1
|
Jin J, Fu C, Xia J, Luo H, Wang X, Chen S, Mao H, Yuan K, Lu L, Xiong W, Zou G. Cannabidiol ameliorates cognitive decline in 5×FAD mouse model of Alzheimer's disease through potentiating the function of extrasynaptic glycine receptors. Mol Psychiatry 2025; 30:1817-1827. [PMID: 39396064 DOI: 10.1038/s41380-024-02789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Emerging evidence supports the therapeutic potential of cannabinoids in Alzheimer's disease (AD), but the underlying mechanism upon how cannabinoids impact brain cognition and AD pathology remains unclear. Here we show that chronic cannabidiol (CBD) administration significantly mitigates cognitive deficiency and hippocampal β-amyloid (Aβ) pathology in 5×FAD mouse model of AD. CBD achieves its curative effect mainly through potentiating the function of inhibitory extrasynaptic glycine receptor (GlyR) in hippocampal dentate gyrus (DG). Based on the in vitro and in vivo electrophysiological recording and calcium imaging, CBD mediated anti-AD effects via GlyR are mainly accomplished by decreasing neuronal hyperactivity of granule cells in the DG of AD mice. Furthermore, the AAV-mediated ablation of DG GlyRα1, or the GlyRα1S296A mutation that exclusively disrupts CBD binding, significantly intercepts the anti-AD effect of CBD. These findings suggest a GlyR dependent mechanism underlying the therapeutic potential of CBD in the treatment of AD.
Collapse
Grants
- 32225020, 91849206, 91942315, 92049304, 32121002, 81901157, 82241032 National Natural Science Foundation of China (National Science Foundation of China)
- 32225020, 91849206, 91942315, 92049304, 32121002 National Natural Science Foundation of China (National Science Foundation of China)
Collapse
Affiliation(s)
- Jin Jin
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Chonglei Fu
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China
| | - Jing Xia
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Heyi Luo
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xianglian Wang
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Si Chen
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Huanhuan Mao
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, 100191, Beijing, China
| | - Lin Lu
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China.
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, 100191, Beijing, China.
| | - Wei Xiong
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China.
| |
Collapse
|
2
|
McGregor ER, Lasky DJ, Rippentrop OJ, Clark JP, Wright S, Jones MV, Anderson RM. Reversal of neuronal tau pathology via adiponectin receptor activation. Commun Biol 2025; 8:8. [PMID: 39755746 PMCID: PMC11700159 DOI: 10.1038/s42003-024-07391-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 12/09/2024] [Indexed: 01/06/2025] Open
Abstract
Changes in brain mitochondrial metabolism are coincident with functional decline; however, direct links between the two have not been established. Here, we show that mitochondrial targeting via the adiponectin receptor activator AdipoRon (AR) clears neurofibrillary tangles (NFTs) and rescues neuronal tauopathy-associated defects. AR reduced levels of phospho-tau and lowered NFT burden by a mechanism involving the energy-sensing kinase AMPK and the growth-sensing kinase GSK3b. The transcriptional response to AR included broad metabolic and functional pathways. Induction of lysosomal pathways involved activation of LC3 and p62, and restoration of neuronal outgrowth required the stress-responsive kinase JNK. Negative consequences of NFTs on mitochondrial activity, ATP production, and lipid stores were corrected. Defects in electrophysiological measures (e.g., resting potential, resistance, spiking profiles) were also corrected. These findings reveal a network linking mitochondrial function, cellular maintenance processes, and electrical aspects of neuronal function that can be targeted via adiponectin receptor activation.
Collapse
Affiliation(s)
- Eric R McGregor
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Danny J Lasky
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Olivia J Rippentrop
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Josef P Clark
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI, USA
| | - Samantha Wright
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Mathew V Jones
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Rozalyn M Anderson
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- GRECC William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
3
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
4
|
Thacker JS, Bettio L, Liang S, Shkolnikov I, Collingridge GL, Christie BR. Adiponectin rescues synaptic plasticity in the dentate gyrus of a mouse model of Fragile X Syndrome. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230221. [PMID: 38853554 PMCID: PMC11343265 DOI: 10.1098/rstb.2023.0221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 06/11/2024] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and is the leading known single-gene cause of autism spectrum disorder. Patients with FXS display varied behavioural deficits that include mild to severe cognitive impairments in addition to mood disorders. Currently, there is no cure for this condition; however, there is an emerging focus on therapies that inhibit mechanistic target of rapamycin (mTOR)-dependent protein synthesis owing to the clinical effectiveness of metformin for alleviating some behavioural symptoms in FXS. Adiponectin (APN) is a neurohormone that is released by adipocytes and provides an alternative means to inhibit mTOR activation in the brain. In these studies, we show that Fmr1 knockout mice, like patients with FXS, show reduced levels of circulating APN and that both long-term potentiation (LTP) and long-term depression (LTD) in the dentate gyrus (DG) are impaired. Brief (20 min) incubation of hippocampal slices in APN (50 nM) was able to rescue both LTP and LTD in the DG and increased both the surface expression and phosphorylation of GluA1 receptors. These results provide evidence for reduced APN levels in FXS playing a role in decreasing bidirectional synaptic plasticity and show that therapies which enhance APN levels may have therapeutic potential for this and related conditions.This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Jonathan S. Thacker
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, OntarioM5G 1X5, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, OntarioM5S 1A8, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Luis Bettio
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Stanley Liang
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Irene Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Graham L. Collingridge
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, OntarioM5G 1X5, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, OntarioM5S 1A8, Canada
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
- Island Medical Program, University of British Columbia, Victoria, British ColumbiaV8P 5C2, Canada
- Center for Behavioral Teratology, San Diego State University, San Diego, CA92120, USA
| |
Collapse
|
5
|
Kourti M, Metaxas A. A systematic review and meta-analysis of tau phosphorylation in mouse models of familial Alzheimer's disease. Neurobiol Dis 2024; 192:106427. [PMID: 38307366 DOI: 10.1016/j.nbd.2024.106427] [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: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024] Open
Abstract
Transgenic models of familial Alzheimer's disease (AD) serve as valuable tools for probing the molecular mechanisms associated with amyloid-beta (Aβ)-induced pathology. In this meta-analysis, we sought to evaluate levels of phosphorylated tau (p-tau) and explore potential age-related variations in tau hyperphosphorylation, within mouse models of AD. The PubMed and Scopus databases were searched for studies measuring soluble p-tau in 5xFAD, APPswe/PSEN1de9, J20 and APP23 mice. Data were extracted and analyzed using standardized procedures. For the 5xFAD model, the search yielded 36 studies eligible for meta-analysis. Levels of p-tau were higher in 5xFAD mice relative to control, a difference that was evident in both the carboxy-terminal (CT) and proline-rich (PR) domains of tau. Age negatively moderated the relationship between genotype and CT phosphorylated tau in studies using hybrid mice, female mice, and preparations from the neocortex. For the APPswe/PSEN1de9 model, the search yielded 27 studies. Analysis showed tau hyperphosphorylation in transgenic vs. control animals, evident in both the CT and PR regions of tau. Age positively moderated the relationship between genotype and PR domain phosphorylated tau in the neocortex of APPswe/PSEN1de9 mice. A meta-analysis was not performed for the J20 and APP23 models, due to the limited number of studies measuring p-tau levels in these mice (<10 studies). Although tau is hyperphosphorylated in both 5xFAD and APPswe/PSEN1de9 mice, the effects of ageing on p-tau are contingent upon the model being examined. These observations emphasize the importance of tailoring model selection to the appropriate disease stage when considering the relationship between Aβ and tau, and suggest that there are optimal intervention points for the administration of both anti-amyloid and anti-tau therapies.
Collapse
Affiliation(s)
- Malamati Kourti
- School of Sciences, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus; Angiogenesis and Cancer Drug Discovery Group, Basic and Translational Cancer Research Centre, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus.
| | - Athanasios Metaxas
- School of Sciences, Department of Life Sciences, European University Cyprus, 2404 Egkomi, Nicosia, Cyprus; Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
6
|
McGregor ER, Lasky DJ, Rippentrop OJ, Clark JP, Wright SLG, Jones MV, Anderson RM. Reversal of neuronal tau pathology, metabolic dysfunction, and electrophysiological defects via adiponectin pathway-dependent AMPK activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579204. [PMID: 38370802 PMCID: PMC10871331 DOI: 10.1101/2024.02.07.579204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Changes in brain mitochondrial metabolism are coincident with functional decline; however, direct links between the two have not been established. Here, we show that mitochondrial targeting via the adiponectin receptor activator AdipoRon (AR) clears neurofibrillary tangles (NFTs) and rescues neuronal tauopathy-associated defects. AR reduced levels of phospho-tau and lowered NFT burden by a mechanism involving the energy-sensing kinase AMPK and the growth-sensing kinase GSK3b. The transcriptional response to AR included broad metabolic and functional pathways. Induction of lysosomal pathways involved activation of LC3 and p62, and restoration of neuronal outgrowth required the stress-responsive kinase JNK. Negative consequences of NFTs on mitochondrial activity, ATP production, and lipid stores were corrected. Defects in electrophysiological measures (e.g., resting potential, resistance, spiking profiles) were also corrected. These findings reveal a network linking mitochondrial function, cellular maintenance processes, and electrical aspects of neuronal function that can be targeted via adiponectin receptor activation.
Collapse
Affiliation(s)
- Eric R McGregor
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Danny J Lasky
- Department. of Neuroscience, Univ. of Wisconsin-Madison, Madison, WI
| | | | - Josef P Clark
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
| | | | - Mathew V Jones
- Department. of Neuroscience, Univ. of Wisconsin-Madison, Madison, WI
| | - Rozalyn M Anderson
- Division of Geriatrics, Department of Medicine, SMPH, University of Wisconsin-Madison, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- GRECC William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
7
|
Bonnet M, Ertlen C, Seblani M, Brezun JM, Coyle T, Cereda C, Zuccotti G, Colli M, Desouches C, Decherchi P, Carelli S, Marqueste T. Activated Human Adipose Tissue Transplantation Promotes Sensorimotor Recovery after Acute Spinal Cord Contusion in Rats. Cells 2024; 13:182. [PMID: 38247873 PMCID: PMC10814727 DOI: 10.3390/cells13020182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Traumatic spinal cord injuries (SCIs) often result in sensory, motor, and vegetative function loss below the injury site. Although preclinical results have been promising, significant solutions for SCI patients have not been achieved through translating repair strategies to clinical trials. In this study, we investigated the effective potential of mechanically activated lipoaspirated adipose tissue when transplanted into the epicenter of a thoracic spinal contusion. Male Sprague Dawley rats were divided into three experimental groups: SHAM (uninjured and untreated), NaCl (spinal cord contusion with NaCl application), and AF (spinal cord contusion with transplanted activated human fat). Pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) were measured to assess endogenous inflammation levels 14 days after injury. Sensorimotor recovery was monitored weekly for 12 weeks, and gait and electrophysiological analyses were performed at the end of this observational period. The results indicated that AF reduced endogenous inflammation post-SCI and there was a significant improvement in sensorimotor recovery. Moreover, activated adipose tissue also reinstated the segmental sensorimotor loop and the communication between supra- and sub-lesional spinal cord regions. This investigation highlights the efficacy of activated adipose tissue grafting in acute SCI, suggesting it is a promising therapeutic approach for spinal cord repair after traumatic contusion in humans.
Collapse
Affiliation(s)
- Maxime Bonnet
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Céline Ertlen
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Mostafa Seblani
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Jean-Michel Brezun
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Thelma Coyle
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
- Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Mattia Colli
- Podgora7 Clinic, Via Podgora 7, 20122 Milano, Italy
| | - Christophe Desouches
- Clinique Phénicia—CD Esthétique, 5 Boulevard Notre Dame, F-13006 Marseille, France
| | - Patrick Decherchi
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
| | - Tanguy Marqueste
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| |
Collapse
|
8
|
Wang C, Cui Y, Xu T, Zhou Y, Yang R, Wang T. New insights into glycogen synthase kinase-3: A common target for neurodegenerative diseases. Biochem Pharmacol 2023; 218:115923. [PMID: 37981175 DOI: 10.1016/j.bcp.2023.115923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Glycogen synthase kinase 3 (GSK-3) is a highly conserved protein serine/threonine kinase that plays a central role in a wide variety of cellular processes to coordinate catabolic and anabolic pathways and regulate cell growth and fate. There is increasing evidence showing that abnormal glycogen synthase kinase 3 (GSK-3) is associated with the pathogenesis and progression of many disorders, such as cancer, diabetes, psychiatric diseases, and neurodegenerative diseases. In this review, we summarize recent findings about the regulatory role of GSK-3 in the occurrence and development of multiple neurodegenerative diseases, mainly focusing on Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. The aim of this study is to provide new insight into the shared working mechanism of GSK-3 as a therapeutic target of multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Chengfeng Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China; Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong 266071, China
| | - Yu Cui
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Tong Xu
- Department of Otorhinolaryngology Head and Neck, The Affiliated Qingdao Third People's Hospital of Qingdao University, Qingdao, Shandong 266021, China
| | - Yu Zhou
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China; Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, Shandong 266071, China; Department of Otorhinolaryngology Head and Neck, The Affiliated Qingdao Third People's Hospital of Qingdao University, Qingdao, Shandong 266021, China; Department of Health and Life Science, University of Health and Rehabilitation Sciences, Qingdao, Shandong 266000, China.
| | - Rong Yang
- Department of Otorhinolaryngology Head and Neck, The Affiliated Qingdao Third People's Hospital of Qingdao University, Qingdao, Shandong 266021, China.
| | - Ting Wang
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, China.
| |
Collapse
|
9
|
de Souza PB, de Araujo Borba L, Castro de Jesus L, Valverde AP, Gil-Mohapel J, Rodrigues ALS. Major Depressive Disorder and Gut Microbiota: Role of Physical Exercise. Int J Mol Sci 2023; 24:16870. [PMID: 38069198 PMCID: PMC10706777 DOI: 10.3390/ijms242316870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Major depressive disorder (MDD) has a high prevalence and is a major contributor to the global burden of disease. This psychiatric disorder results from a complex interaction between environmental and genetic factors. In recent years, the role of the gut microbiota in brain health has received particular attention, and compelling evidence has shown that patients suffering from depression have gut dysbiosis. Several studies have reported that gut dysbiosis-induced inflammation may cause and/or contribute to the development of depression through dysregulation of the gut-brain axis. Indeed, as a consequence of gut dysbiosis, neuroinflammatory alterations caused by microglial activation together with impairments in neuroplasticity may contribute to the development of depressive symptoms. The modulation of the gut microbiota has been recognized as a potential therapeutic strategy for the management of MMD. In this regard, physical exercise has been shown to positively change microbiota composition and diversity, and this can underlie, at least in part, its antidepressant effects. Given this, the present review will explore the relationship between physical exercise, gut microbiota and depression, with an emphasis on the potential of physical exercise as a non-invasive strategy for modulating the gut microbiota and, through this, regulating the gut-brain axis and alleviating MDD-related symptoms.
Collapse
Affiliation(s)
- Pedro Borges de Souza
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Laura de Araujo Borba
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Louise Castro de Jesus
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Ana Paula Valverde
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S. Rodrigues
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| |
Collapse
|
10
|
Colavitta MF, Barrantes FJ. Therapeutic Strategies Aimed at Improving Neuroplasticity in Alzheimer Disease. Pharmaceutics 2023; 15:2052. [PMID: 37631266 PMCID: PMC10459958 DOI: 10.3390/pharmaceutics15082052] [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: 06/25/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer disease (AD) is the most prevalent form of dementia among elderly people. Owing to its varied and multicausal etiopathology, intervention strategies have been highly diverse. Despite ongoing advances in the field, efficient therapies to mitigate AD symptoms or delay their progression are still of limited scope. Neuroplasticity, in broad terms the ability of the brain to modify its structure in response to external stimulation or damage, has received growing attention as a possible therapeutic target, since the disruption of plastic mechanisms in the brain appear to correlate with various forms of cognitive impairment present in AD patients. Several pre-clinical and clinical studies have attempted to enhance neuroplasticity via different mechanisms, for example, regulating glucose or lipid metabolism, targeting the activity of neurotransmitter systems, or addressing neuroinflammation. In this review, we first describe several structural and functional aspects of neuroplasticity. We then focus on the current status of pharmacological approaches to AD stemming from clinical trials targeting neuroplastic mechanisms in AD patients. This is followed by an analysis of analogous pharmacological interventions in animal models, according to their mechanisms of action.
Collapse
Affiliation(s)
- María F. Colavitta
- Laboratory of Molecular Neurobiology, Biomedical Research Institute (BIOMED), Universidad Católica Argentina (UCA)—National Scientific and Technical Research Council (CONICET), Buenos Aires C1107AAZ, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP-UCA), Facultad de Psicología, Av. Alicia Moreau de Justo, Buenos Aires C1107AAZ, Argentina;
| | - Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute (BIOMED), Universidad Católica Argentina (UCA)—National Scientific and Technical Research Council (CONICET), Buenos Aires C1107AAZ, Argentina
| |
Collapse
|
11
|
Gerasimov E, Bezprozvanny I, Vlasova OL. Activation of Gq-Coupled Receptors in Astrocytes Restores Cognitive Function in Alzheimer's Disease Mice Model. Int J Mol Sci 2023; 24:9969. [PMID: 37373117 PMCID: PMC10298315 DOI: 10.3390/ijms24129969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most widespread neurodegenerative diseases. Most of the current AD therapeutic developments are directed towards improving neuronal cell function or facilitating Aβ amyloid clearance from the brain. However, some recent evidence suggests that astrocytes may play a significant role in the pathogenesis of AD. In this paper, we evaluated the effects of the optogenetic activation of Gq-coupled exogenous receptors expressed in astrocytes as a possible way of restoring brain function in the AD mouse model. We evaluated the effects of the optogenetic activation of astrocytes on long-term potentiation, spinal morphology and behavioral readouts in 5xFAD mouse model of AD. We determined that in vivo chronic activation of astrocytes resulted in the preservation of spine density, increased mushroom spine survival, and improved performance in cognitive behavioral tests. Furthermore, chronic optogenetic stimulation of astrocytes resulted in the elevation of EAAT-2 glutamate uptake transporter expression, which could be a possible explanation for the observed in vivo neuroprotective effects. The obtained results suggest that the persistent activation of astrocytes may be considered a potential therapeutic approach for the treatment of AD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Evgenii Gerasimov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (E.G.); (I.B.)
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (E.G.); (I.B.)
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Olga L. Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (E.G.); (I.B.)
| |
Collapse
|
12
|
Tayanloo-Beik A, Nikkhah A, Alaei S, Goodarzi P, Rezaei-Tavirani M, Mafi AR, Larijani B, Shouroki FF, Arjmand B. Brown adipose tissue and alzheimer's disease. Metab Brain Dis 2023; 38:91-107. [PMID: 36322277 DOI: 10.1007/s11011-022-01097-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/01/2022] [Indexed: 01/12/2023]
Abstract
Alzheimer's disease (AD), the most common type of senile dementia, is a chronic neurodegenerative disease characterized by cognitive dysfunction and behavioral disability. The two histopathological hallmarks in this disease are the extraneuronal accumulation of amyloid-β (Aβ) and the intraneuronal deposition of neurofibrillary tangles (NFTs). Despite this, central and peripheral metabolic dysfunction, such as abnormal brain signaling, insulin resistance, inflammation, and impaired glucose utilization, have been indicated to be correlated with AD. There is solid evidence that the age-associated thermoregulatory deficit induces diverse metabolic changes associated with AD development. Brown adipose tissue (BAT) has been known as a thermoregulatory organ particularly vital during infancy. However, in recent years, BAT has been accepted as an endocrine organ, being involved in various functions that prevent AD, such as regulating energy metabolism, secreting hormones, improving insulin sensitivity, and increasing glucose utilization in adult humans. This review focuses on the mechanisms of BAT activation and the effect of aging on BAT production and signaling. Specifically, the evidence demonstrating the effect of BAT on pathological mechanisms influencing the development of AD, including insulin pathway, thermoregulation, and other hormonal pathways, are reviewed in this article.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirabbas Nikkhah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Alaei
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ahmad Rezazadeh Mafi
- Department of Radiation Oncology, Imam Hossein Hospital, Shaheed Beheshti Medical University, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran.
| | - Fatemeh Fazeli Shouroki
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Jin T, Huang W, Cao F, Yu X, Ying Z, Guo S, Cheng Y, Xu C. Causal association between adiponectin and the risk of Alzheimer's disease: A Mendelian randomization study. Front Neurol 2022; 13:1038975. [PMID: 36570466 PMCID: PMC9780387 DOI: 10.3389/fneur.2022.1038975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Background Numerous observational studies have revealed that circulating adiponectin (ADPN) is associated with Alzheimer's disease (AD) risk. However, the causality remains unknown. We aimed to assess the causality of circulating ADPN on AD risk using Mendelian randomization (MR). Methods Fourteen single nucleotide polymorphisms (SNPs) significantly associated with ADPN were selected from publicly available genetic abstract data. We applied these SNPs to two recent large-scale genome-wide association studies (GWAS) of AD, one from the FinnGen consortium and the other from a large meta-analysis. The inverse variance weighted method, MR-Egger method, the weighted median method, the Cochran Q statistic, the MR-Pleiotropy Residual Sum and Outlier methods, and the leave-one-out analysis were applied for MR analyses. Results In MR analysis, no significant genetic association was found between plasma ADPN levels and AD risk by analyzing the FinnGen consortium GWAS database in the inverse variance weighted method [odds ratio (OR): 0.874, 95% confidence interval (CI): 0.701-1.089, p = 0.230], MR-Egger (OR: 0.944, 95% CI: 0.692-1.288, p = 0.721), and weighted median method (OR: 0.900, 95% CI: 0.678-1.194, p = 0.449). Additionally, the same analysis was conducted for the meta-analysis database, and we found no significant association (OR: 1.000, 95% CI: 0.999-1.001, p = 0.683). Conclusion Our findings reveal no significant causal association between circulating ADPN and AD risk.
Collapse
Affiliation(s)
- Tianyu Jin
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China,Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Wei Huang
- Rheumatism and Immunity Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Fangzheng Cao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Yu
- Alberta Institute, Wenzhou Medical University, Wenzhou, China
| | - Zhenhua Ying
- Rheumatism and Immunity Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Shunyuan Guo
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yifan Cheng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China,Yifan Cheng
| | - Chao Xu
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China,*Correspondence: Chao Xu
| |
Collapse
|
14
|
Chen C, Ma X, Wei J, Shakir N, Zhang JK, Zhang L, Nehme A, Cui Y, Ferguson D, Bai F, Qiu S. Early impairment of cortical circuit plasticity and connectivity in the 5XFAD Alzheimer's disease mouse model. Transl Psychiatry 2022; 12:371. [PMID: 36075886 PMCID: PMC9458752 DOI: 10.1038/s41398-022-02132-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Genetic risk factors for neurodegenerative disorders, such as Alzheimer's disease (AD), are expressed throughout the life span. How these risk factors affect early brain development and function remain largely unclear. Analysis of animal models with high constructive validity for AD, such as the 5xFAD mouse model, may provide insights on potential early neurodevelopmental effects that impinge on adult brain function and age-dependent degeneration. The 5XFAD mouse model over-expresses human amyloid precursor protein (APP) and presenilin 1 (PS1) harboring five familial AD mutations. It is unclear how the expression of these mutant proteins affects early developing brain circuits. We found that the prefrontal cortex (PFC) layer 5 (L5) neurons in 5XFAD mice exhibit transgenic APP overloading at an early post-weaning age. Impaired synaptic plasticity (long-term potentiation, LTP) was seen at 6-8 weeks age in L5 PFC circuit, which was correlated with increased intracellular APP. APP overloading was also seen in L5 pyramidal neurons in the primary visual cortex (V1) during the critical period of plasticity (4-5 weeks age). Whole-cell patch clamp recording in V1 brain slices revealed reduced intrinsic excitability of L5 neurons in 5XFAD mice, along with decreased spontaneous miniature excitatory and inhibitory inputs. Functional circuit mapping using laser scanning photostimulation (LSPS) combined with glutamate uncaging uncovered reduced excitatory synaptic connectivity onto L5 neurons in V1, and a more pronounced reduction in inhibitory connectivity, indicative of altered excitation and inhibition during VC critical period. Lastly, in vivo single-unit recording in V1 confirmed that monocular visual deprivation-induced ocular dominance plasticity during critical period was impaired in 5XFAD mice. Our study reveals plasticity deficits across multiple cortical regions and indicates altered early cortical circuit developmental trajectory as a result of mutant APP/PS1 over-expression.
Collapse
Affiliation(s)
- Chang Chen
- grid.41156.370000 0001 2314 964XDepartment of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210008 China ,grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Xiaokuang Ma
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Jing Wei
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Neha Shakir
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Jessica K. Zhang
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Le Zhang
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Antoine Nehme
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Yuehua Cui
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Deveroux Ferguson
- grid.134563.60000 0001 2168 186XBasic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004 USA
| | - Feng Bai
- Department of Neurology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China.
| | - Shenfeng Qiu
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 85004, USA.
| |
Collapse
|
15
|
Formolo DA, Cheng T, Yu J, Kranz GS, Yau SY. Central Adiponectin Signaling – A Metabolic Regulator in Support of Brain Plasticity. Brain Plast 2022; 8:79-96. [DOI: 10.3233/bpl-220138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 12/18/2022] Open
Abstract
Brain plasticity and metabolism are tightly connected by a constant influx of peripheral glucose to the central nervous system in order to meet the high metabolic demands imposed by neuronal activity. Metabolic disturbances highly affect neuronal plasticity, which underlies the prevalent comorbidity between metabolic disorders, cognitive impairment, and mood dysfunction. Effective pro-cognitive and neuropsychiatric interventions, therefore, should consider the metabolic aspect of brain plasticity to achieve high effectiveness. The adipocyte-secreted hormone, adiponectin, is a metabolic regulator that crosses the blood-brain barrier and modulates neuronal activity in several brain regions, where it exerts neurotrophic and neuroprotective properties. Moreover, adiponectin has been shown to improve neuronal metabolism in different animal models, including obesity, diabetes, and Alzheimer’s disease. Here, we aim at linking the adiponectin’s neurotrophic and neuroprotective properties with its main role as a metabolic regulator and to summarize the possible mechanisms of action on improving brain plasticity via its role in regulating the intracellular energetic activity. Such properties suggest adiponectin signaling as a potential target to counteract the central metabolic disturbances and impaired neuronal plasticity underlying many neuropsychiatric disorders.
Collapse
Affiliation(s)
- Douglas A. Formolo
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University3Institute of future foods
- Research Institute for Smart Ageing (RISA), Hong Kong Polytechnic University
| | - Tong Cheng
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University3Institute of future foods
- Research Institute for Smart Ageing (RISA), Hong Kong Polytechnic University
| | - Jiasui Yu
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University3Institute of future foods
- Research Institute for Smart Ageing (RISA), Hong Kong Polytechnic University
| | - Georg S. Kranz
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University3Institute of future foods
- Research Institute for Smart Ageing (RISA), Hong Kong Polytechnic University
| |
Collapse
|
16
|
Zou Y, Gan CL, Xin Z, Zhang HT, Zhang Q, Lee TH, Pan X, Chen Z. Programmed Cell Death Protein 1 Blockade Reduces Glycogen Synthase Kinase 3β Activity and Tau Hyperphosphorylation in Alzheimer's Disease Mouse Models. Front Cell Dev Biol 2022; 9:769229. [PMID: 34977020 PMCID: PMC8716757 DOI: 10.3389/fcell.2021.769229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/22/2021] [Indexed: 12/03/2022] Open
Abstract
Alzheimer’s disease (AD) is a central nervous system degenerative disease, with no effective treatment to date. Administration of immune checkpoint inhibitors significantly reduces neuronal damage and tau hyperphosphorylation in AD, but the specific mechanism is unclear. Here, we found that programmed cell death-receptor 1 (PD1) and its ligand PDL1 were induced by an intracerebroventricular injection of amyloid-β; they were significantly upregulated in the brains of APP/PS1, 5×FAD mice and in SH-SY5Y-APP cell line compared with control. The PD1 and PDL1 levels positively correlated with the glycogen synthase kinase 3 beta (GSK3β) activity in various AD mouse models, and the PDL1-GSK3β immune complex was found in the brain. The application of PD1-blocking antibody reduced tau hyperphosphorylation and GSK3β activity and prevented memory impairments. Mechanistically, we identified PD1 as a critical regulator of GSK3β activity. These results suggest that the immune regulation of the PD1/PDL1 axis is closely involved in AD.
Collapse
Affiliation(s)
- Yulian Zou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Chen-Ling Gan
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Zhiming Xin
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou, China
| | - Hai-Tao Zhang
- Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, National Health Commission, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qi Zhang
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Tae Ho Lee
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiaodong Pan
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhou Chen
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
17
|
Yan XD, Qu XS, Yin J, Qiao J, Zhang J, Qi JS, Wu MN. Adiponectin Ameliorates Cognitive Behaviors and in vivo Synaptic Plasticity Impairments in 3xTg-AD Mice. J Alzheimers Dis 2021; 85:343-357. [PMID: 34806605 DOI: 10.3233/jad-215063] [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] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cognitive deficit is mainly clinical characteristic of Alzheimer's disease (AD). Recent reports showed adiponectin and its analogues could reverse cognitive impairments, lower amyloid-β protein (Aβ) deposition, and exert anti-inflammatory effects in different APP/PS1 AD model mice mainly exhibiting amyloid plaque pathology. However, the potential in vivo electrophysiological mechanism of adiponectin protecting against cognitive deficits in AD and the neuroprotective effects of adiponectin on 3xTg-AD mice including both plaque and tangle pathology are still unclear. OBJECTIVE To observe the effects of adiponectin treatment on cognitive deficits in 3xTg-AD mice, investigate its potential in vivo electrophysiological mechanism, and testify its anti-inflammatory effects. METHODS Barnes maze test, Morris water maze test, and fear conditioning test were used to evaluate the memory-ameliorating effects of adiponectin on 3xTg-AD mice. In vivo hippocampal electrophysiological recording was used to observe the change of basic synaptic transmission, long-term potentiation, and long-term depression. Immunohistochemistry staining and western blot were used to observe the activation of microglia and astroglia, and the expression levels of proinflammatory factors and anti-inflammtory factor IL-10. RESULTS Adiponectin treatment could alleviate spatial memory and conditioned fear memory deficits observed in 3xTg-AD mice, improve in vivo LTP depression and LTD facilitation, inhibit overactivation of microglia and astroglia, decrease the expression of proinflammatory factors NF- κB and IL-1β, and increase the expression level of IL-10 in the hippocampus of 3xTg-AD mice. CONCLUSION Adiponectin could ameliorate cognitive deficits in 3xTg-AD mice through improving in vivo synaptic plasticity impairments and alleviating neuroinflammation in the hippocampus of 3xTg-AD mice.
Collapse
Affiliation(s)
- Xu-Dong Yan
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Xue-Song Qu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China.,Pomology Institute of Shanxi Agricultural University, Taiyuan, China
| | - Jing Yin
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Jin Qiao
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Jun Zhang
- Functional Laboratory Center, Shanxi Medical University, Taiyuan, China
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
18
|
Liang YY, Zhang LD, Luo X, Wu LL, Chen ZW, Wei GH, Zhang KQ, Du ZA, Li RZ, So KF, Li A. All roads lead to Rome - a review of the potential mechanisms by which exerkines exhibit neuroprotective effects in Alzheimer's disease. Neural Regen Res 2021; 17:1210-1227. [PMID: 34782555 PMCID: PMC8643060 DOI: 10.4103/1673-5374.325012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Age-related neurodegenerative disorders such as Alzheimer’s disease (AD) have become a critical public health issue due to the significantly extended human lifespan, leading to considerable economic and social burdens. Traditional therapies for AD such as medicine and surgery remain ineffective, impractical, and expensive. Many studies have shown that a variety of bioactive substances released by physical exercise (called “exerkines”) help to maintain and improve the normal functions of the brain in terms of cognition, emotion, and psychomotor coordination. Increasing evidence suggests that exerkines may exert beneficial effects in AD as well. This review summarizes the neuroprotective effects of exerkines in AD, focusing on the underlying molecular mechanism and the dynamic expression of exerkines after physical exercise. The findings described in this review will help direct research into novel targets for the treatment of AD and develop customized exercise therapy for individuals of different ages, genders, and health conditions.
Collapse
Affiliation(s)
- Yi-Yao Liang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education, Guangzhou, Guangdong Province, China
| | - Li-Dan Zhang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education, Guangzhou, Guangdong Province, China
| | - Xi Luo
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education, Guangzhou, Guangdong Province, China
| | - Li-Li Wu
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-sen University; Guangdong Key Laboratory of Liver Disease Research, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhao-Wei Chen
- Department of Clinical Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Guang-Hao Wei
- Department of Clinical Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Kai-Qing Zhang
- Department of Clinical Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Ze-An Du
- Department of Clinical Medicine, International School, Jinan University, Guangzhou, Guangdong Province, China
| | - Ren-Zhi Li
- International Department of the Affiliated High School of South China Normal University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong Province; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University; Key Laboratory of CNS Regeneration (Jinan University), Ministry of Education; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong Province, China
| |
Collapse
|
19
|
Lopez-Vilaret KM, Cantero JL, Fernandez-Alvarez M, Calero M, Calero O, Lindín M, Zurrón M, Díaz F, Atienza M. Impaired glucose metabolism reduces the neuroprotective action of adipocytokines in cognitively normal older adults with insulin resistance. Aging (Albany NY) 2021; 13:23936-23952. [PMID: 34731089 PMCID: PMC8610113 DOI: 10.18632/aging.203668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022]
Abstract
Evidence suggests that aging-related dysfunctions of adipose tissue and metabolic disturbances increase the risk of diabetes and metabolic syndrome (MtbS), eventually leading to cognitive impairment and dementia. However, the neuroprotective role of adipocytokines in this process has not been specifically investigated. The present study aims to identify metabolic alterations that may prevent adipocytokines from exerting their neuroprotective action in normal ageing. We hypothesize that neuroprotection may occur under insulin resistance (IR) conditions as long as there are no other metabolic alterations that indirectly impair the action of adipocytokines, such as hyperglycemia. This hypothesis was tested in 239 cognitively normal older adults (149 females) aged 52 to 87 years (67.4 ± 5.9 yr). We assessed whether the homeostasis model assessment-estimated insulin resistance (HOMA-IR) and the presence of different components of MtbS moderated the association of plasma adipocytokines (i.e., adiponectin, leptin and the adiponectin to leptin [Ad/L] ratio) with cognitive functioning and cortical thickness. The results showed that HOMA-IR, circulating triglyceride and glucose levels moderated the neuroprotective effect of adipocytokines. In particular, elevated triglyceride levels reduced the beneficial effect of Ad/L ratio on cognitive functioning in insulin-sensitive individuals; whereas under high IR conditions, it was elevated glucose levels that weakened the association of the Ad/L ratio with cognitive functioning and with cortical thickness of prefrontal regions. Taken together, these findings suggest that the neuroprotective action of adipocytokines is conditioned not only by whether cognitively normal older adults are insulin-sensitive or not, but also by the circulating levels of triglycerides and glucose, respectively.
Collapse
Affiliation(s)
| | - Jose L Cantero
- Laboratory of Functional Neuroscience, Universidad Pablo de Olavide, Seville, Spain.,CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Marina Fernandez-Alvarez
- Laboratory of Functional Neuroscience, Universidad Pablo de Olavide, Seville, Spain.,CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Miguel Calero
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain.,Chronic Disease Programme, Instituto de Salud Carlos III, Madrid, Spain
| | - Olga Calero
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain.,Chronic Disease Programme, Instituto de Salud Carlos III, Madrid, Spain
| | - Mónica Lindín
- Cognitive Neuroscience Laboratory, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Montserrat Zurrón
- Cognitive Neuroscience Laboratory, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Fernando Díaz
- Cognitive Neuroscience Laboratory, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Universidad Pablo de Olavide, Seville, Spain.,CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| |
Collapse
|
20
|
Lee TH, Christie BR, Lin K, Siu PMF, Zhang L, Yuan TF, Komal P, Xu A, So KF, Yau SY. Chronic AdipoRon Treatment Mimics the Effects of Physical Exercise on Restoring Hippocampal Neuroplasticity in Diabetic Mice. Mol Neurobiol 2021; 58:4666-4681. [PMID: 34164760 PMCID: PMC8487422 DOI: 10.1007/s12035-021-02441-7] [Citation(s) in RCA: 20] [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: 03/24/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Administration of exercise mimetic drugs could be a novel therapeutic approach to combat comorbid neurodegeneration and metabolic syndromes. Adiponectin is an adipocyte-secreted hormone. In addition to its antidiabetic effect, adiponectin mediates the antidepressant effect of physical exercise associated with adult hippocampal neurogenesis. The antidiabetic effect of the adiponectin receptor agonist AdipoRon has been demonstrated, but its potential pro-cognitive and neurotrophic effects in the hippocampus under diabetic condition are still unclear. This study reported that chronic AdipoRon treatment for 2 weeks improved hippocampal-dependent spatial recognition memory in streptozotocin-induced diabetic mice. Besides, AdipoRon treatment increased progenitor cell proliferation and neuronal differentiation in the hippocampal dentate gyrus (DG) of diabetic mice. Furthermore, AdipoRon treatment significantly increased dendritic complexity, spine density, and N-methyl-D-aspartate receptor-dependent long-term potentiation (LTP) in the dentate region, and increased BDNF levels in the DG of diabetic mice. AdipoRon treatment activated AMPK/PGC-1α signalling in the DG, whereas increases in cell proliferation and LTP were not observed when PGC-1α signalling was pharmacologically inhibited. In sum, chronic AdipoRon treatment partially mimics the benefits of physical exercise for learning and memory and hippocampal neuroplasticity in the diabetic brain. The results suggested that AdipoRon could be a potential physical exercise mimetic to improve hippocampal plasticity and hence rescue learning and memory impairment typically associated with diabetes.
Collapse
Affiliation(s)
- Thomas H Lee
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, 11 Yuk Choi Road, Hung Hom, Hong Kong SAR
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kangguang Lin
- Department of Affective Disorder, Guangzhou Brain Hospital, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Parco Ming-Fai Siu
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Li Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Ti-Fei Yuan
- Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pragya Komal
- Department of Biological Sciences, Birla Institute of Technology and Sciences (BITS-Pilani Hyderabad), Hyderabad, India
| | - Aimin Xu
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Kwok-Fai So
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Brain and Cognitive Science, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, 11 Yuk Choi Road, Hung Hom, Hong Kong SAR.
- University Research Facility in Behavioral and Systems Neuroscience, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR.
| |
Collapse
|
21
|
Tang QY, Li M, Chen L, Jiang JM, Gao SL, Xiao F, Zou W, Zhang P, Chen YJ. Adiponectin Mediates the Protection of H 2S Against Chronic Restraint Stress-Induced Cognitive Impairment via Attenuating Hippocampal Damage. Front Behav Neurosci 2021; 15:623644. [PMID: 34025367 PMCID: PMC8131522 DOI: 10.3389/fnbeh.2021.623644] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/07/2021] [Indexed: 01/11/2023] Open
Abstract
Emerging evidence shows that chronic restraint stress (CRS) can induce cognitive dysfunction, which involves in hippocampal damage. Our recent research reveals that hydrogen sulfide (H2S), a novel gasotransmitter, protects against CRS-induced cognitive impairment, but the underlying mechanism remains unclear. Adiponectin, the most abundant plasma adipokine, has been shown to elicit neuroprotective property and attenuate cognitive impairment. Hence, the present work was aimed to explore whether adiponectin mediates the protective effect of H2S on CRS-induced cognitive impairment by inhibiting hippocampal damage. Results found that administration of Anti-Acrp30, a neutralizing antibody of adiponectin, obviously reverses sodium hydrosulfide (NaHS, an exogenous H2S donor)-induced the inhibition on CRS-induced cognitive impairment according to Y-maze test, Novel object recognition (NOR) test, and Morris water maze (MWM) test. In addition, Anti-Acrp30 blocked the protective effect of NaHS on hippocampal apoptosis in rats-subjected with CRS as evidenced by the pathological changes in hippocampus tissues in hematoxylin and eosin (HE) staining and the increases in the amount of the condensed and stained to yellowish-brown or brownish yellow neuron nucleuses in terminal deoxynucleotidyl transferase transfer-mediated dUTP nick end-labeling (TUNEL) staining as well as the expression of hippocampal pro-apoptotic protein (Bax), and a decrease in the expression of hippocampal anti-apoptotic protein (Bcl-2). Furthermore, Anti-Acrp30 mitigated the inhibitory effect of NaHS on CRS-induced oxidative stress as illustrated by the up-regulation of malondialdehyde (MDA) content and the down-regulation of superoxide dismutase (SOD) activity and glutathione (GSH) level in the hippocampus. Moreover, Anti-Acrp30 eliminated NaHS-induced the reduction of endoplasmic reticulum (ER) stress-related proteins including binding immunoglobulin protein (BIP), C/EBP homologous protein (CHOP), and Cleaved Caspase-12 expressions in the hippocampus of rats-exposed to CRS. Taken together, these results indicated that adiponectin mediates the protection of H2S against CRS-induced cognitive impairment through ameliorating hippocampal damage.
Collapse
Affiliation(s)
- Qiong-Yan Tang
- Department of Neurology, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Min Li
- Department of Neurology, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Lei Chen
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Jia-Mei Jiang
- Institute of Neurology, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Sheng-Lan Gao
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Fan Xiao
- Department of Neurology, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Wei Zou
- Department of Neurology, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Ping Zhang
- Department of Neurology, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Yong-Jun Chen
- Department of Neurology, The Affiliated Nanhua Hospital, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
22
|
Wang M, Yoon G, Song J, Jo J. Exendin-4 improves long-term potentiation and neuronal dendritic growth in vivo and in vitro obesity condition. Sci Rep 2021; 11:8326. [PMID: 33859286 PMCID: PMC8050263 DOI: 10.1038/s41598-021-87809-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Metabolic syndrome, which increases the risk of obesity and type 2 diabetes has emerged as a significant issue worldwide. Recent studies have highlighted the relationship between metabolic imbalance and neurological pathologies such as memory loss. Glucagon-like peptide 1 (GLP-1) secreted from gut L-cells and specific brain nuclei plays multiple roles including regulation of insulin sensitivity, inflammation and synaptic plasticity. Although GLP-1 and GLP-1 receptor agonists appear to have neuroprotective function, the specific mechanism of their action in brain remains unclear. We investigated whether exendin-4, as a GLP-1RA, improves cognitive function and brain insulin resistance in metabolic-imbalanced mice fed a high-fat diet. Considering the result of electrophysiological experiments, exendin-4 inhibits the reduction of long term potentiation (LTP) in high fat diet mouse brain. Further, we identified the neuroprotective effect of exendin-4 in primary cultured hippocampal and cortical neurons in in vitro metabolic imbalanced condition. Our results showed the improvement of IRS-1 phosphorylation, neuronal complexity, and the mature of dendritic spine shape by exendin-4 treatment in metabolic imbalanced in vitro condition. Here, we provides significant evidences on the effect of exendin-4 on synaptic plasticity, long-term potentiation, and neural structure. We suggest that GLP-1 is important to treat neuropathology caused by metabolic syndrome.
Collapse
Affiliation(s)
- Ming Wang
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 seoyangro, Hwasun, 58128, Republic of Korea
| | - Gwangho Yoon
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 seoyangro, Hwasun, 58128, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-do, 58128, Republic of Korea
| | - Juhyun Song
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 seoyangro, Hwasun, 58128, Republic of Korea.
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-do, 58128, Republic of Korea.
| | - Jihoon Jo
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, 264 seoyangro, Hwasun, 58128, Republic of Korea.
- NeuroMedical Convergence Lab, Biomedical Research Institute, Chonnam National University Hospital, Jebong-ro, Gwangju, 501-757, Republic of Korea.
- Department of Neurology, Chonnam National University Medical School, Gwangju, 501-757, Republic of Korea.
| |
Collapse
|
23
|
Lee TH, Christie BR, van Praag H, Lin K, Siu PMF, Xu A, So KF, Yau SY. AdipoRon Treatment Induces a Dose-Dependent Response in Adult Hippocampal Neurogenesis. Int J Mol Sci 2021; 22:2068. [PMID: 33669795 PMCID: PMC7922380 DOI: 10.3390/ijms22042068] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/28/2022] Open
Abstract
AdipoRon, an adiponectin receptor agonist, elicits similar antidiabetic, anti-atherogenic, and anti-inflammatory effects on mouse models as adiponectin does. Since AdipoRon can cross the blood-brain barrier, its chronic effects on regulating hippocampal function are yet to be examined. This study investigated whether AdipoRon treatment promotes hippocampal neurogenesis and spatial recognition memory in a dose-dependent manner. Adolescent male C57BL/6J mice received continuous treatment of either 20 mg/kg (low dose) or 50 mg/kg (high dose) AdipoRon or vehicle intraperitoneally for 14 days, followed by the open field test to examine anxiety and locomotor activity, and the Y maze test to examine hippocampal-dependent spatial recognition memory. Immunopositive cell markers of neural progenitor cells, immature neurons, and newborn cells in the hippocampal dentate gyrus were quantified. Immunosorbent assays were used to measure the serum levels of factors that can regulate hippocampal neurogenesis, including adiponectin, brain-derived neurotrophic factor (BDNF), and corticosterone. Our results showed that 20 mg/kg AdipoRon treatment significantly promoted hippocampal cell proliferation and increased serum levels of adiponectin and BDNF, though there were no effects on spatial recognition memory and locomotor activity. On the contrary, 50 mg/kg AdipoRon treatment impaired spatial recognition memory, suppressed cell proliferation, neuronal differentiation, and cell survival associated with reduced serum levels of BDNF and adiponectin. The results suggest that a low-dose AdipoRon treatment promotes hippocampal cell proliferation, while a high-dose AdipoRon treatment is detrimental to the hippocampus function.
Collapse
Affiliation(s)
- Thomas H. Lee
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong;
| | - Brian R. Christie
- Division of Biomedical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Henriette van Praag
- FAU Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33431, USA;
| | - Kangguang Lin
- Department of Affective Disorder, Guangzhou Brain Hospital, The Brain Affiliated Hospital of Guangzhou Medical University, Guangzhou 510370, China;
| | - Parco Ming-Fai Siu
- Division of Kinesiology, School of Public Health, The University of Hong Kong, Hong Kong;
| | - Aimin Xu
- Department of Medicine, The University of Hong Kong, Hong Kong;
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
- The State Key Laboratory of Pharmacology, The University of Hong Kong, Hong Kong
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China;
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
- Department of Ophthalmology, The University of Hong Kong, Hong Kong
| | - Suk-yu Yau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong;
| |
Collapse
|
24
|
Kim JY, Barua S, Jeong YJ, Lee JE. Adiponectin: The Potential Regulator and Therapeutic Target of Obesity and Alzheimer's Disease. Int J Mol Sci 2020; 21:6419. [PMID: 32899357 PMCID: PMC7504582 DOI: 10.3390/ijms21176419] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 02/08/2023] Open
Abstract
Animal and human mechanistic studies have consistently shown an association between obesity and Alzheimer's disease (AD). AD, a degenerative brain disease, is the most common cause of dementia and is characterized by the presence of extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles disposition. Some studies have recently demonstrated that Aβ and tau cannot fully explain the pathophysiological development of AD and that metabolic disease factors, such as insulin, adiponectin, and antioxidants, are important for the sporadic onset of nongenetic AD. Obesity prevention and treatment can be an efficacious and safe approach to AD prevention. Adiponectin is a benign adipokine that sensitizes the insulin receptor signaling pathway and suppresses inflammation. It has been shown to be inversely correlated with adipose tissue dysfunction and may enhance the risk of AD because a range of neuroprotection adiponectin mechanisms is related to AD pathology alleviation. In this study, we summarize the recent progress that addresses the beneficial effects and potential mechanisms of adiponectin in AD. Furthermore, we review recent studies on the diverse medications of adiponectin that could possibly be related to AD treatment, with a focus on their association with adiponectin. A better understanding of the neuroprotection roles of adiponectin will help clarify the precise underlying mechanism of AD development and progression.
Collapse
Affiliation(s)
- Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
| | - Sumit Barua
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
| | - Ye Jun Jeong
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
- BK21 Plus Project for Medical Sciences, and Brain Research Institute, Yonsei University College of Medicine, Seoul 120-752, Korea
| |
Collapse
|
25
|
King MR, Anderson NJ, Deciu M, Guernsey LS, Cundiff M, Hajizadeh S, Jolivalt CG. Insulin deficiency, but not resistance, exaggerates cognitive deficits in transgenic mice expressing human amyloid and tau proteins. Reversal by Exendin-4 treatment. J Neurosci Res 2020; 98:2357-2369. [PMID: 32737929 DOI: 10.1002/jnr.24706] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 06/29/2020] [Accepted: 07/12/2020] [Indexed: 12/24/2022]
Abstract
Epidemiological studies have pointed at diabetes as a risk factor for Alzheimer's disease (AD) and this has been supported by several studies in animal models of both type 1 and type 2 diabetes. However, side-by-side comparison of the two types of diabetes is limited. We investigated the role of insulin deficiency and insulin resistance in the development of memory impairments and the effect of Exendin-4 (Ex4) treatment in a mouse model of AD. Three-4-month-old female wild type (WT) mice and mice overexpressing human tau and amyloid precursor protein (TAPP) were injected with streptozotocin (STZ) or fed a high-fat diet (HFD). A second study was performed in TAPP-STZ mice treated with Ex4, a long-lasting analog of GLP-1. Plasma and brain were collected at study termination for ELISA, Western blot, and immunohistochemistry analysis. Learning and memory deficits were impaired in TAPP transgenic mice compared with WT mice at the end of the study. Deficits were exaggerated by insulin deficiency in TAPP mice but 12 weeks of insulin resistance did not affect memory performances in either WT or TAPP mice. Levels of phosphorylated tau were increased in the brain of WT-STZ and TAPP-STZ mice but not in the brain of WT or TAPP mice on HFD. In the TAPP-STZ mice, treatment with Ex4 initiated after established cognitive deficits ameliorated learning, but not memory, impairments. This was accompanied by the reduction of amyloid β and phosphorylated tau expression. Theses studies support the role of Ex4 in AD, independently from its actions on diabetes.
Collapse
Affiliation(s)
- Matthew R King
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Nicholas J Anderson
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Mihaela Deciu
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Lucie S Guernsey
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Morgan Cundiff
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Shohreh Hajizadeh
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Corinne G Jolivalt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Cai M, Lee JH, Yang EJ. Electroacupuncture attenuates cognition impairment via anti-neuroinflammation in an Alzheimer's disease animal model. J Neuroinflammation 2019; 16:264. [PMID: 31836020 PMCID: PMC6909515 DOI: 10.1186/s12974-019-1665-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressive loss of cognitive abilities and memory leading to dementia. Electroacupuncture (EA) is a complementary alternative medicine approach, applying an electrical current to acupuncture points. In clinical and animal studies, EA causes cognitive improvements in AD and vascular dementia. However, EA-induced changes in cognition and microglia-mediated amyloid β (Aβ) degradation have not been determined yet in AD animals. Therefore, this study investigated the EA-induced molecular mechanisms causing cognitive improvement and anti-inflammatory activity in five familial mutation (5XFAD) mice, an animal model of AD. Methods 5XFAD mice were bilaterally treated with EA at the Taegye (KI3) acupoints three times per week for 2 weeks. To evaluate the effects of EA treatment on cognitive functions, novel object recognition and Y-maze tests were performed with non-Tg, 5XFAD (Tg), and EA-treated 5XFAD (Tg + KI3) mice. To examine the molecular mechanisms underlying EA effects, western blots, immunohistochemistry, and micro-positron emission tomography scans were performed. Furthermore, we studied synapse ultrastructures with transmission electron microscopy and used electrophysiology to investigate EA effects on synaptic plasticity in 5XFAD mice. Results EA treatment significantly improved working memory and synaptic plasticity, alleviated neuroinflammation, and reduced ultrastructural degradation of synapses via upregulation of synaptophysin and postsynaptic density-95 protein in 5XFAD mice. Furthermore, microglia-mediated Aβ deposition was reduced after EA treatment and coincided with a reduction in amyloid precursor protein. Conclusions Our findings demonstrate that EA treatment ameliorates cognitive impairment via inhibition of synaptic degeneration and neuroinflammation in a mouse model of AD.
Collapse
Affiliation(s)
- Mudan Cai
- Department of Herbal Medicine Research, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 305-811, Republic of Korea
| | - Jun-Hwan Lee
- Department of Clinical Research, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 305-811, Republic of Korea
| | - Eun Jin Yang
- Department of Clinical Research, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 305-811, Republic of Korea.
| |
Collapse
|
27
|
The Novel Perspectives of Adipokines on Brain Health. Int J Mol Sci 2019; 20:ijms20225638. [PMID: 31718027 PMCID: PMC6887733 DOI: 10.3390/ijms20225638] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
First seen as a fat-storage tissue, the adipose tissue is considered as a critical player in the endocrine system. Precisely, adipose tissue can produce an array of bioactive factors, including cytokines, lipids, and extracellular vesicles, which target various systemic organ systems to regulate metabolism, homeostasis, and immune response. The global effects of adipokines on metabolic events are well defined, but their impacts on brain function and pathology remain poorly defined. Receptors of adipokines are widely expressed in the brain. Mounting evidence has shown that leptin and adiponectin can cross the blood–brain barrier, while evidence for newly identified adipokines is limited. Significantly, adipocyte secretion is liable to nutritional and metabolic states, where defective circuitry, impaired neuroplasticity, and elevated neuroinflammation are symptomatic. Essentially, neurotrophic and anti-inflammatory properties of adipokines underlie their neuroprotective roles in neurodegenerative diseases. Besides, adipocyte-secreted lipids in the bloodstream can act endocrine on the distant organs. In this article, we have reviewed five adipokines (leptin, adiponectin, chemerin, apelin, visfatin) and two lipokines (palmitoleic acid and lysophosphatidic acid) on their roles involving in eating behavior, neurotrophic and neuroprotective factors in the brain. Understanding and regulating these adipokines can lead to novel therapeutic strategies to counteract metabolic associated eating disorders and neurodegenerative diseases, thus promote brain health.
Collapse
|
28
|
Bloemer J, Pinky PD, Smith WD, Bhattacharya D, Chauhan A, Govindarajulu M, Hong H, Dhanasekaran M, Judd R, Amin RH, Reed MN, Suppiramaniam V. Adiponectin Knockout Mice Display Cognitive and Synaptic Deficits. Front Endocrinol (Lausanne) 2019; 10:819. [PMID: 31824431 PMCID: PMC6886372 DOI: 10.3389/fendo.2019.00819] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022] Open
Abstract
Adiponectin is an adipokine that has recently been under investigation for potential neuroprotective effects in various brain disorders including Alzheimer's disease, stroke, and depression. Adiponectin receptors (AdipoR1 and AdipoR2) are found throughout various brain regions, including the hippocampus. However, the role of these receptors in synaptic and cognitive function is not clear. Therefore, the goal of the current study was to evaluate synaptic and cognitive function in the absence of adiponectin. The current study utilized 12-month-old adiponectin knockout (APN-KO) mice and age-matched controls to study cognitive and hippocampal synaptic alterations. We determined that AdipoR1 and AdipoR2 are present in the synaptosome, with AdipoR2 displaying increased presynaptic vs. postsynaptic localization, whereas AdipoR1 was enriched in both the presynaptic and postsynaptic fractions. APN-KO mice displayed cognitive deficits in the novel object recognition (NOR) and Y-maze tests. This was mirrored by deficits in long-term potentiation (LTP) of the hippocampal Schaefer collateral pathway in APN-KO mice. APN-KO mice also displayed a reduction in basal synaptic transmission and an increase in presynaptic release probability. Deficits in LTP were rescued through hippocampal slice incubation with the adiponectin receptor agonist, AdipoRon, indicating that acute alterations in adiponectin receptor signaling influence synaptic function. Along with the deficits in LTP, altered levels of key presynaptic and postsynaptic proteins involved in glutamatergic neurotransmission were observed in APN-KO mice. Taken together, these results indicate that adiponectin is an important regulator of cognition and synaptic function in the hippocampus. Future studies should examine the role of specific adiponectin receptors in synaptic processes.
Collapse
Affiliation(s)
- Jenna Bloemer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Warren D. Smith
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Dwipayan Bhattacharya
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Alisa Chauhan
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Hao Hong
- Center for Neuroscience, Auburn University, Auburn, AL, United States
- Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Robert Judd
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Rajesh H. Amin
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
- *Correspondence: Miranda N. Reed
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
- Center for Neuroscience, Auburn University, Auburn, AL, United States
- Vishnu Suppiramaniam
| |
Collapse
|