeneration. Cell Mol Neurobiol 2015; 35:101-10. [PMID: 25352419 PMCID: PMC11486181 DOI: 10.1007/s10571-014-0127-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/19/2014] [Indexed: 01/09/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) is a common consequence of various cerebral vascular disorders and hemodynamic and blood changes. Recent studies have revealed an important role of CCH in neurodegeneration and dementia, including vascular dementia and Alzheimer's disease (AD). This article reviews the recent advances in understanding CCH-induced neurodegeneration and AD-related brain pathology and cognitive impairment. We discuss the causes and assessment of CCH, the possible mechanisms by which CCH promotes Alzheimer-like pathology and neurodegeneration, and animal models of CCH. It appears that CCH promotes neurodegeneration and AD through multiple mechanisms, including induction of oxidative stress, Aβ accumulation and aggravation, tau hyperphosphorylation, synaptic dysfunction, neuronal loss, white matter lesion, and neuroinflammation. Better understanding of the mechanisms of CCH will help develop therapeutic strategies for preventing and treating neurodegeneration, including sporadic AD and vascular dementia, caused by CCH.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Neurology, The First Hospital of Jilin University, Xinmin Street, Changchun, 130021, Jilin, China,
| | | |
Collapse
|
29
|
Cognitive Reserve and Alzheimer’s Disease. Mol Neurobiol 2014; 51:187-208. [DOI: 10.1007/s12035-014-8720-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 04/17/2014] [Indexed: 12/13/2022]
|
30
|
Zhao Y, Gu JH, Dai CL, Liu Q, Iqbal K, Liu F, Gong CX. Chronic cerebral hypoperfusion causes decrease of O-GlcNAcylation, hyperphosphorylation of tau and behavioral deficits in mice. Front Aging Neurosci 2014; 6:10. [PMID: 24575038 PMCID: PMC3918671 DOI: 10.3389/fnagi.2014.00010] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 01/16/2014] [Indexed: 11/13/2022] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is one of the causes of vascular dementia (VaD) and is also an etiological factor for Alzheimer’s disease (AD). However, how CCH causes cognitive impairment and contributes to Alzheimer’s pathology is poorly understood. Here we produced a mouse model of CCH by unilateral common carotid artery occlusion (UCCAO) and studied the behavioral changes and brain abnormalities in mice 2.5 months after UCCAO. We found that CCH caused significant short-term memory deficits and mild long-term spatial memory impairment, as well as decreased level of protein O-GlcNAcylation, increased level of tau phosphorylation, dysregulated synaptic proteins and insulin signaling, and selective neurodegeneration in the brain. These findings provide mechanistic insight into the effects of CCH on memory and cognition and the likely link between AD and VaD.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun Jilin, China ; Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities Staten Island, NY, USA
| | - Jin-Hua Gu
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities Staten Island, NY, USA ; Department of Pharmacology, Medical School, Nantong University Nantong, Jiangsu, China
| | - Chun-Ling Dai
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities Staten Island, NY, USA
| | - Qun Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun Jilin, China
| | - Khalid Iqbal
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities Staten Island, NY, USA
| | - Fei Liu
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities Staten Island, NY, USA
| | - Cheng-Xin Gong
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities Staten Island, NY, USA
| |
Collapse
|