1
|
Li D, Jia J, Zeng H, Zhong X, Chen H, Yi C. Efficacy of exercise rehabilitation for managing patients with Alzheimer's disease. Neural Regen Res 2024; 19:2175-2188. [PMID: 38488551 PMCID: PMC11034587 DOI: 10.4103/1673-5374.391308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/16/2023] [Accepted: 11/25/2023] [Indexed: 04/24/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and degenerative neurological disease characterized by the deterioration of cognitive functions. While a definitive cure and optimal medication to impede disease progression are currently unavailable, a plethora of studies have highlighted the potential advantages of exercise rehabilitation for managing this condition. Those studies show that exercise rehabilitation can enhance cognitive function and improve the quality of life for individuals affected by AD. Therefore, exercise rehabilitation has been regarded as one of the most important strategies for managing patients with AD. Herein, we provide a comprehensive analysis of the currently available findings on exercise rehabilitation in patients with AD, with a focus on the exercise types which have shown efficacy when implemented alone or combined with other treatment methods, as well as the potential mechanisms underlying these positive effects. Specifically, we explain how exercise may improve the brain microenvironment and neuronal plasticity. In conclusion, exercise is a cost-effective intervention to enhance cognitive performance and improve quality of life in patients with mild to moderate cognitive dysfunction. Therefore, it can potentially become both a physical activity and a tailored intervention. This review may aid the development of more effective and individualized treatment strategies to address the challenges imposed by this debilitating disease, especially in low- and middle-income countries.
Collapse
Affiliation(s)
- Dan Li
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Jinning Jia
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Haibo Zeng
- Department of Pathology, Huichang County People’s Hospital, Ganzhou, Jiangxi Province, China
| | - Xiaoyan Zhong
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Chenju Yi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Dhungana A, Becchi S, Leake J, Morris G, Avgan N, Balleine BW, Vissel B, Bradfield LA. Goal-Directed Action Is Initially Impaired in a hAPP-J20 Mouse Model of Alzheimer's Disease. eNeuro 2023; 10:ENEURO.0363-22.2023. [PMID: 36650070 PMCID: PMC9927544 DOI: 10.1523/eneuro.0363-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Cognitive-behavioral testing in preclinical models of Alzheimer's disease has failed to capture deficits in goal-directed action control. Here, we provide the first comprehensive investigation of goal-directed action in a transgenic mouse model of Alzheimer's disease. Specifically, we tested outcome devaluation performance in male and female human amyloid precursor protein (hAPP)-J20 mice. Mice were first trained to press left and right levers for pellet and sucrose outcomes, respectively (counterbalanced), over 4 d. On test, mice were prefed one of the outcomes to satiety and given a choice between levers. Devaluation performance was intact for 36-week-old wild-types of both sexes, who responded more on the valued relative to the devalued lever (Valued > Devalued). By contrast, devaluation was impaired (Valued = Devalued) for J20 mice of both sexes, and for 52-week-old male mice regardless of genotype. After additional lever press training (i.e., 8-d lever pressing in total), devaluation was intact for all mice, demonstrating that the initial deficits were not a result of a nonspecific impairment in reward processing, depression, or locomotor activity in J20 or aging mice. Follow-up analyses revealed that microglial expression in the dorsal CA1 region of the hippocampus was associated with poorer outcome devaluation performance on initial, but not later tests. Together, these data demonstrate that goal-directed action is initially impaired in J20 mice of both sexes and in aging male mice regardless of genotype, and that this impairment is related to neuroinflammation in the dorsal CA1 hippocampal region.
Collapse
Affiliation(s)
- Amolika Dhungana
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia
| | - Serena Becchi
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Jessica Leake
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia
- School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Gary Morris
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Nesli Avgan
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia
| | - Bernard W Balleine
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia
- School of Clinical Medicine, University of New South Wales Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, New South Wales 2052, Australia
| | - Laura A Bradfield
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Centre for Neuroscience and Regenerative Medicine, St. Vincent's Centre for Applied Medical Research, St. Vincent's Health Network, Sydney, New South Wales 2010, Australia
| |
Collapse
|
3
|
Dysfunction of the glutamatergic photoreceptor synapse in the P301S mouse model of tauopathy. Acta Neuropathol Commun 2023; 11:5. [PMID: 36631898 PMCID: PMC9832799 DOI: 10.1186/s40478-022-01489-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/04/2022] [Indexed: 01/13/2023] Open
Abstract
Tauopathies, including Alzheimer's disease, are characterized by retinal ganglion cell loss associated with amyloid and phosphorylated tau deposits. We investigated the functional impact of these histopathological alterations in the murine P301S model of tauopathy. Visual impairments were demonstrated by a decrease in visual acuity already detectable at 6 months, the onset of disease. Visual signals to the cortex and retina were delayed at 6 and 9 months, respectively. Surprisingly, the retinal output signal was delayed at the light onset and advanced at the light offset. This antagonistic effect, due to a dysfunction of the cone photoreceptor synapse, was associated with changes in the expression of the vesicular glutamate transporter and a microglial reaction. This dysfunction of retinal glutamatergic synapses suggests a novel interpretation for visual deficits in tauopathies and it highlights the potential value of the retina for the diagnostic assessment and the evaluation of therapies in Alzheimer's disease and other tauopathies.
Collapse
|
4
|
Lansdell TA, Xu H, Galligan JJ, Dorrance AM. Effects of Striatal Amyloidosis on the Dopaminergic System and Behavior: A Comparative Study in Male and Female 5XFAD Mice. J Alzheimers Dis 2023; 94:1361-1375. [PMID: 37424461 DOI: 10.3233/jad-220905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
BACKGROUND Nearly two-thirds of patients diagnosed with Alzheimer's disease (AD) are female. In addition, female patients with AD have more significant cognitive impairment than males at the same disease stage. This disparity suggests there are sex differences in AD progression. While females appear to be more affected by AD, most published behavioral studies utilize male mice. In humans, there is an association between antecedent attention-deficit/hyperactivity disorder and increased risk of dementia. Functional connectivity studies indicate that dysfunctional cortico-striatal networks contribute to hyperactivity in attention deficit hyperactivity disorder. Higher plaque density in the striatum accurately predicts the presence of clinical AD pathology. In addition, there is a link between AD-related memory dysfunction and dysfunctional dopamine signaling. OBJECTIVE With the need to consider sex as a biological variable, we investigated the influence of sex on striatal plaque burden, dopaminergic signaling, and behavior in prodromal 5XFAD mice. METHODS Six-month-old male and female 5XFAD and C57BL/6J mice were evaluated for striatal amyloid plaque burden, locomotive behavior, and changes in dopaminergic machinery in the striatum. RESULTS 5XFAD female mice had a higher striatal amyloid plaque burden than male 5XFAD mice. 5XFAD females, but not males, were hyperactive. Hyperactivity in female 5XFAD mice was associated with increased striatal plaque burden and changes in dopamine signaling in the dorsal striatum. CONCLUSION Our results indicate that the progression of amyloidosis involves the striatum in females to a greater extent than in males. These studies have significant implications for using male-only cohorts in the study of AD progression.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Hui Xu
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
5
|
Cheng KC, Hwang YL, Chiang HC. The double-edged sword effect of HDAC6 in Aβ toxicities. FASEB J 2021; 36:e22072. [PMID: 34907598 DOI: 10.1096/fj.202101061r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/25/2021] [Accepted: 11/12/2021] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease (AD) is marked by cognitive impairment, massive cell death, and reduced life expectancy. Pathologically, accumulated beta-amyloid (Aβ) aggregates and hyperphosphorylated tau protein is the hallmark of the disease. Although changes in cellular function and protein accumulates have been demonstrated in many different AD animal models, the molecular mechanism involved in different cellular functions and the correlation and causative relation between different protein accumulations remain elusive. Our in vivo genetic studies revealed that the molecular mechanisms involved in memory loss and lifespan shortening are different and that tau plays an essential role in mediating Aβ-induced early death. We found that when the first deacetylase (DAC) domain of histone deacetylase 6 (HDAC6) activity was increased, it regulated cortactin deacetylation to reverse Aβ-induced learning and memory deficit, but with no effect on the lifespan of the Aβ flies. On the other hand, an increased amount of the second DAC domain of HDAC6 promoted tau phosphorylation to facilitate Aβ-induced lifespan shortening without affecting learning performance in the Aβ flies. Our data also confirmed decreased acetylation in two major HDAC6 downstream proteins, suggesting increased HDAC6 activity in Aβ flies. Our data established the double-edged sword effect of HDAC6 activity in Aβ-induced pathologies. Not only did we segregate memory loss and lifespan shortening in Aβ flies, but we also provided evidence to link the Aβ with tau signaling.
Collapse
Affiliation(s)
- Kuan-Chung Cheng
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Luen Hwang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsueh-Cheng Chiang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
6
|
Ni R. Magnetic Resonance Imaging in Animal Models of Alzheimer's Disease Amyloidosis. Int J Mol Sci 2021; 22:12768. [PMID: 34884573 PMCID: PMC8657987 DOI: 10.3390/ijms222312768] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid-beta (Aβ) plays an important role in the pathogenesis of Alzheimer's disease. Aberrant Aβ accumulation induces neuroinflammation, cerebrovascular alterations, and synaptic deficits, leading to cognitive impairment. Animal models recapitulating the Aβ pathology, such as transgenic, knock-in mouse and rat models, have facilitated the understanding of disease mechanisms and the development of therapeutics targeting Aβ. There is a rapid advance in high-field MRI in small animals. Versatile high-field magnetic resonance imaging (MRI) sequences, such as diffusion tensor imaging, arterial spin labeling, resting-state functional MRI, anatomical MRI, and MR spectroscopy, as well as contrast agents, have been developed for preclinical imaging in animal models. These tools have enabled high-resolution in vivo structural, functional, and molecular readouts with a whole-brain field of view. MRI has been used to visualize non-invasively the Aβ deposits, synaptic deficits, regional brain atrophy, impairment in white matter integrity, functional connectivity, and cerebrovascular and glymphatic system in animal models of Alzheimer's disease amyloidosis. Many of the readouts are translational toward clinical MRI applications in patients with Alzheimer's disease. In this review, we summarize the recent advances in MRI for visualizing the pathophysiology in amyloidosis animal models. We discuss the outstanding challenges in brain imaging using MRI in small animals and propose future outlook in visualizing Aβ-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
7
|
BACE inhibitor treatment of mice induces hyperactivity in a Seizure-related gene 6 family dependent manner without altering learning and memory. Sci Rep 2021; 11:15084. [PMID: 34302009 PMCID: PMC8302682 DOI: 10.1038/s41598-021-94369-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/07/2021] [Indexed: 01/15/2023] Open
Abstract
BACE inhibitors, which decrease BACE1 (β-secretase 1) cleavage of the amyloid precursor protein, are a potential treatment for Alzheimer’s disease. Clinical trials using BACE inhibitors have reported a lack of positive effect on patient symptoms and, in some cases, have led to increased adverse events, cognitive worsening and hippocampal atrophy. A potential drawback of this strategy is the effect of BACE inhibition on other BACE1 substrates such as Seizure-related gene 6 (Sez6) family proteins which are known to have a role in neuronal function. Mice were treated with an in-diet BACE inhibitor for 4–8 weeks to achieve a clinically-relevant level of amyloid-β40 reduction in the brain. Mice underwent behavioural testing and postmortem analysis of dendritic spine number and morphology with Golgi-Cox staining. Sez6 family triple knockout mice were tested alongside wild-type mice to identify whether any effects of the treatment were due to altered cleavage of Sez6 family proteins. Wild-type mice treated with BACE inhibitor displayed hyperactivity on the elevated open field, as indicated by greater distance travelled, but this effect was not observed in treated Sez6 triple knockout mice. BACE inhibitor treatment did not lead to significant changes in spatial or fear learning, reference memory, cognitive flexibility or anxiety in mice as assessed by the Morris water maze, context fear conditioning, or light–dark box tests. Chronic BACE inhibitor treatment reduced the density of mushroom-type spines in the somatosensory cortex, regardless of genotype, but did not affect steady-state dendritic spine density or morphology in the CA1 region of the hippocampus. Chronic BACE inhibition for 1–2 months in mice led to increased locomotor output but did not alter memory or cognitive flexibility. While the mechanism underlying the treatment-induced hyperactivity is unknown, the absence of this response in Sez6 triple knockout mice indicates that blocking ectodomain shedding of Sez6 family proteins is a contributing factor. In contrast, the decrease in mature spine density in cortical neurons was not attributable to lack of shed Sez6 family protein ectodomains. Therefore, other BACE1 substrates are implicated in this effect and, potentially, in the cognitive decline in longer-term chronically treated patients.
Collapse
|
8
|
Ma DL, Luo Y, Huang R, Zhao ZR, Zhang L, Li YL, Wang Q, Li L, Zhang L. Cornel Iridoid Glycoside Suppresses Hyperactivity Phenotype in rTg4510 Mice through Reducing Tau Pathology and Improving Synaptic Dysfunction. Curr Med Sci 2021; 40:1031-1039. [PMID: 33428130 DOI: 10.1007/s11596-020-2284-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/19/2020] [Indexed: 12/15/2022]
Abstract
rTg4510 mice are transgenic mice expressing P301L mutant tau and have been developed as an animal model of tauopathies including Alzheimer's disease (AD). Besides cognitive impairments, rTg4510 mice also show abnormal hyperactivity behavior. Cornel iridoid glycoside (CIG) is an active ingredient extracted from Cornus officinalis, a traditional Chinese herb. The purpose of the present study was to investigate the effects of CIG on the emotional disorders such as hyperactivity, and related mechanisms. The emotional hyperactivity was detected by locomotor activity test and Y maze test. Immunofluorescent and immunohistochemical analyses were conducted to measure neuron loss and phosphorylated tau. Western blotting was used to detect the expression of related proteins. The results showed that intragastric administration of CIG for 3 months decreased the hyperactivity phenotype, prevented neuronal loss, reduced tau hyperphosphorylation and aggregation in the amygdala of rTg4510 mice. Meanwhile, CIG alleviated the synaptic dysfunction by increasing the expression of N-methyl-D-aspartate receptors (NMDARs) subunits GluN1 and GluN2A and αamino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) subunits GluA1 and GluA2, and increased the level of phosphorylated Ca2+/calmodulin dependent protein kinase II α (p-CaMK IIα) in the brain of rTg4510 mice. In conclusion, CIG may have potential to treat the emotional disorders in tauopathies such as AD through reducing tau pathology and improving synaptic dysfunction.
Collapse
Affiliation(s)
- Deng-Lei Ma
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.,Beijing Institute for Brain Disorders, Beijing, 100053, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, 100053, China
| | - Yi Luo
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.,Beijing Institute for Brain Disorders, Beijing, 100053, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, 100053, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Rui Huang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.,Beijing Institute for Brain Disorders, Beijing, 100053, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, 100053, China
| | - Zi-Run Zhao
- Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, 11794, USA
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.,Beijing Institute for Brain Disorders, Beijing, 100053, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, 100053, China
| | - Ya-Li Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.,Beijing Institute for Brain Disorders, Beijing, 100053, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, 100053, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China. .,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China. .,Beijing Institute for Brain Disorders, Beijing, 100053, China. .,Beijing Engineering Research Center for Nerve System Drugs, Beijing, 100053, China.
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China. .,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China. .,Beijing Institute for Brain Disorders, Beijing, 100053, China. .,Beijing Engineering Research Center for Nerve System Drugs, Beijing, 100053, China.
| |
Collapse
|
9
|
Tweedy C, Kindred N, Curry J, Williams C, Taylor JP, Atkinson P, Randall F, Erskine D, Morris CM, Reeve AK, Clowry GJ, LeBeau FEN. Hippocampal network hyperexcitability in young transgenic mice expressing human mutant alpha-synuclein. Neurobiol Dis 2020; 149:105226. [PMID: 33347975 PMCID: PMC7859835 DOI: 10.1016/j.nbd.2020.105226] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/12/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Abnormal excitability in cortical networks has been reported in patients and animal models of Alzheimer's disease (AD), and other neurodegenerative conditions. Whether hyperexcitability is a core feature of alpha(α)-synucleinopathies, including dementia with Lewy bodies (DLB) is unclear. To assess this, we used two murine models of DLB that express either human mutant α-synuclein (α-syn) the hA30P, or human wild-type α-syn (hWT-α-syn) mice. We observed network hyperexcitability in vitro in young (2–5 months), pre-symptomatic transgenic α-syn mice. Interictal discharges (IIDs) were seen in the extracellular local field potential (LFP) in the hippocampus in hA30P and hWT-α-syn mice following kainate application, while only gamma frequency oscillations occurred in control mice. In addition, the concentration of the GABAA receptor antagonist (gabazine) needed to evoke IIDs was lower in slices from hA30P mice compared to control mice. hA30P mice also showed increased locomotor activity in the open field test compared to control mice. Intracellular recordings from CA3 pyramidal cells showed a more depolarised resting membrane potential in hA30P mice. Quadruple immunohistochemistry for human α-syn, and the mitochondrial markers, porin and the complex IV enzyme cytochrome c oxidase subunit 1 (COX1) in parvalbumin (PV+)-expressing interneurons showed that 25% of PV+ cells contained human α-syn in hA30P mice. While there was no change in PV expression, COX1 expression was significantly increased in PV+ cells in hA30P mice, perhaps reflecting a compensatory change to support PV+ interneuron activity. Our findings suggest that hippocampal network hyperexcitability may be an important early consequence of α-syn-mediated impairment of neuronal/synaptic function, which occurs without any overt loss of PV interneurons. The therapeutic benefit of targeting network excitability early in the disease stage should be explored with respect to α-synucleinopathies such as DLB. Young transgenic α-syn mice exhibit network hyperexcitability in the hippocampus in vitro. Young transgenic α-syn mice have increased locomotor activity in an open field test. Hippocampal pyramidal cells are more depolarised in young transgenic α-syn mice. Increased mitochondrial cytochrome c oxidase (complex IV) function in PV+ interneurons in young transgenic a-syn mice.
Collapse
Affiliation(s)
- Clare Tweedy
- Biosciences Institute, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Nathan Kindred
- Biosciences Institute, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Joshua Curry
- Biosciences Institute, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Christopher Williams
- Biosciences Institute, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - John-Paul Taylor
- Institute of Clinical and Translational Research, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Peter Atkinson
- Eisai Hatfield Research Laboratories, Eisai Ltd., European Knowledge Centre, Mosquito Way, Hatfield, Herts AL10 9SN, UK
| | - Fiona Randall
- Previously Eisai AiM Institute, Eisai Inc., 4 Corporate Drive, Andover, MA 01810, USA
| | - Daniel Erskine
- Institute of Clinical and Translational Research, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Christopheer M Morris
- Institute of Clinical and Translational Research, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Amy K Reeve
- Institute of Clinical and Translational Research, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Gavin J Clowry
- Biosciences Institute, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Fiona E N LeBeau
- Biosciences Institute, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK.
| |
Collapse
|
10
|
Soeda Y, Takashima A. New Insights Into Drug Discovery Targeting Tau Protein. Front Mol Neurosci 2020; 13:590896. [PMID: 33343298 PMCID: PMC7744460 DOI: 10.3389/fnmol.2020.590896] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Microtubule-associated protein tau is characterized by the fact that it is an intrinsically disordered protein due to its lack of a stable conformation and high flexibility. Intracellular inclusions of fibrillar forms of tau with a β-sheet structure accumulate in the brain of patients with Alzheimer's disease and other tauopathies. Accordingly, detachment of tau from microtubules and transition of tau from a disordered state to an abnormally aggregated state are essential events preceding the onset of tau-related diseases. Many reports have shown that this transition is caused by post-translational modifications, including hyperphosphorylation and acetylation. The misfolded tau is self-assembled and forms a tau oligomer before the appearance of tau inclusions. Animal and pathological studies using human samples have demonstrated that tau oligomer formation contributes to neuronal loss. During the progression of tauopathies, tau seeds are released from cells and incorporated into other cells, leading to the propagation of pathological tau aggregation. Accumulating evidence suggests several potential approaches for blocking tau-mediated toxicity: (1) direct inhibition of pathological tau aggregation and (2) inhibition of tau post-translational modifications that occur prior to pathological tau aggregation, (3) inhibition of tau propagation and (4) stabilization of microtubules. In addition to traditional low-molecular-weight compounds, newer drug discovery approaches such as the development of medium-molecular-weight drugs (peptide- or oligonucleotide-based drugs) and high-molecular-weight drugs (antibody-based drugs) provide alternative pathways to preventing the formation of abnormal tau. Of particular interest are recent studies suggesting that tau droplet formation by liquid-liquid phase separation may be the initial step in aberrant tau aggregation, as well results that implicate roles for tau in dendritic and nuclear functions. Here, we review the mechanisms through which drugs can target tau and consider recent clinical trials for the treatment of tauopathies. In addition, we discuss the utility of these newer strategies and propose future directions for research on tau-targeted therapeutics.
Collapse
Affiliation(s)
- Yoshiyuki Soeda
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Akihiko Takashima
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| |
Collapse
|
11
|
Visser PJ, Reus LM, Gobom J, Jansen I, Dicks E, Tsolaki M, Verhey FRJ, Popp J, Martinez-Lage P, Vandenberghe R, Lleó A, Molinuevo JL, Engelborghs S, Freund-Levi Y, Froelich L, Sleegers K, Dobricic V, Hong S, Lovestone S, Streffer J, Vos SJB, Bos I, Smit AB, Blennow K, Scheltens P, Teunissen CE, Bertram L, Zetterberg H, Tijms BM. Cerebrospinal fluid total tau levels indicate aberrant neuronal plasticity in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 33173883 DOI: 10.1101/2020.10.29.20211920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is characterised by abnormal amyloid beta and tau processing. Previous studies reported that cerebrospinal fluid (CSF) total tau (t-tau) levels vary between patients. Here we show that CSF t-tau variability is associated with distinct impairments in neuronal plasticity mediated by gene repression factors SUZ12 and REST. AD individuals with abnormal t-tau levels have increased CSF concentrations of plasticity proteins regulated by SUZ12 and REST. AD individuals with normal t-tau, on the contrary, have decreased concentrations of these plasticity proteins and increased concentrations in proteins associated with blood-brain and blood CSF-barrier dysfunction. Genomic analyses suggested that t-tau levels in part depend on genes involved in gene expression. The distinct plasticity abnormalities in AD as signaled by t-tau urge the need for personalised treatment.
Collapse
|
12
|
Brandt R, Trushina NI, Bakota L. Much More Than a Cytoskeletal Protein: Physiological and Pathological Functions of the Non-microtubule Binding Region of Tau. Front Neurol 2020; 11:590059. [PMID: 33193056 PMCID: PMC7604284 DOI: 10.3389/fneur.2020.590059] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Tau protein (MAPT) is classified as a microtubule-associated protein (MAP) and is believed to regulate the axonal microtubule arrangement. It belongs to the tau/MAP2/MAP4 family of MAPs that have a similar microtubule binding region at their carboxy-terminal half. In tauopathies, such as Alzheimer's disease, tau is distributed more in the somatodendritic compartment, where it aggregates into filamentous structures, the formation of which correlates with cognitive impairments in patients. While microtubules are the dominant interaction partners of tau under physiological conditions, tau has many additional interaction partners that can contribute to its physiological and pathological role. In particular, the amino-terminal non-microtubule binding domain (N-terminal projection region, NTR) of tau interacts with many partners that are involved in membrane organization. The NTR contains intrinsically disordered regions (IDRs) that show a strong evolutionary increase in the disorder and may have been the basis for the development of new, tau-specific interactions. In this review we discuss the functional organization of the tau protein and the special features of the tau non-microtubule binding region also in the connection with the results of Tau KO models. We consider possible physiological and pathological functions of tau's non-microtubule interactions, which could indicate that interactions mediated by tau's NTR and regulated by far-reaching functional interactions of the PRR and the extreme C-terminus of tau contribute to the pathological processes.
Collapse
Affiliation(s)
- Roland Brandt
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany.,Center for Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany.,Institute of Cognitive Science, University of Osnabrück, Osnabrück, Germany
| | | | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
13
|
Zhao F, Tian H, Chinnathambi A, Alharbi SA, Yang H. Neuroprotective effect of rhaponticin against Parkinson disease: Insights from in vitro BV-2 model and in vivo MPTP-induced mice model. J Biochem Mol Toxicol 2020; 35:e22631. [PMID: 32926752 DOI: 10.1002/jbt.22631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/22/2020] [Accepted: 09/02/2020] [Indexed: 12/26/2022]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative illness associated with the loss or damage to neurons of the dopaminergic system in the brain. Few therapeutic approaches and considerable side effects of conventional drugs necessitate a new therapeutic agent to treat patients with PD. Rhaponticin is a natural hydroxystilbene, found in herbal plants such as Rheum rhaponticum, and known to have desirable biological activity including anti-inflammatory properties. However, the neuroinflammation on rhaponticin levels has only been investigated partially so far. So, the current study explored whether rhaponticin could ameliorate the pathophysiology observed in both the in vitro microglial BV-2 cells and the in vivo (1-methyl-4-phenyl-1,2,3,5-tetrahydropyridine [MPTP])-mediated PD model. The results show rhaponticin significantly attenuated lipopolysaccharide (LPS)-mediated microglial activation by suppressing nitric oxide synthase in conjunction with abridged reactive oxygen species production together with proinflammatory mediator reduction. In vivo rhaponticin treatment improves motor impairments as well as the loss of dopaminergic neurons in MPTP-treated mice possibly through suppression via mediators of inflammation. Taken together, these results offer evidence that rhaponticin exerts anti-inflammatory effects and neuroprotection in an LPS-induced microglial model and the MPTP-induced mouse models of PD.
Collapse
Affiliation(s)
- Fangfang Zhao
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng City, Henan, China
| | - Huijun Tian
- Department of Neurosurgery, Beijing Beiya Orthopedics Hospital, Beijing, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hongan Yang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
14
|
Rodriguez GA, Barrett GM, Duff KE, Hussaini SA. Chemogenetic attenuation of neuronal activity in the entorhinal cortex reduces Aβ and tau pathology in the hippocampus. PLoS Biol 2020; 18:e3000851. [PMID: 32822389 PMCID: PMC7467290 DOI: 10.1371/journal.pbio.3000851] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/02/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
High levels of the amyloid-beta (Aβ) peptide have been shown to disrupt neuronal function and induce hyperexcitability, but it is unclear what effects Aβ-associated hyperexcitability may have on tauopathy pathogenesis or propagation in vivo. Using a novel transgenic mouse line to model the impact of human APP (hAPP)/Aβ accumulation on tauopathy in the entorhinal cortex–hippocampal (EC-HIPP) network, we demonstrate that hAPP overexpression aggravates EC-Tau aggregation and accelerates pathological tau spread into the hippocampus. In vivo recordings revealed a strong role for hAPP/Aβ, but not tau, in the emergence of EC neuronal hyperactivity and impaired theta rhythmicity. Chronic chemogenetic attenuation of EC neuronal hyperactivity led to reduced hAPP/Aβ accumulation and reduced pathological tau spread into downstream hippocampus. These data strongly support the hypothesis that in Alzheimer’s disease (AD), Aβ-associated hyperactivity accelerates the progression of pathological tau along vulnerable neuronal circuits, and demonstrates the utility of chronic, neuromodulatory approaches in ameliorating AD pathology in vivo. A novel, triple transgenic mouse model of Alzheimer's disease reveals that amyloid beta-associated neuronal hyperactivity and network dysfunction accelerates the spread of pathological tau from the entorhinal cortex into the hippocampus. Chronic attenuation of neuronal activity using chemogenetics reduces this effect, supporting a role for neuronal hyperactivity in Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Gustavo A. Rodriguez
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Geoffrey M. Barrett
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Karen E. Duff
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
- UK Dementia Research Institute at University College London, London, United Kingdom
- * E-mail: (SAH); (KED)
| | - S. Abid Hussaini
- Taub Institute for Research on Alzheimer’s disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
- * E-mail: (SAH); (KED)
| |
Collapse
|
15
|
Al Rihani SB, Darakjian LI, Kaddoumi A. Oleocanthal-Rich Extra-Virgin Olive Oil Restores the Blood-Brain Barrier Function through NLRP3 Inflammasome Inhibition Simultaneously with Autophagy Induction in TgSwDI Mice. ACS Chem Neurosci 2019; 10:3543-3554. [PMID: 31244050 DOI: 10.1021/acschemneuro.9b00175] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by multiple hallmarks including extracellular amyloid (Aβ) plaques, neurofibrillary tangles, dysfunctional blood-brain barrier (BBB), neuroinflammation, and impaired autophagy. Thus, novel strategies that target multiple disease pathways would be essential to prevent, halt, or treat the disease. A growing body of evidence including our studies supports a protective effect of oleocanthal (OC) and extra-virgin olive oil (EVOO) at early AD stages before the onset of pathology. In addition, we reported previously that OC and EVOO exhibited such effect by restoring the BBB function; however, the mechanism(s) by which OC and EVOO exert such an effect and whether this effect extends to a later stage of AD remain unknown. In this work, we sought first to test the effect of OC-rich EVOO consumption at an advanced stage of the disease in TgSwDI mice, an AD mouse model, starting at the age of 6 months for 3 months treatment, and then to elucidate the mechanism(s) by which OC-rich EVOO exerts the observed beneficial effect. Overall findings demonstrated that OC-rich EVOO restored the BBB function and reduced AD-associated pathology by reducing neuroinflammation through inhibition of NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome and inducing autophagy through activation of AMP-activated protein kinase (AMPK)/Unc-51-like autophagy activating kinase 1 (ULK1) pathway. Thus, diet supplementation with OC-rich EVOO could provide beneficial effect to slow or halt the progression of AD.
Collapse
Affiliation(s)
- Sweilem B. Al Rihani
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Pharmacy Research Building, Auburn University, Auburn, Alabama 36849, United States
- Center for Neuroscience Initiative, Auburn University, Auburn, Alabama 36849, United States
| | - Lucy I. Darakjian
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Pharmacy Research Building, Auburn University, Auburn, Alabama 36849, United States
| | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Pharmacy Research Building, Auburn University, Auburn, Alabama 36849, United States
- Center for Neuroscience Initiative, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
16
|
Progressive age-dependent motor impairment in human tau P301S overexpressing mice. Behav Brain Res 2019; 376:112158. [PMID: 31442549 DOI: 10.1016/j.bbr.2019.112158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/13/2019] [Accepted: 08/17/2019] [Indexed: 12/31/2022]
Abstract
This study assessed the development of motor deficits in female hTau.P301S transgenic mice from 1.5 to 5.5 months of age. The test battery included clasping reflex, grid hanging, Rotarod test, spontaneous explorative activity, Catwalk gait analysis, and nest building. Starting from the age of 2-3 months the mice showed marked hyperactivity, abnormal placing of weight on the hindlimbs and defective nest building in their home cage. These behavioral impairments did not progress with age. In addition, there was a progressive development of hindlimb clasping, inability to stay on a rotating rod or hang on a metal grid, and gait impairment. Depending on the measured output parameter, the motor impairment became significant from 3 to 4 months onwards and rapidly worsened until the age of 5.5 months with little inter-individual variation. The progressive motor impairment was paralleled by a robust increase in AT8 p-tau positive neurons in deep cerebellar nuclei and pontine brainstem between 3 and 5.5 months of age. The quick and steadily progressive motor impairment between 3 and 5.5 months of age accompanied by robust development of tau pathology in the hindbrain makes this mouse well suited for preclinical studies aiming at slowing down tau pathology associated with primary or secondary tauopathies.
Collapse
|
17
|
Ano Y, Yoshikawa M, Takaichi Y, Michikawa M, Uchida K, Nakayama H, Takashima A. Iso-α-Acids, Bitter Components in Beer, Suppress Inflammatory Responses and Attenuate Neural Hyperactivation in the Hippocampus. Front Pharmacol 2019; 10:81. [PMID: 30804789 PMCID: PMC6378368 DOI: 10.3389/fphar.2019.00081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/21/2019] [Indexed: 11/19/2022] Open
Abstract
Due to the growth in aging populations worldwide, prevention and therapy for age-related cognitive decline and dementia are in great demand. We previously demonstrated that long-term intake of iso-α-acids, which are hop-derived bitter compounds found in beer, prevent Alzheimer’s pathology in a rodent model. On the other hand, the effects of iso-α-acids on neural activity in Alzheimer’s disease model mice have not been investigated. Here, we demonstrated that short-term intake of iso-α-acids suppresses inflammation in the hippocampus and improves memory impairment even after disease onset. Importantly, we demonstrated that short-term administration of iso-α-acids attenuated the neural hyperactivation in hippocampus. In 6-month-old 5 × FAD mice exhibiting hippocampus inflammation and memory impairment, oral administration of iso-α-acids for 7 days reduced inflammatory cytokines, including MIP-1α and soluble Aβ and improved object memory in the novel object recognition test. In 12-month-old J20 mice, intake of iso-α-acids for 7 days also suppressed inflammatory cytokines and soluble Aβ in the brain. Manganese-enhanced magnetic resonance imaging (MEMRI) of hippocampi of J20 mice showed increased manganese compared with wild type mice, but iso-α-acids canceled this increased MEMRI signal in J20 mice, particularly in the hippocampus CA1 and CA3 region. Taken together, these findings suggest that short-term intake of iso-α-acids can suppress hippocampus inflammation even after disease onset and improve hyper neural activity in Alzheimer’s disease model mice.
Collapse
Affiliation(s)
- Yasuhisa Ano
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.,Research Laboratories for Health Science & Food Technologies, Kirin Company Ltd., Kanagawa, Japan
| | - Misato Yoshikawa
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yuta Takaichi
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Makoto Michikawa
- Department of Biochemistry, School of Medicine, Nagoya City University, Nagoya, Japan
| | - Kazuyuki Uchida
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Nakayama
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Akihiko Takashima
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Obu, Japan.,Faculty of Science, Gakushuin University, Tokyo, Japan
| |
Collapse
|
18
|
Liu X, Hebron ML, Mulki S, Wang C, Lekah E, Ferrante D, Shi W, Kurd-Misto B, Moussa C. Ubiquitin Specific Protease 13 Regulates Tau Accumulation and Clearance in Models of Alzheimer's Disease. J Alzheimers Dis 2019; 72:425-441. [PMID: 31594232 DOI: 10.3233/jad-190635] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ubiquitin Specific Protease-13 (USP13) is a de-ubiquinating enzyme that regulates protein ubiquitination and clearance. The role of USP13 is largely unknown in neurodegeneration. In this study we aim to demonstrate whether tau accumulation and/or clearance depends on ubiquitination/de-ubiquitination via USP-13. We used transgenic animal models of human amyloid precursor protein (APP) or P301L tau mutations and genetically knocked-down USP13 expression via shRNA to determine USP13 effects on tau ubiquitination and levels. We found a two-fold increase of USP13 levels in postmortem Alzheimer's disease (AD) brains. USP13 knockdown significantly increased the activity of the 20S proteasome and reduced the levels of hyper-phosphorylated tau (p-tau) in primary cortical neurons. USP13 knockdown also reduced the levels of amyloid and increased p-tau ubiquitination and clearance in transgenic animal models that overexpress murine tau as a result of the expression of familial APP mutations (TgAPP) and the human mutant P301L tau (rTg4510), respectively. Clearance of p-tau appears to be mediated by autophagy in these animal models. Taken together, these data suggest that USP13 knockdown reduces p-tau accumulation via regulation of ubiquitination/de-ubiquitination and mediates its clearance via autophagy and/or the proteasome. These results suggest that USP13 inhibition may be a therapeutic strategy to reduce accumulation of plaques and toxic p-tau in AD and human tauopathies.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Sanjana Mulki
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Chen Wang
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Elizabeth Lekah
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Dalila Ferrante
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Wangke Shi
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Bahjat Kurd-Misto
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
19
|
Bushen-Yizhi Formula Alleviates Neuroinflammation via Inhibiting NLRP3 Inflammasome Activation in a Mouse Model of Parkinson's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3571604. [PMID: 30224927 PMCID: PMC6129340 DOI: 10.1155/2018/3571604] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/20/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disease, is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Although the molecular mechanisms underlying dopaminergic neuronal degeneration in PD remain unclear, neuroinflammation is considered as the vital mediator in the pathogenesis and progression of PD. Bushen-Yizhi Formula (BSYZ), a traditional Chinese medicine, has been demonstrated to exert antineuroinflammation in our previous studies. However, it remains unclear whether BSYZ is effective for PD. Here, we sought to assess the neuroprotective effects and explore the underlying mechanisms of BSYZ in a 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine- (MPTP-) induced mouse model of PD. Our results indicate that BSYZ significantly alleviates the motor impairments and dopaminergic neuron degeneration of MPTP-treated mice. Furthermore, BSYZ remarkably attenuates microglia activation, inhibits NLPR3 activation, and decreases the levels of inflammatory cytokines in MPTP-induced mouse brain. Also, BSYZ inhibits NLRP3 activation and interleukin-1β production of the 1-methyl-4-phenyl-pyridinium (MPP+) stimulated BV-2 microglia cells. Taken together, our results indicate that BSYZ alleviates MPTP-induced neuroinflammation probably via inhibiting NLRP3 inflammasome activation in microglia. Collectively, BSYZ may be a potential therapeutic agent for PD and the related neurodegeneration diseases.
Collapse
|
20
|
Zhang WF, Wang X, Wang K, Duan LP. Early life esophageal acid exposure reduces expression of NMDAR1 in the adult rat dorsal hippocampus and medial prefrontal cortex: Potential relationship with hyperlocomotion. J Dig Dis 2018; 19:485-497. [PMID: 30058264 DOI: 10.1111/1751-2980.12650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 07/13/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Early life esophageal acid exposure causes long-term molecular alterations in the rostral cingulate cortex; however, whether it induces behavioral changes remains unverified. Little is known about the molecular changes resulting from this event in the developing hippocampus and medial prefrontal cortex (mPFC). This study aimed to investigate the influence of early life esophageal acid exposure on spontaneous locomotor behavior and N-methyl-D-aspartate receptor (NMDAR), expression in these brain regions of adult rats. METHODS Male Sprague-Dawley rats were administered with an esophageal acid or saline infusion once per day (postnatal days 7-14). Some of these rats were given acute esophageal acid rechallenge in adulthood (postnatal day 60). The spontaneous locomotor behavior and expressions of esophageal epithelial caludin-1 and NMDAR subunits in the dorsal hippocampus (DH), ventral hippocampus (VH) and mPFC of the adult rats were recorded. RESULTS Neonatal esophageal acid stimulation caused long-term impairment of the tight junctions in the adult esophagus. Simultaneously, hyperlocomotion and reduced expression of NMDAR1 subunits in both the DH and mPFC were observed, but not in the VH regions. Adult acute acid rechallenge reversed the decreased NMDAR1 expression in the DH and mPFC. The glycine ligand to NMDAR1 subunits was also changed. CONCLUSIONS Peripheral visceral stimulation such as esophageal acid exposure during cerebral development induces increased locomotor activity, which may be related to the alteration of central sensitivity via NMDAR1 subunit reduction in the DH and mPFC. The impairment of tight junctions in the esophageal epithelium may contribute to the formation of central neuroplasticity.
Collapse
Affiliation(s)
- Wei Fang Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Kun Wang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Li Ping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| |
Collapse
|