1
|
Xia L, Qiu Y, Li J, Xu M, Dong Z. The Potential Role of Artemisinins Against Neurodegenerative Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024:1-20. [PMID: 39343990 DOI: 10.1142/s0192415x24500642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Artemisinin (ART) and its derivatives, collectively referred to as artemisinins (ARTs), have been approved for the treatment of malaria for decades. ARTs are converted into dihydroartemisinin (DHA), the only active form, which is reductive in vivo. In this review, we provide a brief overview of the neuroprotective potential of ARTs and the underlying mechanisms on several of the most common neurodegenerative diseases, particularly considering their potential application in those associated with cognitive and motor impairments including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). ARTs act as autophagy balancers to alleviate AD and PD. They inhibit neuroinflammatory responses by regulating phosphorylation of signal transduction proteins, such as AKT, PI3K, ERK, NF-[Formula: see text]B, p38 MAPK, I[Formula: see text]B[Formula: see text]. In addition, ARTs regulate GABAergic signaling in a dose-dependent manner. Although they competitively inhibit the binding of gephyrin to GABAergic receptors, low doses of ARTs enhance GABAergic signaling. ARTs can also inhibit ferroptosis, activate the Akt/Bcl-2, AMPK, or ERK/CREB pathways to reduce oxidative stress, and maintain mitochondrial homeostasis, protecting neurons from oxidative stress injury. More importantly, ARTs structurally combine with and suppress [Formula: see text]-Amyloid (A[Formula: see text]-induced neurotoxicity, reduce P-tau, and maintain O-GlcNAcylation/Phosphorylation balance, leading to relieved pathological changes in neurodegenerative diseases. Collectively, these natural properties endow ARTs with unique potential for application in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lei Xia
- Growth, Development, and Mental Health of Children and Adolescence Center Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders National Clinical Research Center for Child Health and Disorders Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| | - Yiqiong Qiu
- Medical Laboratory of Changshou District Hospital of Traditional Chinese Medicine Chongqing 401220, P. R. China
| | - Junjie Li
- Growth, Development, and Mental Health of Children and Adolescence Center Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders National Clinical Research Center for Child Health and Disorders Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| | - Mingliang Xu
- Growth, Development, and Mental Health of Children and Adolescence Center Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders National Clinical Research Center for Child Health and Disorders Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| | - Zhifang Dong
- Growth, Development, and Mental Health of Children and Adolescence Center Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders National Clinical Research Center for Child Health and Disorders Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders Children's Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| |
Collapse
|
2
|
Di Domenico F, Lanzillotta C, Perluigi M. Redox imbalance and metabolic defects in the context of Alzheimer disease. FEBS Lett 2024; 598:2047-2066. [PMID: 38472147 DOI: 10.1002/1873-3468.14840] [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: 10/17/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
Redox reactions play a critical role for intracellular processes, including pathways involved in metabolism and signaling. Reactive oxygen species (ROS) act either as second messengers or generators of protein modifications, fundamental mechanisms for signal transduction. Disturbance of redox homeostasis is associated with many disorders. Among these, Alzheimer's disease is a neurodegenerative pathology that presents hallmarks of oxidative damage such as increased ROS production, decreased activity of antioxidant enzymes, oxidative modifications of macromolecules, and changes in mitochondrial homeostasis. Interestingly, alteration of redox homeostasis is closely associated with defects of energy metabolism, involving both carbohydrates and lipids, the major energy fuels for the cell. As the brain relies exclusively on glucose metabolism, defects of glucose utilization represent a harmful event for the brain. During aging, a progressive perturbation of energy metabolism occurs resulting in brain hypometabolism. This condition contributes to increase neuronal cell vulnerability ultimately resulting in cognitive impairment. The current review discusses the crosstalk between alteration of redox homeostasis and brain energy defects that seems to act in concert in promoting Alzheimer's neurodegeneration.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
3
|
Hroudová J, Fišar Z. Alzheimer's disease approaches - Focusing on pathology, biomarkers and clinical trial candidates. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111069. [PMID: 38917881 DOI: 10.1016/j.pnpbp.2024.111069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024]
Abstract
The strategy for the development of new drugs for Alzheimer's disease (AD) recognizes that an effective therapy requires early therapeutic intervention and a multifactorial approach that considers the individual initiators of AD development. Current knowledge of AD includes the understanding of pathophysiology, risk factors, biomarkers, and the evolving patterns of biomarker abnormalities. This knowledge is essential in identifying potential molecular targets for new drug development. This review summarizes promising AD drug candidates, many of which are currently in phase 2 or 3 clinical trials. New agents are classified according to the Common Alzheimer's Disease Research Ontology (CADRO). The main targets of new drugs for AD are processes related to amyloid beta and tau neurotoxicity, neurotransmission, inflammation, metabolism and bioenergetics, synaptic plasticity, and oxidative stress. These interventions are aimed at preventing disease onset and slowing or eliminating disease progression. The efficacy of pharmacotherapy may be enhanced by combining these drugs with other treatments, antioxidants, and dietary supplements. Ongoing research into AD pathophysiology, risk factors, biomarkers, and the dynamics of biomarker abnormalities may contribute to the understanding of AD and offer hope for effective therapeutic strategies in the near future.
Collapse
Affiliation(s)
- Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| |
Collapse
|
4
|
Majumder M, Dutta D. Oligodendrocyte Dysfunction in Tauopathy: A Less Explored Area in Tau-Mediated Neurodegeneration. Cells 2024; 13:1112. [PMID: 38994964 PMCID: PMC11240328 DOI: 10.3390/cells13131112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Aggregation of the microtubule-associated protein tau (MAPT) is the hallmark pathology in a spectrum of neurodegenerative disorders collectively called tauopathies. Physiologically, tau is an inherent neuronal protein that plays an important role in the assembly of microtubules and axonal transport. However, disease-associated mutations of this protein reduce its binding to the microtubule components and promote self-aggregation, leading to formation of tangles in neurons. Tau is also expressed in oligodendrocytes, where it has significant developmental roles in oligodendrocyte maturation and myelin synthesis. Oligodendrocyte-specific tau pathology, in the form of fibrils and coiled coils, is evident in major tauopathies including progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick's disease (PiD). Multiple animal models of tauopathy expressing mutant forms of MAPT recapitulate oligodendroglial tau inclusions with potential to cause degeneration/malfunction of oligodendrocytes and affecting the neuronal myelin sheath. Till now, mechanistic studies heavily concentrated on elucidating neuronal tau pathology. Therefore, more investigations are warranted to comprehensively address tau-induced pathologies in oligodendrocytes. The present review provides the current knowledge available in the literature about the intricate relations between tau and oligodendrocytes in health and diseases.
Collapse
Affiliation(s)
- Moumita Majumder
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Debashis Dutta
- Department of Pediatrics, Darby’s Children Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
5
|
El Hajjar L, Page A, Bridot C, Cantrelle FX, Landrieu I, Smet-Nocca C. Regulation of Glycogen Synthase Kinase-3β by Phosphorylation and O-β-Linked N-Acetylglucosaminylation: Implications on Tau Protein Phosphorylation. Biochemistry 2024; 63:1513-1533. [PMID: 38788673 DOI: 10.1021/acs.biochem.4c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Glycogen synthase kinase 3 (GSK3) plays a pivotal role in signaling pathways involved in insulin metabolism and the pathogenesis of neurodegenerative disorders. In particular, the GSK3β isoform is implicated in Alzheimer's disease (AD) as one of the key kinases involved in the hyperphosphorylation of tau protein, one of the neuropathological hallmarks of AD. As a constitutively active serine/threonine kinase, GSK3 is inactivated by Akt/PKB-mediated phosphorylation of Ser9 in the N-terminal disordered domain, and for most of its substrates, requires priming (prephosphorylation) by another kinase that targets the substrate to a phosphate-specific pocket near the active site. GSK3 has also been shown to be post-translationally modified by O-linked β-N-acetylglucosaminylation (O-GlcNAcylation), with still unknown functions. Here, we have found that binding of Akt inhibits GSK3β kinase activity on both primed and unprimed tau substrates. Akt-mediated Ser9 phosphorylation restores the GSK3β kinase activity only on primed tau, thereby selectively inactivating GSK3β toward unprimed tau protein. Additionally, we have shown that GSK3β is highly O-GlcNAcylated at multiple sites within the kinase domain and the disordered N- and C-terminal domains, including Ser9. In contrast to Akt-mediated regulation, neither the O-GlcNAc transferase nor O-GlcNAcylation significantly alters GSK3β kinase activity, but high O-GlcNAc levels reduce Ser9 phosphorylation by Akt. Reciprocally, Akt phosphorylation downregulates the overall O-GlcNAcylation of GSK3β, indicating a crosstalk between both post-translational modifications. Our results indicate that specific O-GlcNAc profiles may be involved in the phosphorylation-dependent Akt-mediated regulation of GSK3β kinase activity.
Collapse
Affiliation(s)
- Léa El Hajjar
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Adeline Page
- Protein Science Facility, SFR Biosciences Univ Lyon, ENS de Lyon, CNRS UAR3444, Inserm US8, Université Claude Bernard Lyon 1, 50 Avenue Tony Garnier, Lyon F-69007, France
| | - Clarisse Bridot
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - François-Xavier Cantrelle
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Isabelle Landrieu
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Caroline Smet-Nocca
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| |
Collapse
|
6
|
de Veij Mestdagh CF, Witte ME, Scheper W, Smit AB, Henning RH, van Kesteren RE. Torpor induces reversible tau hyperphosphorylation and accumulation in mice expressing human tau. Acta Neuropathol Commun 2024; 12:86. [PMID: 38835043 PMCID: PMC11149198 DOI: 10.1186/s40478-024-01800-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Tau protein hyperphosphorylation and aggregation are key pathological events in neurodegenerative tauopathies such as Alzheimer's disease. Interestingly, seasonal hibernators show extensive tau hyperphosphorylation during torpor, i.e., the hypothermic and hypometabolic state of hibernation, which is completely reversed during arousal. Torpor-associated mechanisms that reverse tau hyperphosphorylation may be of therapeutic relevance, however, it is currently not known to what extent they apply to human tau. Here we addressed this issue using daily torpor in wildtype mice that express mouse tau (mtau) and in mice that lack mtau expression and instead express human tau (htau). AT8, AT100 and Ser396 immunoblotting and immunohistochemistry were used to assess tau (hyper)phosphorylation at clinically relevant phosphorylation sites. We found that torpor robustly and reversibly increases the levels of phosphorylated tau in both mtau and htau mice. Immunohistochemistry revealed four brain areas that show prominent tau phosphorylation: the hippocampus, posterior parietal cortex, piriform cortex and cortical amygdala. Whereas wildtype mice primarily showed increased levels of diffusely organized hyperphosphorylated tau during torpor, htau mice contained clear somato-dendritic accumulations of AT8 reactivity resembling tau pre-tangles as observed in the Alzheimer brain. Interestingly, AT8-positive accumulations disappeared upon arousal, and tau phosphorylation levels at 24 h after arousal were lower than observed at baseline, suggesting a beneficial effect of torpor-arousal cycles on preexisting hyperphosphorylated tau. In conclusion, daily torpor in mice offers a quick and standardized method to study tau phosphorylation, accumulation and clearance in mouse models relevant for neurodegeneration, as well as opportunities to discover new targets for the treatment of human tauopathies.
Collapse
Affiliation(s)
- C F de Veij Mestdagh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands.
- Alzheimer Center Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
| | - M E Witte
- Department of Molecular Cell Biology and Immunology, MS Center, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - W Scheper
- Department of Human Genetics, Amsterdam UMC - location Vrije Universiteit, Amsterdam, The Netherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - A B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - R H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - R E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Ma B, Khan KS, Xu T, Xeque Amada J, Guo Z, Huang Y, Yan Y, Lam H, Cheng ASL, Ng BWL. Targeted Protein O-GlcNAcylation Using Bifunctional Small Molecules. J Am Chem Soc 2024; 146:9779-9789. [PMID: 38561350 PMCID: PMC11009946 DOI: 10.1021/jacs.3c14380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Protein O-linked β-N-acetylglucosamine modification (O-GlcNAcylation) plays a crucial role in regulating essential cellular processes. The disruption of the homeostasis of O-GlcNAcylation has been linked to various human diseases, including cancer, diabetes, and neurodegeneration. However, there are limited chemical tools for protein- and site-specific O-GlcNAc modification, rendering the precise study of the O-GlcNAcylation challenging. To address this, we have developed heterobifunctional small molecules, named O-GlcNAcylation TArgeting Chimeras (OGTACs), which enable protein-specific O-GlcNAcylation in living cells. OGTACs promote O-GlcNAcylation of proteins such as BRD4, CK2α, and EZH2 in cellulo by recruiting FKBP12F36V-fused O-GlcNAc transferase (OGT), with temporal, magnitude, and reversible control. Overall, the OGTACs represent a promising approach for inducing protein-specific O-GlcNAcylation, thus enabling functional dissection and offering new directions for O-GlcNAc-targeting therapeutic development.
Collapse
Affiliation(s)
- Bowen Ma
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
| | - Khadija Shahed Khan
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
- School
of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Tongyang Xu
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
| | - Josefina Xeque Amada
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
| | - Zhihao Guo
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
| | - Yunpeng Huang
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
| | - Yu Yan
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
| | - Henry Lam
- Department
of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Sai Kung, Hong Kong
| | - Alfred Sze-Lok Cheng
- School
of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Billy Wai-Lung Ng
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, Hong Kong
- Li Ka
Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong
Kong
| |
Collapse
|
8
|
Zhang JF, Fang ZT, Zhao JN, Liu GP, Shen X, Jiang GF, Liu Q. Acetylated tau exacerbates apoptosis by disturbing mitochondrial dynamics in HEK293 cells. J Neurochem 2024; 168:288-302. [PMID: 38275215 DOI: 10.1111/jnc.16053] [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: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
An increase in tau acetylation at K274 and K281 and abnormal mitochondrial dynamics have been observed in the brains of Alzheimer's disease (AD) patients. Here, we constructed three types of tau plasmids, TauKQ (acetylated tau mutant, by mutating its K274/K281 into glutamine to mimic disease-associated lysine acetylation), TauKR (non-acetylated tau mutant, by mutating its K274/K281 into arginine), and TauWT (wild-type human full-length tau). By transfecting these tau plasmids in HEK293 cells, we found that TauWT and TauKR induced mitochondrial fusion by increasing the level of mitochondrial fusion proteins. Conversely, TauKQ induced mitochondrial fission by reducing mitochondrial fusion proteins, exacerbating mitochondrial dysfunction and apoptosis. BGP-15 ameliorated TauKQ-induced mitochondrial dysfunction and apoptosis by improving mitochondrial dynamics. Our findings suggest that acetylation of K274/281 represents an important post-translational modification site regulating mitochondrial dynamics, and that BGP-15 holds potential as a therapeutic agent for mitochondria-associated diseases such as AD.
Collapse
Affiliation(s)
- Jun-Fei Zhang
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhi-Ting Fang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun-Ning Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Shen
- School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Gao-Feng Jiang
- Center for Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Qian Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
9
|
Du P, Zhang X, Lian X, Hölscher C, Xue G. O-GlcNAcylation and Its Roles in Neurodegenerative Diseases. J Alzheimers Dis 2024; 97:1051-1068. [PMID: 38250776 DOI: 10.3233/jad-230955] [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] [Indexed: 01/23/2024]
Abstract
As a non-classical post-translational modification, O-linked β-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation) is widely found in human organ systems, particularly in our brains, and is indispensable for healthy cell biology. With the increasing age of the global population, the incidence of neurodegenerative diseases is increasing, too. The common characteristic of these disorders is the aggregation of abnormal proteins in the brain. Current research has found that O-GlcNAcylation dysregulation is involved in misfolding or aggregation of these abnormal proteins to mediate disease progression, but the specific mechanism has not been defined. This paper reviews recent studies on O-GlcNAcylation's roles in several neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, Machado-Joseph's disease, and giant axonal neuropathy, and shows that O-GlcNAcylation, as glucose metabolism sensor, mediating synaptic function, participating in oxidative stress response and signaling pathway conduction, directly or indirectly regulates characteristic pathological protein toxicity and affects disease progression. The existing results suggest that targeting O-GlcNAcylation will provide new ideas for clinical diagnosis, prevention, and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Pengyang Du
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaomin Zhang
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xia Lian
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Christian Hölscher
- Academy of Chinese Medical Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Guofang Xue
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
10
|
Kalyaanamoorthy S, Opare SK, Xu X, Ganesan A, Rao PPN. Post-Translational Modifications in Tau and Their Roles in Alzheimer's Pathology. Curr Alzheimer Res 2024; 21:24-49. [PMID: 38623984 DOI: 10.2174/0115672050301407240408033046] [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: 01/01/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024]
Abstract
Microtubule-Associated Protein Tau (also known as tau) has been shown to accumulate into paired helical filaments and neurofibrillary tangles, which are known hallmarks of Alzheimer's disease (AD) pathology. Decades of research have shown that tau protein undergoes extensive post-translational modifications (PTMs), which can alter the protein's structure, function, and dynamics and impact the various properties such as solubility, aggregation, localization, and homeostasis. There is a vast amount of information describing the impact and role of different PTMs in AD pathology and neuroprotection. However, the complex interplay between these PTMs remains elusive. Therefore, in this review, we aim to comprehend the key post-translational modifications occurring in tau and summarize potential connections to clarify their impact on the physiology and pathophysiology of tau. Further, we describe how different computational modeling methods have helped in understanding the impact of PTMs on the structure and functions of the tau protein. Finally, we highlight the tau PTM-related therapeutics strategies that are explored for the development of AD therapy.
Collapse
Affiliation(s)
| | - Stanley Kojo Opare
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Xiaoxiao Xu
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Aravindhan Ganesan
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Praveen P N Rao
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
11
|
Jiang X, Yang Q. Recent advances in glycoside hydrolase family 20 and 84 inhibitors: Structures, inhibitory mechanisms and biological activities. Bioorg Chem 2023; 142:106870. [PMID: 39492366 DOI: 10.1016/j.bioorg.2023.106870] [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: 06/21/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 11/05/2024]
Abstract
Glycoside hydrolase family 20 (GH20) β-N-acetyl-d-hexosaminidase (Hex) catalyzes the cleavage of glycosidic linkages in glycans, glycolipids and glycoproteins, and is involved in glycoprotein modification, metabolism of glycoconjugate and the degradation of chitin in fungal cell walls and arthropod exoskeletons. GH84 O-β-N-acetyl-d-glucosaminidase (OGA), which is mechanistically similar related to GH20, participates in the O-GlcNAcylation modification, hydrolyzing the O-GlcNAc moiety from protein acceptors. Hex and OGA are of interest due to their potential for the treatment of disorder diseases and plant protection. Hex inhibitors act as molecular chaperones to treat lysosomal storage disease and as growth regulators to arrest insect molting. Inhibition of OGA is a promising therapeutic approach to treat tau pathology in neurodegenerative diseases such as Alzheimer's disease. However, since Hex and OGA exhibit similar active sites, there are challenges in designing highly selective inhibitors. The elucidation of the structural basis of the catalytic mechanism and substrate binding mode of Hex and OGA has provided core information for virtual screening and rational design of inhibitors. A large number of high-potency and selective inhibitors have been developed in the last five years. In this review, we focus on the recent advances in the structural modification, inhibitory activity, binding mechanisms and biological evaluation of Hex and OGA inhibitors, which will facilitate the development of new drugs and agrochemicals.
Collapse
Affiliation(s)
- Xi Jiang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Qing Yang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China; State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
12
|
Abstract
Deposits of the microtubule-associated protein Tau (MAPT) serve as a hallmark of neurodegenerative diseases known as tauopathies. Numerous studies have demonstrated that in diseases such as Alzheimer's disease (AD), Tau undergoes extensive remodeling. The attachment of post-translational modifications distributed throughout the entire sequence of the protein correlates with clinical presentation. A systematic examination of these protein alterations can shed light on their roles in both healthy and diseased states. However, the ability to access these modifications in the entire protein chain is limited as Tau can only be produced recombinantly or through semisynthesis. In this article, we describe the first chemical synthesis of the longest 2N4R isoform of Tau, consisting of 441 amino acids. The 2N4R Tau was divided into 3 major segments and a total of 11 fragments, all of which were prepared via solid-phase peptide synthesis. The successful chemical strategy has relied on the strategic use of two cysteine sites (C291 and C322) for the native chemical ligations (NCLs). This was combined with modern preparative protein chemistries, such as mercaptothreonine ligation (T205), diselenide-selenoester ligation (D358), and mutations of mercaptoamino acids into native residues via homogeneous radical desulfurization (A40, A77, A119, A157, A246, and A390). The successful completion of the synthesis has established a robust and scalable route to the native protein in multimilligram quantities and high purity. In broader terms, the presented strategy can be applied to the preparation of other shorter isoforms of Tau as well as to introduce all post-translational modifications that are characteristic of tauopathies such as AD.
Collapse
Affiliation(s)
- Wyatt C Powell
- Department of Chemistry, University of Colorado, Boulder, Colorado 80309, United States
| | - Ruiheng Jing
- Department of Chemistry, University of Colorado, Boulder, Colorado 80309, United States
| | - Maciej A Walczak
- Department of Chemistry, University of Colorado, Boulder, Colorado 80309, United States
| |
Collapse
|
13
|
Gupta S, Jinka SKA, Khanal S, Bhavnani N, Almashhori F, Lallo J, Mathias A, Al-Rhayyel Y, Herman D, Holden JG, Fleming SM, Raman P. Cognitive dysfunction and increased phosphorylated tau are associated with reduced O-GlcNAc signaling in an aging mouse model of metabolic syndrome. J Neurosci Res 2023; 101:1324-1344. [PMID: 37031439 DOI: 10.1002/jnr.25196] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 12/15/2022] [Accepted: 03/21/2023] [Indexed: 04/10/2023]
Abstract
Metabolic syndrome (MetS), characterized by hyperglycemia, obesity, and hyperlipidemia, can increase the risk of developing late-onset dementia. Recent studies in patients and mouse models suggest a putative link between hyperphosphorylated tau, a component of Alzheimer's disease-related dementia (ADRD) pathology, and cerebral glucose hypometabolism. Impaired glucose metabolism reduces glucose flux through the hexosamine metabolic pathway triggering attenuated O-linked N-acetylglucosamine (O-GlcNAc) protein modification. The goal of the current study was to investigate the link between cognitive function, tau pathology, and O-GlcNAc signaling in an aging mouse model of MetS, agouti KKAy+/- . Male and female C57BL/6, non-agouti KKAy-/- , and agouti KKAy+/- mice were aged 12-18 months on standard chow diet. Body weight, blood glucose, total cholesterol, and triglyceride were measured to confirm the MetS phenotype. Cognition, sensorimotor function, and emotional reactivity were assessed for each genotype followed by plasma and brain tissue collection for biochemical and molecular analyses. Body weight, blood glucose, total cholesterol, and triglyceride levels were significantly elevated in agouti KKAy+/- mice versus C57BL/6 controls and non-agouti KKAy-/- . Behaviorally, agouti KKAy+/- revealed impairments in sensorimotor and cognitive function versus age-matched C57BL/6 and non-agouti KKAy-/- mice. Immunoblotting demonstrated increased phosphorylated tau accompanied with reduced O-GlcNAc protein expression in hippocampal-associated dorsal midbrain of female agouti KKAy+/- versus C57BL/6 control mice. Together, these data demonstrate that impaired cognitive function and AD-related pathology are associated with reduced O-GlcNAc signaling in aging MetS KKAy+/- mice. Overall, our study suggests that interaction of tau pathology with O-GlcNAc signaling may contribute to MetS-induced cognitive dysfunction in aging.
Collapse
Affiliation(s)
- Shreya Gupta
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Sanjay K A Jinka
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Saugat Khanal
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Neha Bhavnani
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Fayez Almashhori
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Jason Lallo
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Amy Mathias
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Yasmine Al-Rhayyel
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Danielle Herman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - John G Holden
- Department of Psychology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sheila M Fleming
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Priya Raman
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| |
Collapse
|
14
|
Liu Q, Wang X, Hu Y, Zhao JN, Huang CH, Li T, Zhang BG, He Y, Wu YQ, Zhang ZJ, Wang GP, Liu GP. Acetylated tau exacerbates learning and memory impairment by disturbing with mitochondrial homeostasis. Redox Biol 2023; 62:102697. [PMID: 37037158 PMCID: PMC10114242 DOI: 10.1016/j.redox.2023.102697] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/22/2023] [Accepted: 04/05/2023] [Indexed: 04/09/2023] Open
Abstract
Increased tau acetylation at K274 and K281 has been observed in the brains of Alzheimer's disease (AD) patients and animal models, and mitochondrial dysfunction are noticeable and early features of AD. However, the effect of acetylated tau on mitochondria has been unclear until now. Here, we constructed three type of tau forms, acetylated tau mutant by mutating its K274/K281 into Glutamine (TauKQ) to mimic disease-associated lysine acetylation, the non-acetylation tau mutant by mutating its K274/K281 into Arginine (TauKR) and the wild-type human full-length tau (TauWT). By overexpression of these tau forms in vivo and in vitro, we found that, TauKQ induced more severe cognitive deficits with neuronal loss, dendritic plasticity damage and mitochondrial dysfunctions than TauWT. Unlike TauWT induced mitochondria fusion, TauKQ not only induced mitochondria fission by decreasing mitofusion proteins, but also inhibited mitochondrial biogenesis via reduction of PGC-1a/Nrf1/Tfam levels. TauKR had no significant difference in the cognitive and mitochondrial abnormalities compared with TauWT. Treatment with BGP-15 rescued impaired learning and memory by attenuation of mitochondrial dysfunction, neuronal loss and dendritic complexity damage, which caused by TauKQ. Our data suggested that, acetylation at K274/281 was an important post translational modification site for tau neurotoxicity, and BGP-15 is a potential therapeutic drug for AD.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Yu Hu
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun-Ning Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chun-Hui Huang
- Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research, Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Ting Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bing-Ge Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan-Qing Wu
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Zai-Jun Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research, Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Guo-Ping Wang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
15
|
Hart G, Huang CW, Rust N, Wu HF. Altered O-GlcNAcylation and mitochondrial dysfunction, a molecular link between brain glucose dysregulation and sporadic Alzheimer’s disease. Neural Regen Res 2023; 18:779-783. [DOI: 10.4103/1673-5374.354515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
16
|
Effect of cx-DHED on Abnormal Glucose Transporter Expression Induced by AD Pathologies in the 5xFAD Mouse Model. Int J Mol Sci 2022; 23:ijms231810602. [PMID: 36142509 PMCID: PMC9505457 DOI: 10.3390/ijms231810602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a form of dementia associated with abnormal glucose metabolism resulting from amyloid-beta (Aβ) plaques and intracellular neurofibrillary tau protein tangles. In a previous study, we confirmed that carboxy-dehydroevodiamine∙HCl (cx-DHED), a derivative of DHED, was effective at improving cognitive impairment and reducing phosphorylated tau levels and synaptic loss in an AD mouse model. However, the specific mechanism of action of cx-DHED is unclear. In this study, we investigated how the cx-DHED attenuates AD pathologies in the 5xFAD mouse model, focusing particularly on abnormal glucose metabolism. We analyzed behavioral changes and AD pathologies in mice after intraperitoneal injection of cx-DHED for 2 months. As expected, cx-DHED reversed memory impairment and reduced Aβ plaques and astrocyte overexpression in the brains of 5xFAD mice. Interestingly, cx-DHED reversed the abnormal expression of glucose transporters in the brains of 5xFAD mice. In addition, otherwise low O-GlcNac levels increased, and the overactivity of phosphorylated GSK-3β decreased in the brains of cx-DHED-treated 5xFAD mice. Finally, the reduction in synaptic proteins was found to also improve by treatment with cx-DHED. Therefore, we specifically demonstrated the protective effects of cx-DHED against AD pathologies and suggest that cx-DHED may be a potential therapeutic drug for AD.
Collapse
|
17
|
Xiong J, Deng I, Kelliny S, Lin L, Bobrovskaya L, Zhou XF. Long term high fat diet induces metabolic disorders and aggravates behavioral disorders and cognitive deficits in MAPT P301L transgenic mice. Metab Brain Dis 2022; 37:1941-1957. [PMID: 35704147 DOI: 10.1007/s11011-022-01029-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/07/2022] [Indexed: 01/18/2023]
Abstract
Most Alzheimer disease (AD) patients present as sporadic late onset AD, with metabolic factors playing an important role in the occurrence and development of AD. Given the link between peripheral insulin resistance and tau pathology in streptozotocin-injected and db/db mouse models of diabetes, we fed high fat diet (HFD) to pR5 mice expressing P301L mutant human tau, with the aim of developing a new model with characteristics of obesity, T2DM and AD to mimic AD patients exacerbated by obesity and T2DM, an increasing trend in modern society. In our study, pR5 and C57BL/6 (WT) mice were randomly allocated to a standard diet (STD) or HFD for 30 weeks starting at 8 weeks of age. Food intake was measured weekly, body weight and fasting glucose levels were measured fortnightly, and a comprehensive behavioral test battery was performed to assess anxiety, depression and cognitive dysfunction. Glucose and insulin tolerance tests were performed after 30 weeks of HFD. We also investigated the effect of long term HFD on tau pathology in the brains of WT and P301L mice by performing western blotting of whole brain homogenates for total tau, phosphorylated tau at Ser396 and Thr231. Our results show that pR5 mice fed with HFD are more vulnerable to diet induced obesity compared to WT, especially with increasing age. In addition, pR5 mice on HFD developed glucose intolerance and insulin resistance. It was identified that long term HFD significantly aggravates depression like behavior and impairs cognitive function in pR5 mice, and also induces anxiety like behavior in both pR5 and WT mice. Long term HFD was also shown to aggravate tau hyperphosphorylation in pR5 transgenic mice, and increase total and hyperphosphorylated tau in WT mice. These results indicate that diet induced obesity of pR5 transgenic mice expressing P301L mutant human tau generates T2DM, and aggravates tau phosphorylation, and is therefore a model useful for investigations that seek to understand the relationships between AD, T2DM and obesity, and the underlying biochemical changes and mechanisms associated with metabolic disorders and AD tauopathy.
Collapse
Affiliation(s)
- Jing Xiong
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia.
- Department of Neurology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan Province, China.
| | - Isaac Deng
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sally Kelliny
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
- Faculty of Phamacy, Assiut University, Assiut, Egypt
| | - Liying Lin
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia.
| |
Collapse
|
18
|
Velueta-Viveros M, Martínez-Bailén M, Puerta A, Romero-Hernández LL, Křen V, Merino-Montiel P, Montiel-Smith S, Fernandes MX, Moreno-Vargas AJ, Padrón JM, López Ó, Fernández-Bolaños JG. Carbohydrate-derived bicyclic selenazolines as new dual inhibitors (cholinesterases/OGA) against Alzheimer’s disease. Bioorg Chem 2022; 127:105983. [DOI: 10.1016/j.bioorg.2022.105983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022]
|
19
|
Ye H, Han Y, Li P, Su Z, Huang Y. The Role of Post-Translational Modifications on the Structure and Function of Tau Protein. J Mol Neurosci 2022; 72:1557-1571. [PMID: 35325356 DOI: 10.1007/s12031-022-02002-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/14/2022] [Indexed: 12/14/2022]
Abstract
Involving addition of chemical groups or protein units to specific residues of the target protein, post-translational modifications (PTMs) alter the charge, hydrophobicity, and conformation of a protein, which in tune influences protein function, protein - protein interaction, and protein aggregation. While the occurrence of PTMs is dynamic and subject to regulations, conformational disorder of the target protein facilitates PTMs. The microtubule-associated protein tau is a typical intrinsically disordered protein that undergoes a variety of PTMs including phosphorylation, acetylation, ubiquitination, methylation, and oxidation. Accumulated evidence shows that these PTMs play a critical role in regulating tau-microtubule interaction, tau localization, tau degradation and aggregation, and reinforces the correlation between tau PTMs and pathogenesis of neurodegenerative disease. Here, we review tau PTMs with an emphasis on their influence on tau structure. With available biophysical characterization results, we describe how PTMs induce conformational changes in tau monomer and regulate tau aggregation. Compared to functional analysis of tau PTMs, biophysical characterization of tau PTMs is lagging. While it is challenging, characterizing the specific effects of PTMs on tau conformation and interaction is indispensable to unravel the tau PTM code.
Collapse
Affiliation(s)
- Haiqiong Ye
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430068, China.,Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, China
| | - Yue Han
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430068, China.,Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, China
| | - Ping Li
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430068, China.,Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, China
| | - Zhengding Su
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430068, China.,Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, China
| | - Yongqi Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China. .,Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430068, China. .,Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, China.
| |
Collapse
|
20
|
Abstract
Post-translational modification with O-linked β-N-acetylglucosamine (O-GlcNAc), a process referred to as O-GlcNAcylation, occurs on a vast variety of proteins. Mounting evidence in the past several decades has clearly demonstrated that O-GlcNAcylation is a unique and ubiquitous modification. Reminiscent of a code, protein O-GlcNAcylation functions as a crucial regulator of nearly all cellular processes studied. The primary aim of this review is to summarize the developments in our understanding of myriad protein substrates modified by O-GlcNAcylation from a systems perspective. Specifically, we provide a comprehensive survey of O-GlcNAcylation in multiple species studied, including eukaryotes (e.g., protists, fungi, plants, Caenorhabditis elegans, Drosophila melanogaster, murine, and human), prokaryotes, and some viruses. We evaluate features (e.g., structural properties and sequence motifs) of O-GlcNAc modification on proteins across species. Given that O-GlcNAcylation functions in a species-, tissue-/cell-, protein-, and site-specific manner, we discuss the functional roles of O-GlcNAcylation on human proteins. We focus particularly on several classes of relatively well-characterized human proteins (including transcription factors, protein kinases, protein phosphatases, and E3 ubiquitin-ligases), with representative O-GlcNAc site-specific functions presented. We hope the systems view of the great endeavor in the past 35 years will help demystify the O-GlcNAc code and lead to more fascinating studies in the years to come.
Collapse
Affiliation(s)
- Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, United States
| | - Chunyan Hou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, United States
| | - Ci Wu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, United States
| |
Collapse
|
21
|
Gong CX, Dai CL, Liu F, Iqbal K. Multi-Targets: An Unconventional Drug Development Strategy for Alzheimer's Disease. Front Aging Neurosci 2022; 14:837649. [PMID: 35222001 PMCID: PMC8864545 DOI: 10.3389/fnagi.2022.837649] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/20/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that eventually leads to dementia and death of the patient. Despite the enormous amounts of resources and efforts for AD drug development during the last three decades, no effective treatments have been developed that can slow or halt the progression of the disease. Currently available drugs for treating AD can only improve clinical symptoms temporarily with moderate efficacies. In recent years, the scientific community has realized these challenges and reconsidered the future directions of AD drug development. The most significant recent changes in AD drug development strategy include shifting from amyloid-based targets to other targets, such as tau, and efforts toward better designs for clinical trials. However, most AD drug development is still focused on a single mechanism or target, which is the conventional strategy for drug development. Although multifactorial mechanisms and, on this basis, multi-target strategies have been proposed in recent years, this approach has not been widely recognized and accepted by the mainstream of AD drug development. Here, we emphasize the multifactorial mechanisms of AD and discuss the urgent need for a paradigm shift in AD drug development from a single target to multiple targets, either with the multi-target-directed ligands approach or the combination therapy approach. We hope this article will increase the recognition of the multifactorial nature of AD and promote this paradigm shift. We believe that such a shift will facilitate successful development of effective AD therapies.
Collapse
Affiliation(s)
- Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, New York, NY, United States
| | | | | | | |
Collapse
|
22
|
Nanomolar inhibition of human OGA by 2-acetamido-2-deoxy-d-glucono-1,5-lactone semicarbazone derivatives. Eur J Med Chem 2021; 223:113649. [PMID: 34186233 DOI: 10.1016/j.ejmech.2021.113649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 12/25/2022]
Abstract
O-GlcNAcylation is a dynamic post-translational modification mediated by O-linked β-N-acetylglucosamine transferase (OGT) and O-GlcNAc hydrolase (OGA), that adds or removes a single β-N-acetylglucosamine (GlcNAc) moiety to or from serine/threonine residues of nucleocytosolic and mitochondrial proteins, respectively. The perturbed homeostasis of O-GlcNAc cycling results in several pathological conditions. Human OGA is a promising therapeutic target in diseases where aberrantly low levels of O-GlcNAc are experienced, such as tauopathy in Alzheimer's disease. A new class of potent OGA inhibitors, 2-acetamido-2-deoxy-d-glucono-1,5-lactone (thio)semicarbazones, have been identified. Eight inhibitors were designed and synthesized in five steps starting from d-glucosamine and with 15-55% overall yields. A heterologous OGA expression protocol with strain selection and isolation has been optimized that resulted in stable, active and full length human OGA (hOGA) isomorph. Thermal denaturation kinetics of hOGA revealed environmental factors affecting hOGA stability. From kinetics experiments, the synthesized compounds proved to be efficient competitive inhibitors of hOGA with Ki-s in the range of ∼30-250 nM and moderate selectivity with respect to lysosomal β-hexosaminidases. In silico studies consisting of Prime protein-ligand refinements, QM/MM optimizations and QM/MM-PBSA binding free energy calculations revealed the factors governing the observed potencies, and led to design of the most potent analogue 2-acetamido-2-deoxy-d-glucono-1,5-lactone 4-(2-naphthyl)-semicarbazone 6g (Ki = 36 nM). The protocol employed has applications in future structure based inhibitor design targeting OGA.
Collapse
|
23
|
Chamberlain S, Gabriel H, Strittmatter W, Didsbury J. An Exploratory Phase IIa Study of the PPAR delta/gamma Agonist T3D-959 Assessing Metabolic and Cognitive Function in Subjects with Mild to Moderate Alzheimer's Disease. J Alzheimers Dis 2021; 73:1085-1103. [PMID: 31884472 PMCID: PMC7081093 DOI: 10.3233/jad-190864] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background: T3D-959 is a chemically unique, brain penetrant, dual PPAR delta/gamma agonist with 15-fold higher PPAR delta selectivity. Ubiquitous brain expression of PPAR delta, its critical role in regulating glucose and lipid metabolism, and the Alzheimer’s disease (AD)-like phenotype of PPAR delta null mice motivated this study. Objective: To determine safety and tolerability of multiple doses of T3D-959 in subjects with mild to moderate AD, examine systemic and central drug pharmacology and in an exploratory manner, perform cognitive assessments. Methods: Thirty-four subjects with mild-to-moderate AD were orally administered 3, 10, 30, or 90 mg of T3D-959 daily for 14 days. There was no inclusion of a placebo arm. Safety and tolerability were monitored. Systemic drug pharmacology was examined via plasma metabolomics LC-MS-MS analysis, cerebral drug pharmacology via FDG-PET measures of changes in Relative CMRgl (R CMRgl, AD-effected regions relative to brain reference regions), and cognitive function assessed before and after drug treatment and again one week after completion of drug treatment, by ADAS-cog11 and the Digit Symbol Substitution Test (DSST). Results: T3D-959 was in general safe and well tolerated. Single point pharmacokinetics at the Tmax showed dose dependent exposure. Plasma metabolome profile changes showed dose-dependent systemic effects on lipid metabolism and metabolism related to insulin sensitization. Relative FDG-PET imaging demonstrated dose-dependent, regional, effects of T3D-959 on R CMRgl based on the use of multiple reference regions. ADAS-cog11 and DSST cognitive assessments showed improvements with possible ApoE genotype association and pharmacodynamics related to the mechanism of drug action. Conclusions: Exploratory data from this Phase IIa clinical trial supports further clinical investigation of T3D-959 in a larger placebo-controlled clinical study.
Collapse
Affiliation(s)
| | - Hoda Gabriel
- T3D Therapeutics, Inc., Research Triangle Park, NC, USA
| | | | - John Didsbury
- T3D Therapeutics, Inc., Research Triangle Park, NC, USA
| |
Collapse
|
24
|
Akan I, Halim A, Vakhrushev SY, Clausen H, Hanover JA. Drosophila O-GlcNAcase Mutants Reveal an Expanded Glycoproteome and Novel Growth and Longevity Phenotypes. Cells 2021; 10:cells10051026. [PMID: 33925313 PMCID: PMC8145559 DOI: 10.3390/cells10051026] [Citation(s) in RCA: 3] [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: 01/31/2021] [Revised: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 12/17/2022] Open
Abstract
The reversible posttranslational O-GlcNAc modification of serine or threonine residues of intracellular proteins is involved in many cellular events from signaling cascades to epigenetic and transcriptional regulation. O-GlcNAcylation is a conserved nutrient-dependent process involving two enzymes, with O-GlcNAc transferase (OGT) adding O-GlcNAc and with O-GlcNAcase (OGA) removing it in a manner that’s protein- and context-dependent. O-GlcNAcylation is essential for epigenetic regulation of gene expression through its action on Polycomb and Trithorax and COMPASS complexes. However, the important role of O-GlcNAc in adult life and health span has been largely unexplored, mainly due the lack of available model systems. Cataloging the O-GlcNAc proteome has proven useful in understanding the biology of this modification in vivo. In this study, we leveraged a recently developed oga knockout fly mutant to identify the O-GlcNAcylated proteins in adult Drosophilamelanogaster. The adult O-GlcNAc proteome revealed many proteins related to cell and organismal growth, development, differentiation, and epigenetics. We identified many O-GlcNAcylated proteins that play a role in increased growth and decreased longevity, including HCF, SIN3A, LOLA, KISMET, ATX2, SHOT, and FOXO. Interestingly, oga mutant flies are larger and have a shorter life span compared to wild type flies, suggesting increased O-GlcNAc results in increased growth. Our results suggest that O-GlcNAc alters the function of many proteins related to transcription, epigenetic modification and signaling pathways that regulate growth rate and longevity. Therefore, our findings highlight the importance of O-GlcNAc in growth and life span in adult Drosophila.
Collapse
Affiliation(s)
- Ilhan Akan
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Adnan Halim
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; (A.H.); (S.Y.V.); (H.C.)
| | - Sergey Y. Vakhrushev
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; (A.H.); (S.Y.V.); (H.C.)
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark; (A.H.); (S.Y.V.); (H.C.)
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
- Correspondence:
| |
Collapse
|
25
|
Ruegenberg S, Mayr FAMC, Atanassov I, Baumann U, Denzel MS. Protein kinase A controls the hexosamine pathway by tuning the feedback inhibition of GFAT-1. Nat Commun 2021; 12:2176. [PMID: 33846315 PMCID: PMC8041777 DOI: 10.1038/s41467-021-22320-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 03/10/2021] [Indexed: 02/01/2023] Open
Abstract
The hexosamine pathway (HP) is a key anabolic pathway whose product uridine 5'-diphospho-N-acetyl-D-glucosamine (UDP-GlcNAc) is an essential precursor for glycosylation processes in mammals. It modulates the ER stress response and HP activation extends lifespan in Caenorhabditis elegans. The highly conserved glutamine fructose-6-phosphate amidotransferase 1 (GFAT-1) is the rate-limiting HP enzyme. GFAT-1 activity is modulated by UDP-GlcNAc feedback inhibition and via phosphorylation by protein kinase A (PKA). Molecular consequences of GFAT-1 phosphorylation, however, remain poorly understood. Here, we identify the GFAT-1 R203H substitution that elevates UDP-GlcNAc levels in C. elegans. In human GFAT-1, the R203H substitution interferes with UDP-GlcNAc inhibition and with PKA-mediated Ser205 phosphorylation. Our data indicate that phosphorylation affects the interactions of the two GFAT-1 domains to control catalytic activity. Notably, Ser205 phosphorylation has two discernible effects: it lowers baseline GFAT-1 activity and abolishes UDP-GlcNAc feedback inhibition. PKA controls the HP by uncoupling the metabolic feedback loop of GFAT-1.
Collapse
Affiliation(s)
- Sabine Ruegenberg
- grid.419502.b0000 0004 0373 6590Max Planck Institute for Biology of Ageing, Cologne, Germany ,grid.6190.e0000 0000 8580 3777Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Felix A. M. C. Mayr
- grid.419502.b0000 0004 0373 6590Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Ilian Atanassov
- grid.419502.b0000 0004 0373 6590Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Ulrich Baumann
- grid.6190.e0000 0000 8580 3777Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Martin S. Denzel
- grid.419502.b0000 0004 0373 6590Max Planck Institute for Biology of Ageing, Cologne, Germany ,grid.6190.e0000 0000 8580 3777CECAD - Cluster of Excellence, University of Cologne, Cologne, Germany ,grid.6190.e0000 0000 8580 3777Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
26
|
Pan D, Gu JH, Zhang J, Hu Y, Liu F, Iqbal K, Cekic N, Vocadlo DJ, Dai CL, Gong CX. Thiamme2-G, a Novel O-GlcNAcase Inhibitor, Reduces Tau Hyperphosphorylation and Rescues Cognitive Impairment in Mice. J Alzheimers Dis 2021; 81:273-286. [PMID: 33814439 DOI: 10.3233/jad-201450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Abnormal hyperphosphorylation of microtubule-associated protein tau plays a pivotal role in Alzheimer's disease (AD). We previously found that O-GlcNAcylation inversely correlates to hyperphosphorylation of tau in AD brain, and downregulation of brain O-GlcNAcylation promotes tau hyperphosphorylation and AD-like neurodegeneration in mice. OBJECTIVE Herein we investigated the effect of increasing O-GlcNAcylation by using intermittent dosing with low doses of a potent novel O-GlcNAcase (OGA) inhibitor on AD-like brain changes and cognitive function in a mouse model of sporadic AD (sAD) induced by intracerebroventricular (ICV) injection of streptozotocin (STZ). METHODS STZ was injected into the lateral ventricle of C57BL/6J mice. From the second day, Thiamme2-G (TM2G) or saline, as a vehicle control, was orally administered to the ICV-STZ mice three times per week for five weeks. A separate group of ICV-saline mice treated with saline was used as a baseline control. Behavioral tests, including open field and novel object recognition, were conducted three weeks after the first dose of the TM2G or saline. Protein O-GlcNAcylation, tau hyperphosphorylation, synaptic proteins, and neuroinflammation in the mouse brain were assessed by western blotting. RESULTS ICV-STZ caused decreased protein O-GlcNAcylation. Enhancement of O-GlcNAcylation to moderate levels by using low-dose OGA inhibitor in ICV-STZ mice prevented STZ-induced body weight loss, rescued cognitive impairments, and restored AD-like pathologies, including hyperphosphorylation of tau and abnormalities in synaptic proteins and neuroinflammation. CONCLUSION These findings suggest that moderately increasing protein O-GlcNAcylation by using low doses of OGA inhibitor may be a suitable therapeutic strategy for sAD.
Collapse
Affiliation(s)
- Danmin Pan
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Jin-Hua Gu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Jin Zhang
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Yae Hu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Nevena Cekic
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - David J Vocadlo
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Chun-Ling Dai
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
27
|
McCarty MF, Lerner A. The second phase of brain trauma can be controlled by nutraceuticals that suppress DAMP-mediated microglial activation. Expert Rev Neurother 2021; 21:559-570. [PMID: 33749495 DOI: 10.1080/14737175.2021.1907182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION A delayed second wave of brain trauma is mediated in large part by microglia that are activated to a pro-inflammatory M1 phenotype by DAMP proteins released by dying neurons. These microglia can promote apoptosis or necrosis in neighboring neurons by producing a range of pro-inflammatory cytokines and the deadly oxidant peroxynitrite. This second wave could therefore be mitigated with agents that blunt the post-traumatic M1 activation of microglia and that preferentially promote a pro-healing M2 phenotype. AREAS COVERED The literature on nutraceuticals that might have clinical potential in this regard. EXPERT OPINION The chief signaling pathway whereby DAMPs promote M1 microglial activation involves activation of toll-like receptor 4 (TLR4), NADPH oxidase, NF-kappaB, and the stress activated kinases JNK and p38. The green tea catechin EGCG can suppress TLR4 expression. Phycocyanobilin can inhibit NOX2-dependent NADPH oxidase, ferulate and melatonin can oppose pro-inflammatory signal modulation by NADPH oxidase-derived oxidants. Long-chain omega-3 fatty acids, the soy isoflavone genistein, the AMPK activator berberine, glucosamine, and ketone bodies can down-regulate NF-kappaB activation. Vitamin D activity can oppose JNK/p38 activation. A sophisticated program of nutraceutical supplementation may have important potential for mitigating the second phase of neuronal death and aiding subsequent healing.
Collapse
Affiliation(s)
- Mark F McCarty
- Department of research, Catalytic Longevity Foundation, San Diego, California, USA
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| |
Collapse
|
28
|
Zuliani I, Lanzillotta C, Tramutola A, Barone E, Perluigi M, Rinaldo S, Paone A, Cutruzzolà F, Bellanti F, Spinelli M, Natale F, Fusco S, Grassi C, Di Domenico F. High-Fat Diet Leads to Reduced Protein O-GlcNAcylation and Mitochondrial Defects Promoting the Development of Alzheimer's Disease Signatures. Int J Mol Sci 2021; 22:ijms22073746. [PMID: 33916835 PMCID: PMC8038495 DOI: 10.3390/ijms22073746] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/20/2021] [Accepted: 04/01/2021] [Indexed: 02/05/2023] Open
Abstract
The disturbance of protein O-GlcNAcylation is emerging as a possible link between altered brain metabolism and the progression of neurodegeneration. As observed in brains with Alzheimer's disease (AD), flaws of the cerebral glucose uptake translate into reduced protein O-GlcNAcylation, which promote the formation of pathological hallmarks. A high-fat diet (HFD) is known to foster metabolic dysregulation and insulin resistance in the brain and such effects have been associated with the reduction of cognitive performances. Remarkably, a significant role in HFD-related cognitive decline might be played by aberrant protein O-GlcNAcylation by triggering the development of AD signature and mitochondrial impairment. Our data support the impairment of total protein O-GlcNAcylation profile both in the brain of mice subjected to a 6-week high-fat-diet (HFD) and in our in vitro transposition on SH-SY5Y cells. The reduction of protein O-GlcNAcylation was associated with the development of insulin resistance, induced by overfeeding (i.e., defective insulin signaling and reduced mitochondrial activity), which promoted the dysregulation of the hexosamine biosynthetic pathway (HBP) flux, through the AMPK-driven reduction of GFAT1 activation. Further, we observed that a HFD induced the selective impairment of O-GlcNAcylated-tau and of O-GlcNAcylated-Complex I subunit NDUFB8, thus resulting in tau toxicity and reduced respiratory chain functionality respectively, highlighting the involvement of this posttranslational modification in the neurodegenerative process.
Collapse
Affiliation(s)
- Ilaria Zuliani
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Chiara Lanzillotta
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Antonella Tramutola
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Eugenio Barone
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Marzia Perluigi
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Serena Rinaldo
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Alessio Paone
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Francesca Cutruzzolà
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
| | - Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.S.); (F.N.); (S.F.); (C.G.)
| | - Francesca Natale
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.S.); (F.N.); (S.F.); (C.G.)
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.S.); (F.N.); (S.F.); (C.G.)
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.S.); (F.N.); (S.F.); (C.G.)
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Fabio Di Domenico
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (C.L.); (A.T.); (E.B.); (M.P.); (S.R.); (A.P.); (F.C.)
- Correspondence:
| |
Collapse
|
29
|
Lee JH, Veronese M, Liow JS, Morse CL, Montero Santamaria JA, Haskali MB, Zoghbi SS, Pike VW, Innis RB, Zanotti-Fregonara P. Region- and voxel-based quantification in human brain of [ 18F]LSN3316612, a radioligand for O-GlcNAcase. EJNMMI Res 2021; 11:35. [PMID: 33796956 PMCID: PMC8017047 DOI: 10.1186/s13550-021-00780-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/23/2021] [Indexed: 11/25/2022] Open
Abstract
Background Previous studies found that the positron emission tomography (PET) radioligand [18F]LSN3316612 accurately quantified O-GlcNAcase in human brain using a two-tissue compartment model (2TCM). This study sought to assess kinetic model(s) as an alternative to 2TCM for quantifying [18F]LSN3316612 binding, particularly in order to generate good-quality parametric images. Methods The current study reanalyzed data from a previous study of 10 healthy volunteers who underwent both test and retest PET scans with [18F]LSN3316612. Kinetic analysis was performed at the region level with 2TCM using 120-min PET data and arterial input function, which was considered as the gold standard. Quantification was then obtained at both the region and voxel levels using Logan plot, Ichise's multilinear analysis-1 (MA1), standard spectral analysis (SA), and impulse response function at 120 min (IRF120). To avoid arterial sampling, a noninvasive relative quantification (standardized uptake value ratio (SUVR)) was also tested using the corpus callosum as a pseudo-reference region. Venous samples were also assessed to see whether they could substitute for arterial ones. Results Logan and MA1 generated parametric images of good visual quality and their total distribution volume (VT) values at both the region and voxel levels were strongly correlated with 2TCM-derived VT (r = 0.96–0.99) and showed little bias (up to − 8%). SA was more weakly correlated to 2TCM-derived VT (r = 0.93–0.98) and was more biased (~ 16%). IRF120 showed a strong correlation with 2TCM-derived VT (r = 0.96) but generated noisier parametric images. All techniques were comparable to 2TCM in terms of test–retest variability and reliability except IRF120, which gave significantly worse results. Noninvasive SUVR values were not correlated with 2TCM-derived VT, and arteriovenous equilibrium was never reached. Conclusions Compared to SA and IRF, Logan and MA1 are more suitable alternatives to 2TCM for quantifying [18F]LSN3316612 and generating good-quality parametric images.
Collapse
Affiliation(s)
- Jae-Hoon Lee
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA. .,Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Jose A Montero Santamaria
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Mohammad B Haskali
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
30
|
Ma J, Wu C, Hart GW. Analytical and Biochemical Perspectives of Protein O-GlcNAcylation. Chem Rev 2021; 121:1513-1581. [DOI: 10.1021/acs.chemrev.0c00884] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington D.C. 20057, United States
| | - Ci Wu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington D.C. 20057, United States
| | - Gerald W. Hart
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
31
|
Alquezar C, Arya S, Kao AW. Tau Post-translational Modifications: Dynamic Transformers of Tau Function, Degradation, and Aggregation. Front Neurol 2021; 11:595532. [PMID: 33488497 PMCID: PMC7817643 DOI: 10.3389/fneur.2020.595532] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Post-translational modifications (PTMs) on tau have long been recognized as affecting protein function and contributing to neurodegeneration. The explosion of information on potential and observed PTMs on tau provides an opportunity to better understand these modifications in the context of tau homeostasis, which becomes perturbed with aging and disease. Prevailing views regard tau as a protein that undergoes abnormal phosphorylation prior to its accumulation into the toxic aggregates implicated in Alzheimer's disease (AD) and other tauopathies. However, the phosphorylation of tau may, in fact, represent part of the normal but interrupted function and catabolism of the protein. In addition to phosphorylation, tau undergoes another forms of post-translational modification including (but not limited to), acetylation, ubiquitination, glycation, glycosylation, SUMOylation, methylation, oxidation, and nitration. A holistic appreciation of how these PTMs regulate tau during health and are potentially hijacked in disease remains elusive. Recent studies have reinforced the idea that PTMs play a critical role in tau localization, protein-protein interactions, maintenance of levels, and modifying aggregate structure. These studies also provide tantalizing clues into the possibility that neurons actively choose how tau is post-translationally modified, in potentially competitive and combinatorial ways, to achieve broad, cellular programs commensurate with the distinctive environmental conditions found during development, aging, stress, and disease. Here, we review tau PTMs and describe what is currently known about their functional impacts. In addition, we classify these PTMs from the perspectives of protein localization, electrostatics, and stability, which all contribute to normal tau function and homeostasis. Finally, we assess the potential impact of tau PTMs on tau solubility and aggregation. Tau occupies an undoubtedly important position in the biology of neurodegenerative diseases. This review aims to provide an integrated perspective of how post-translational modifications actively, purposefully, and dynamically remodel tau function, clearance, and aggregation. In doing so, we hope to enable a more comprehensive understanding of tau PTMs that will positively impact future studies.
Collapse
Affiliation(s)
| | | | - Aimee W. Kao
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Zuliani I, Lanzillotta C, Tramutola A, Francioso A, Pagnotta S, Barone E, Perluigi M, Di Domenico F. The Dysregulation of OGT/OGA Cycle Mediates Tau and APP Neuropathology in Down Syndrome. Neurotherapeutics 2021; 18:340-363. [PMID: 33258073 PMCID: PMC8116370 DOI: 10.1007/s13311-020-00978-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
Protein O-GlcNAcylation is a nutrient-related post-translational modification that, since its discovery some 30 years ago, has been associated with the development of neurodegenerative diseases. As reported in Alzheimer's disease (AD), flaws in the cerebral glucose uptake translate into reduced hexosamine biosynthetic pathway flux and subsequently lead to aberrant protein O-GlcNAcylation. Notably, the reduction of O-GlcNAcylated proteins involves also tau and APP, thus promoting their aberrant phosphorylation in AD brain and the onset of AD pathological markers. Down syndrome (DS) individuals are characterized by the early development of AD by the age of 60 and, although the two conditions present the same pathological hallmarks and share the alteration of many molecular mechanisms driving brain degeneration, no evidence has been sought on the implication of O-GlcNAcylation in DS pathology. Our study aimed to unravel for the first time the role of protein O-GlcNacylation in DS brain alterations positing the attention of potential trisomy-related mechanisms triggering the aberrant regulation of OGT/OGA cycle. We demonstrate the disruption of O-GlcNAcylation homeostasis, as an effect of altered OGT and OGA regulatory mechanism, and confirm the relevance of O-GlcNAcylation in the appearance of AD hallmarks in the brain of a murine model of DS. Furthermore, we provide evidence for the neuroprotective effects of brain-targeted OGA inhibition. Indeed, the rescue of OGA activity was able to restore protein O-GlcNAcylation, and reduce AD-related hallmarks and decreased protein nitration, possibly as effect of induced autophagy.
Collapse
Affiliation(s)
- Ilaria Zuliani
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Antonio Francioso
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Sara Pagnotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
33
|
Flax J, Wilkins HM, Miller R, Griffith S, Cork GK, Qiang A, Thompson J, Swerdlow RH, Slawson C. OGA Inhibition Alters Energetics and Nutrient Sensing in Alzheimer's Disease Cytoplasmic Hybrids. J Alzheimers Dis 2020; 78:1743-1753. [PMID: 33285636 DOI: 10.3233/jad-200996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) features reductions in key bioenergetic fluxes and perturbed mitochondrial function. Cytoplasmic hybrids (cybrids) generated through the transfer of AD subject mitochondria to mtDNA-depleted SH-SY5Y neuroblastoma cells recapitulate some of these features in an in vitro setting. OBJECTIVE For this study, we used the AD cybrid model to assess the impact of a nutrient-excess like-state via increasing O-GlcNAcylation on whole cell and mitochondrial homeostasis. METHODS We induced increased O-GlcNAc by treating AD and control cybrid cell lines with Thiamet G (TMG), an inhibitor of the O-GlcNAcase enzyme that mediates removal of the nutrient-dependent O-GlcNAc modification. RESULTS Relative to control cybrid cell lines, AD cybrid lines showed a blunted response to TMG-induced O-GlcNAcylation. At baseline, AD cybrid cell line mitochondria showed partial activation of several proteins that help maintain bioenergetic homeostasis such as AMP-Regulated Kinase suggesting that AD mitochondria initiate a state of nutrient stress promoting energetic compensation; however, this compensation reduces the capacity of cells to respond to additional nutrient-related stresses such as TMG treatment. Also, TMG caused disruptions in acetylation and Sirtuin 3 expression, while lowing total energetic output of the cell. CONCLUSION Together, these findings suggest that modulation of O-GlcNAc is essential for proper energetic function of the mitochondria, and AD mitochondrial capacity to handle nutrient-excess is limited.
Collapse
Affiliation(s)
- Jarrod Flax
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather M Wilkins
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Reegan Miller
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sarah Griffith
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Gentry K Cork
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Amy Qiang
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffrey Thompson
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Chad Slawson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
34
|
Hart GW, Huang CW. Increased O-GlcNAcylation prevents degeneration of dopamine neurons. Brain 2020; 143:3515-3518. [PMID: 33439984 DOI: 10.1093/brain/awaa398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 11/13/2022] Open
Abstract
This scientific commentary refers to ‘O-GlcNAcylation regulates dopamine neuron function, survival and degeneration in Parkinson disease’, by Lee et al. (doi:10.1093/brain/awaa320).
Collapse
Affiliation(s)
- Gerald W Hart
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Chia-Wei Huang
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
35
|
Martínez-Viturro CM, Trabanco AA, Royes J, Fernández E, Tresadern G, Vega JA, del Cerro A, Delgado F, García Molina A, Tovar F, Shaffer P, Ebneth A, Bretteville A, Mertens L, Somers M, Alonso JM, Bartolomé-Nebreda JM. Diazaspirononane Nonsaccharide Inhibitors of O-GlcNAcase (OGA) for the Treatment of Neurodegenerative Disorders. J Med Chem 2020; 63:14017-14044. [DOI: 10.1021/acs.jmedchem.0c01479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Carlos M. Martínez-Viturro
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Andrés A. Trabanco
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Jordi Royes
- Department Química Física i Inorgànica, University Rovira i Virgili, C/Marcel·lí Domingo s/n, 43007 Tarragona, Spain
| | - Elena Fernández
- Department Química Física i Inorgànica, University Rovira i Virgili, C/Marcel·lí Domingo s/n, 43007 Tarragona, Spain
| | - Gary Tresadern
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Juan A. Vega
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Alcira del Cerro
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Francisca Delgado
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Aránzazu García Molina
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Fulgencio Tovar
- Villapharma Research S.L., Parque Tecnológico de Fuente Álamo, Ctra. El Estrecho-Lobosillo, Km. 2.5—Av. Azul, 30320 Fuente Álamo de Murcia, Spain
| | - Paul Shaffer
- X-Ray Crystallography, Janssen Pharmaceutical Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Andreas Ebneth
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Alexis Bretteville
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Liesbeth Mertens
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Marijke Somers
- Discovery DMPK, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Jose M. Alonso
- Analytical Sciences, Janssen Research & Development, Janssen-Cilag, S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - José M. Bartolomé-Nebreda
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| |
Collapse
|
36
|
Joseph CR. Novel MRI Techniques Identifying Vascular Leak and Paravascular Flow Reduction in Early Alzheimer Disease. Biomedicines 2020; 8:biomedicines8070228. [PMID: 32698354 PMCID: PMC7400582 DOI: 10.3390/biomedicines8070228] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
With beta amyloid and tau antibody treatment trial failures, avenues directed to other facets of the disease pathophysiology are being explored to treat in the preclinical or early clinical state. Clear evidence of blood–brain barrier (BBB) breakdown occurring early in the AD process has recently been established. Likewise, the glymphatic system regulating water and solute inflow and outflow in parallel with the vascular system is affected causing delayed clearance of fluid waste. Its dysfunction as a component of AD along with BBB leak are reasonable candidates to explore for future treatments. Ideally, human medication trials require a minimally invasive method of quantifying both improvements in BBB integrity and glymphatic fluid clearance correlated with clinical outcomes. We will review the known physiology and anatomy of the BBB system, and its relationship to the glymphatic system and the microglial surveillance system. Dysfunction of this tripart system occurring in preclinical Alzheimer disease (AD) will be reviewed along with existing MRI tools for identifying altered flow dynamics useful for monitoring improved functionality with future treatments. High-resolution dynamic contrast enhanced MRI imaging demonstrating BBB leak and the recently reported non-invasive 3D PASL MRI pilot study demonstrating significant delay in glymphatic clearance in AD subjects appear to be the best candidates.
Collapse
Affiliation(s)
- Charles R Joseph
- Department of Internal Medicine, Liberty University College of Osteopathic Medicine, Lynchburg, VA 24502, USA
| |
Collapse
|
37
|
Elbatrawy AA, Kim EJ, Nam G. O‐GlcNAcase: Emerging Mechanism, Substrate Recognition and Small‐Molecule Inhibitors. ChemMedChem 2020; 15:1244-1257. [DOI: 10.1002/cmdc.202000077] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/22/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Ahmed A. Elbatrawy
- Center for Neuro-Medicine Brain Science Institute Korea Institutes of Science and Technology Seoul 02792 (Republic of Korea
- Division of Bio-Med KIST school Korea University of Science and Technology (UST) Gajungro 217 Youseong-gu Daejeon (Republic of Korea
| | - Eun Ju Kim
- Daegu University Department of Science Education-Chemistry Gyeongsan-si, Gyeongsangbuk-do Gyeongbuk 38453 (Republic of Korea
| | - Ghilsoo Nam
- Center for Neuro-Medicine Brain Science Institute Korea Institutes of Science and Technology Seoul 02792 (Republic of Korea
- Division of Bio-Med KIST school Korea University of Science and Technology (UST) Gajungro 217 Youseong-gu Daejeon (Republic of Korea
| |
Collapse
|
38
|
Wang X, Li W, Marcus J, Pearson M, Song L, Smith K, Terracina G, Lee J, Hong KLK, Lu SX, Hyde L, Chen SC, Kinsley D, Melchor JP, Rubins DJ, Meng X, Hostetler E, Sur C, Zhang L, Schachter JB, Hess JF, Selnick HG, Vocadlo DJ, McEachern EJ, Uslaner JM, Duffy JL, Smith SM. MK-8719, a Novel and Selective O-GlcNAcase Inhibitor That Reduces the Formation of Pathological Tau and Ameliorates Neurodegeneration in a Mouse Model of Tauopathy. J Pharmacol Exp Ther 2020; 374:252-263. [PMID: 32493725 DOI: 10.1124/jpet.120.266122] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/01/2020] [Indexed: 01/01/2023] Open
Abstract
Deposition of hyperphosphorylated and aggregated tau protein in the central nervous system is characteristic of Alzheimer disease and other tauopathies. Tau is subject to O-linked N-acetylglucosamine (O-GlcNAc) modification, and O-GlcNAcylation of tau has been shown to influence tau phosphorylation and aggregation. Inhibition of O-GlcNAcase (OGA), the enzyme that removes O-GlcNAc moieties, is a novel strategy to attenuate the formation of pathologic tau. Here we described the in vitro and in vivo pharmacological properties of a novel and selective OGA inhibitor, MK-8719. In vitro, this compound is a potent inhibitor of the human OGA enzyme with comparable activity against the corresponding enzymes from mouse, rat, and dog. In vivo, oral administration of MK-8719 elevates brain and peripheral blood mononuclear cell O-GlcNAc levels in a dose-dependent manner. In addition, positron emission tomography imaging studies demonstrate robust target engagement of MK-8719 in the brains of rats and rTg4510 mice. In the rTg4510 mouse model of human tauopathy, MK-8719 significantly increases brain O-GlcNAc levels and reduces pathologic tau. The reduction in tau pathology in rTg4510 mice is accompanied by attenuation of brain atrophy, including reduction of forebrain volume loss as revealed by volumetric magnetic resonance imaging analysis. These findings suggest that OGA inhibition may reduce tau pathology in tauopathies. However, since hundreds of O-GlcNAcylated proteins may be influenced by OGA inhibition, it will be critical to understand the physiologic and toxicological consequences of chronic O-GlcNAc elevation in vivo. SIGNIFICANCE STATEMENT: MK-8719 is a novel, selective, and potent O-linked N-acetylglucosamine (O-GlcNAc)-ase (OGA) inhibitor that inhibits OGA enzyme activity across multiple species with comparable in vitro potency. In vivo, MK-8719 elevates brain O-GlcNAc levels, reduces pathological tau, and ameliorates brain atrophy in the rTg4510 mouse model of tauopathy. These findings indicate that OGA inhibition may be a promising therapeutic strategy for the treatment of Alzheimer disease and other tauopathies.
Collapse
Affiliation(s)
- Xiaohai Wang
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Wenping Li
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Jacob Marcus
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Michelle Pearson
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Lixin Song
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Karen Smith
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Giuseppe Terracina
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Julie Lee
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Kwok-Lam Karen Hong
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Sherry X Lu
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Lynn Hyde
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Shu-Cheng Chen
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - David Kinsley
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Jerry P Melchor
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Daniel J Rubins
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Xiangjun Meng
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Eric Hostetler
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Cyrille Sur
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Lili Zhang
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Joel B Schachter
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - J Fred Hess
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Harold G Selnick
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - David J Vocadlo
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Ernest J McEachern
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Jason M Uslaner
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Joseph L Duffy
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| | - Sean M Smith
- MRL, Merck & Co., Inc., Kenilworth, New Jersey (X.W., W.L., J.M., M.P., L.S., K.S., G.T., J.L., K.-L.K.H., S.X.L., L.H., S.-C.C., D.K., J.P.M., D.J.R., X.M., E.H., C.S., L.Z., J.B.S., J.F.H., H.G.S., J.M.U., J.L.D., S.M.S.) and Alectos Therapeutics Inc., Burnaby, British Columbia, Canada (D.J.V., E.J.M.)
| |
Collapse
|
39
|
Sáez-Orellana F, Octave JN, Pierrot N. Alzheimer's Disease, a Lipid Story: Involvement of Peroxisome Proliferator-Activated Receptor α. Cells 2020; 9:E1215. [PMID: 32422896 PMCID: PMC7290654 DOI: 10.3390/cells9051215] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Mutations in genes encoding proteins involved in amyloid-β peptide (Aβ) production are responsible for inherited AD cases. The amyloid cascade hypothesis was proposed to explain the pathogeny. Despite the fact that Aβ is considered as the main culprit of the pathology, most clinical trials focusing on Aβ failed and suggested that earlier interventions are needed to influence the course of AD. Therefore, identifying risk factors that predispose to AD is crucial. Among them, the epsilon 4 allele of the apolipoprotein E gene that encodes the major brain lipid carrier and metabolic disorders such as obesity and type 2 diabetes were identified as AD risk factors, suggesting that abnormal lipid metabolism could influence the progression of the disease. Among lipids, fatty acids (FAs) play a fundamental role in proper brain function, including memory. Peroxisome proliferator-activated receptor α (PPARα) is a master metabolic regulator that regulates the catabolism of FA. Several studies report an essential role of PPARα in neuronal function governing synaptic plasticity and cognition. In this review, we explore the implication of lipid metabolism in AD, with a special focus on PPARα and its potential role in AD therapy.
Collapse
Affiliation(s)
- Francisco Sáez-Orellana
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Jean-Noël Octave
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Nathalie Pierrot
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| |
Collapse
|
40
|
Lu S, Yin X, Wang J, Gu Q, Huang Q, Jin N, Chu D, Xu Z, Liu F, Qian W. SIRT1 regulates O-GlcNAcylation of tau through OGT. Aging (Albany NY) 2020; 12:7042-7055. [PMID: 32310828 PMCID: PMC7202539 DOI: 10.18632/aging.103062] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/09/2020] [Indexed: 04/19/2023]
Abstract
Tau is modified with O-GlcNAcylation extensively in human brain. The O-GlcNAcylation levels of tau are decreased in Alzheimer's disease (AD) brain. Sirtuin type 1 (SIRT1) is an enzyme that deacetylates proteins including transcriptional factors and associates with neurodegenerative diseases, such as AD. Aberrant SIRT1 expression levels in AD brain is in parallel with the accumulation of tau. cAMP response element binding protein (CREB), a cellular transcription factor, plays a critical role in learning and memory. In this present study, we found SIRT1 deacetylates CREB and inhibits phosphorylation of CREB at Ser133. The inactivated CREB suppresses OGT expression and therefore decreases the O-GlcNAcylation of tau and thus increases the phosphorylation of tau at specific sites. These findings suggest that SIRT1 may be a potential therapeutic target for treating tauopathies.
Collapse
Affiliation(s)
- Shu Lu
- Department of Intensive Care Unit, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, P. R. China
| | - Xiaomin Yin
- Department of Biochemistry and Molecular Biology, Medical School, Nantong, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
| | - Jia Wang
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
| | - Qun Gu
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
| | - Qin Huang
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
| | - Nana Jin
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
| | - Dandan Chu
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
| | - Ziqi Xu
- Department of Biochemistry and Molecular Biology, Medical School, Nantong, Jiangsu, P. R. China
| | - Fei Liu
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Wei Qian
- Department of Biochemistry and Molecular Biology, Medical School, Nantong, Jiangsu, P. R. China
- Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, P. R. China
| |
Collapse
|
41
|
Ogawa M, Tashima Y, Sakaguchi Y, Takeuchi H, Okajima T. Contribution of extracellular O-GlcNAc to the stability of folded epidermal growth factor-like domains and Notch1 trafficking. Biochem Biophys Res Commun 2020; 526:184-190. [PMID: 32201074 DOI: 10.1016/j.bbrc.2020.03.066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/10/2020] [Indexed: 01/05/2023]
Abstract
The Notch signaling pathway is highly conserved and essential in animal development and tissue homeostasis. Regulation of Notch signaling is a crucial process for human health. Ligands initiate a signal cascade by binding to Notch receptors expressed on the neighboring cell. Notch receptors interact with ligands through their epidermal growth factor-like repeats (EGF repeats). Most EGF repeats are modified by O-glycosylation with residues, such as O-linked N-acetylglucosamine (O-GlcNAc), O-fucose, and O-glucose. A recent study revealed the distinct roles of these O-glycans in ligand binding, processing, and trafficking of Notch receptors. In particular, O-GlcNAc glycans are essential for Delta-like (DLL) ligand-mediated Notch signaling. In this study, we showed that O-GlcNAc promotes Notch1 trafficking to the cell surfaces under the condition that O-fucose and O-glucose are removed from consecutive EGF repeats of Notch1. Through in vitro experiments, we showed that O-GlcNAc mediates the stability of EGF domains in the same manner as O-fucose and O-glucose. Thus, O-GlcNAc on EGF domains possesses a shared function in the stability of EGF domains and Notch1 trafficking.
Collapse
Affiliation(s)
- Mitsutaka Ogawa
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan
| | - Yuko Tashima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan
| | - Yamato Sakaguchi
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan
| | - Hideyuki Takeuchi
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan
| | - Tetsuya Okajima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
42
|
Lee JH, Liow JS, Paul S, Morse CL, Haskali MB, Manly L, Shcherbinin S, Ruble JC, Kant N, Collins EC, Nuthall HN, Zanotti-Fregonara P, Zoghbi SS, Pike VW, Innis RB. PET quantification of brain O-GlcNAcase with [ 18F]LSN3316612 in healthy human volunteers. EJNMMI Res 2020; 10:20. [PMID: 32172476 PMCID: PMC7072082 DOI: 10.1186/s13550-020-0616-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/02/2020] [Indexed: 01/11/2023] Open
Abstract
Background Previous studies found that [18F]LSN3316612 was a promising positron emission tomography (PET) radioligand for imaging O-GlcNAcase in nonhuman primates and human volunteers. This study sought to further evaluate the suitability of [18F]LSN3316612 for human clinical research. Methods Kinetic evaluation of [18F]LSN3316612 was conducted in a combined set of baseline brain scans from 17 healthy human volunteers and test-retest imaging was conducted in 10 of these volunteers; another 6 volunteers had whole-body scans to measure radiation exposure to body organs. Total distribution volume (VT) estimates were compared for the one- and two-tissue compartment models with the arterial input function. Test-retest variability and reliability were evaluated via mean difference and intraclass correlation coefficient (ICC). The time stability of VT was assessed down to a 30-min scan time. An alternative quantification method for [18F]LSN3316612 binding without blood was also investigated to assess the possibility of eliminating arterial sampling. Results Brain uptake was generally high and could be quantified as VT with excellent identifiability using the two-tissue compartment model. [18F]LSN3316612 exhibited good absolute test-retest variability (12.5%), but the arithmetic test-retest variability was far from 0 (11.3%), reflecting a near-uniform increase of VT on the retest scan in nine of 10 volunteers. VT values were stable after 110 min in all brain regions, suggesting that no radiometabolites accumulated in the brain. Measurements obtained using only brain activity (i.e., area under the curve (AUC) from 150–180 min) correlated strongly with regional VT values during test-retest conditions (R2 = 0.84), exhibiting similar reliability to VT (ICC = 0.68 vs. 0.64). Estimated radiation exposure for [18F]LSN3316612 PET was 20.5 ± 2.1 μSv/MBq, comparable to other 18F-labeled radioligands for brain imaging. Conclusions [18F]LSN3316612 is an excellent PET radioligand for imaging O-GlcNAcase in the human brain. Alternative quantification without blood is possible, at least for within-subject repeat studies. However, the unexplained increase of VT under retest conditions requires further investigation.
Collapse
Affiliation(s)
- Jae-Hoon Lee
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA. .,Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, South Korea.
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Soumen Paul
- Molecular Imaging Core, University of Virginia, Charlottesville, VA, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Mohammad B Haskali
- The Centre for Molecular Imaging and Translational Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Lester Manly
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | | | | | - Nancy Kant
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | | | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
43
|
Much More Than a Scaffold: Cytoskeletal Proteins in Neurological Disorders. Cells 2020; 9:cells9020358. [PMID: 32033020 PMCID: PMC7072452 DOI: 10.3390/cells9020358] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/08/2023] Open
Abstract
Recent observations related to the structure of the cytoskeleton in neurons and novel cytoskeletal abnormalities involved in the pathophysiology of some neurological diseases are changing our view on the function of the cytoskeletal proteins in the nervous system. These efforts allow a better understanding of the molecular mechanisms underlying neurological diseases and allow us to see beyond our current knowledge for the development of new treatments. The neuronal cytoskeleton can be described as an organelle formed by the three-dimensional lattice of the three main families of filaments: actin filaments, microtubules, and neurofilaments. This organelle organizes well-defined structures within neurons (cell bodies and axons), which allow their proper development and function through life. Here, we will provide an overview of both the basic and novel concepts related to those cytoskeletal proteins, which are emerging as potential targets in the study of the pathophysiological mechanisms underlying neurological disorders.
Collapse
|
44
|
Ruegenberg S, Horn M, Pichlo C, Allmeroth K, Baumann U, Denzel MS. Loss of GFAT-1 feedback regulation activates the hexosamine pathway that modulates protein homeostasis. Nat Commun 2020; 11:687. [PMID: 32019926 PMCID: PMC7000685 DOI: 10.1038/s41467-020-14524-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 01/14/2020] [Indexed: 01/03/2023] Open
Abstract
Glutamine fructose-6-phosphate amidotransferase (GFAT) is the key enzyme in the hexosamine pathway (HP) that produces uridine 5′-diphospho-N-acetyl-d-glucosamine (UDP-GlcNAc), linking energy metabolism with posttranslational protein glycosylation. In Caenorhabditis elegans, we previously identified gfat-1 gain-of-function mutations that elevate UDP-GlcNAc levels, improve protein homeostasis, and extend lifespan. GFAT is highly conserved, but the gain-of-function mechanism and its relevance in mammalian cells remained unclear. Here, we present the full-length crystal structure of human GFAT-1 in complex with various ligands and with important mutations. UDP-GlcNAc directly interacts with GFAT-1, inhibiting catalytic activity. The longevity-associated G451E variant shows drastically reduced sensitivity to UDP-GlcNAc inhibition in enzyme activity assays. Our structural and functional data point to a critical role of the interdomain linker in UDP-GlcNAc inhibition. In mammalian cells, the G451E variant potently activates the HP. Therefore, GFAT-1 gain-of-function through loss of feedback inhibition constitutes a potential target for the treatment of age-related proteinopathies. Mutations in the hexosamine pathway key enzyme glutamine fructose-6-phosphate amidotransferase (GFAT-1) improve protein quality control and extend C. elegans lifespan. Here the authors present the crystal structures of full-length human GFAT-1 alone and with bound ligands and perform activity assays, which show that gain-of-function in the longevity-associated G451E variant is caused by a loss of feedback regulation.
Collapse
Affiliation(s)
- Sabine Ruegenberg
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany.,University of Cologne, Institute of Biochemistry, 50674, Cologne, Germany
| | - Moritz Horn
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Christian Pichlo
- University of Cologne, Institute of Biochemistry, 50674, Cologne, Germany
| | - Kira Allmeroth
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Ulrich Baumann
- University of Cologne, Institute of Biochemistry, 50674, Cologne, Germany.
| | - Martin S Denzel
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany. .,CECAD-Cluster of Excellence, University of Cologne, 50931, Cologne, Germany. .,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
45
|
Guo J, Zhang G, Ma J, Zhao C, Xue Q, Wang J, Liu W, Liu K, Wang H, Liu N, Song Q, Li J. Detection and identification of O-GlcNAc-modified proteins using 6-azido-6-deoxy-N-acetyl-galactosamine. Org Biomol Chem 2019; 17:4326-4334. [PMID: 30976765 DOI: 10.1039/c9ob00516a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
An unnatural monosaccharide with a C6-azide, Ac36AzGalNAc, has been developed as a potent and selective probe for O-GlcNAc-modified proteins. Combined with click chemistry, we demonstrate that Ac36AzGalNAc can robustly label O-GlcNAc glycosylation in a wide range of cell lines. Meanwhile, cell imaging and LC-MS/MS proteomics verify its selective activity on O-GlcNAc. More importantly, the protocol presented here provides a general methodology for tracking, capturing and identifying unnatural monosaccharide modified proteins in cells or cell lysates.
Collapse
Affiliation(s)
- Jianshuang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Selnick HG, Hess JF, Tang C, Liu K, Schachter JB, Ballard JE, Marcus J, Klein DJ, Wang X, Pearson M, Savage MJ, Kaul R, Li TS, Vocadlo DJ, Zhou Y, Zhu Y, Mu C, Wang Y, Wei Z, Bai C, Duffy JL, McEachern EJ. Discovery of MK-8719, a Potent O-GlcNAcase Inhibitor as a Potential Treatment for Tauopathies. J Med Chem 2019; 62:10062-10097. [DOI: 10.1021/acs.jmedchem.9b01090] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Harold G. Selnick
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - J. Fred Hess
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Cuyue Tang
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kun Liu
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Joel B. Schachter
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jeanine E. Ballard
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jacob Marcus
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Daniel J. Klein
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaohai Wang
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michelle Pearson
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Mary J. Savage
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ramesh Kaul
- Alectos Therapeutics Inc., 8999 Nelson Way, Burnaby, British Columbia V5A 4B5, Canada
| | - Tong-Shuang Li
- Alectos Therapeutics Inc., 8999 Nelson Way, Burnaby, British Columbia V5A 4B5, Canada
| | - David J. Vocadlo
- Alectos Therapeutics Inc., 8999 Nelson Way, Burnaby, British Columbia V5A 4B5, Canada
| | - Yuanxi Zhou
- Alectos Therapeutics Inc., 8999 Nelson Way, Burnaby, British Columbia V5A 4B5, Canada
| | - Yongbao Zhu
- Alectos Therapeutics Inc., 8999 Nelson Way, Burnaby, British Columbia V5A 4B5, Canada
| | - Changwei Mu
- Pharmaron Beijing Co. Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Yaode Wang
- Pharmaron Beijing Co. Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Zhongyong Wei
- Pharmaron Beijing Co. Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Chang Bai
- Pharmaron Beijing Co. Ltd., 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - Joseph L. Duffy
- Merck & Co., Inc., 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ernest J. McEachern
- Alectos Therapeutics Inc., 8999 Nelson Way, Burnaby, British Columbia V5A 4B5, Canada
| |
Collapse
|
47
|
Chen S, Zhou M, Sun J, Guo A, Fernando RL, Chen Y, Peng P, Zhao G, Deng Y. DPP-4 inhibitor improves learning and memory deficits and AD-like neurodegeneration by modulating the GLP-1 signaling. Neuropharmacology 2019; 157:107668. [PMID: 31199957 DOI: 10.1016/j.neuropharm.2019.107668] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) signaling in the brain plays an important role in the regulation of glucose metabolism, which is impaired in Alzheimer's disease (AD). Here, we detected the GLP-1 and GLP-1 receptor (GLP-1R) in AD human brain and APP/PS1/Tau transgenic (3xTg) mice brain, finding that they were both decreased in AD human and mice brain. Enhanced GLP-1 exerts its protective effects on AD, however, this is rapidly degraded into inactivated metabolites by dipeptidyl peptidase-4 (DPP-4), resulting in its extremely short half-time. DPP-4 inhibitors, thus, was applied to improve the level of GLP-1 and GLP-1R expression in the hippocampus and cortex of AD mice brains. It is also protected learning and memory and synaptic proteins, increased the O-Glycosylation and decreased abnormal phosphorylation of tau and neurofilaments (NFs), degraded intercellular β-amyloid (Aβ) accumulation and alleviated neurodegeneration related to GLP-1 signaling pathway.
Collapse
Affiliation(s)
- Shuyi Chen
- Pathophysiology Department, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mei Zhou
- Pathophysiology Department, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jie Sun
- Department of Pathology, Tianjin People's Hospital, Tianjin, China
| | - Ai Guo
- Pathophysiology Department, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Roger Lakmal Fernando
- Pathophysiology Department, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanlin Chen
- Pathophysiology Department, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Peng Peng
- Pathophysiology Department, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Gang Zhao
- Department of Pathology, Tianjin Tumor Hospital, Tianjin Medical University, Tianjin, China
| | - Yanqiu Deng
- Pathophysiology Department, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
48
|
Ansari SA, Emerald BS. The Role of Insulin Resistance and Protein O-GlcNAcylation in Neurodegeneration. Front Neurosci 2019; 13:473. [PMID: 31143098 PMCID: PMC6521730 DOI: 10.3389/fnins.2019.00473] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome including obesity and type 2 diabetes is increasing at an alarming rate worldwide. Similarly, there has been an increase in the cases of neurodegenerative diseases such as Alzheimer’s disease (AD) possibility due to increase in elderly population in the past few decades. Both, metabolic diseases and AD have one common feature that is insulin resistance. Recent studies suggest a link between the regulatory functions of insulin in the brain and AD. Hypoglycemia, a characteristic feature of AD may be a result of impaired insulin signaling in the affected regions of the brain. O-GlcNAcylation is a post-translational protein modification, the levels of which are dependent on the availability of glucose inside the cells. Hyperphosphorylation of Tau is a major molecular feature, which leads to its aggregation and neurotoxicity in AD. In addition, impaired processing of Amyloid precursor protein (APP) leading to toxic amyloid β (Aβ) aggregation is also implicated in the pathogenesis of AD. Both APP and Tau are also found to be O-GlcNAcylated. Reduced O-GlcNAcylation of APP and Tau due to hypoglycemia is found to be associated with their pathological features in AD brain. Recent studies have also identified perturbed O-GlcNAcylation/phosphorylation of several other proteins important for normal neuronal function, which may be contributing to the neuropathological development in AD. Herein, we discuss about the uptake and distribution of insulin inside the brain, brain insulin signaling and insulin resistance as well as its relation to neurodegenerative diseases with a special focus on protein O-GlcNAcylation and its potential role in the treatment of AD.
Collapse
Affiliation(s)
- Suraiya A Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
49
|
Abstract
In the early 1980s, while using purified glycosyltransferases to probe glycan structures on surfaces of living cells in the murine immune system, we discovered a novel form of serine/threonine protein glycosylation (O-linked β-GlcNAc; O-GlcNAc) that occurs on thousands of proteins within the nucleus, cytoplasm, and mitochondria. Prior to this discovery, it was dogma that protein glycosylation was restricted to the luminal compartments of the secretory pathway and on extracellular domains of membrane and secretory proteins. Work in the last 3 decades from several laboratories has shown that O-GlcNAc cycling serves as a nutrient sensor to regulate signaling, transcription, mitochondrial activity, and cytoskeletal functions. O-GlcNAc also has extensive cross-talk with phosphorylation, not only at the same or proximal sites on polypeptides, but also by regulating each other's enzymes that catalyze cycling of the modifications. O-GlcNAc is generally not elongated or modified. It cycles on and off polypeptides in a time scale similar to phosphorylation, and both the enzyme that adds O-GlcNAc, the O-GlcNAc transferase (OGT), and the enzyme that removes O-GlcNAc, O-GlcNAcase (OGA), are highly conserved from C. elegans to humans. Both O-GlcNAc cycling enzymes are essential in mammals and plants. Due to O-GlcNAc's fundamental roles as a nutrient and stress sensor, it plays an important role in the etiologies of chronic diseases of aging, including diabetes, cancer, and neurodegenerative disease. This review will present an overview of our current understanding of O-GlcNAc's regulation, functions, and roles in chronic diseases of aging.
Collapse
Affiliation(s)
- Gerald W Hart
- From the Complex Carbohydrate Research Center and Biochemistry and Molecular Biology Department, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
50
|
Ellmer D, Brehs M, Haj‐Yahya M, Lashuel HA, Becker CFW. Single Posttranslational Modifications in the Central Repeat Domains of Tau4 Impact its Aggregation and Tubulin Binding. Angew Chem Int Ed Engl 2019; 58:1616-1620. [PMID: 30549369 PMCID: PMC6391969 DOI: 10.1002/anie.201805238] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/29/2018] [Indexed: 12/22/2022]
Abstract
A variety of methods have been employed to study the impact of posttranslational modifications on Tau protein function. Here, a semisynthesis strategy is described that enables selective modification within the central repeat domain of Tau4 (residues 291-321), comprising a major interaction motive with tubulin as well as one of the key hexapeptides involved in Tau aggregation. This strategy has led to the preparation of four semisynthetic Tau variants with phosphoserine residues in different positions and one with a so far largely ignored carboxymethyllysine modification that results from a non-enzymatic posttranslational modification (nPTM). The latter modification inhibits tubulin polymerization but exhibits an aggregation behavior very similar to unmodified Tau. In contrast, phosphorylated Tau variants exhibit similar binding to tubulin as unmodified Tau4 but show lower tendencies to aggregate.
Collapse
Affiliation(s)
- Doris Ellmer
- University of ViennaFaculty of ChemistryInstitute of Biological ChemistryWähringer Str. 381090ViennaAustria
| | - Manuel Brehs
- University of ViennaFaculty of ChemistryInstitute of Biological ChemistryWähringer Str. 381090ViennaAustria
| | - Mahmood Haj‐Yahya
- École Polytechnique Fédérale de Lausanne (EPFL), Brain Mind InstituteLaboratory of Molecular and Chemical Biology of Neurodegeneration1015LausanneSwitzerland
| | - Hilal A. Lashuel
- École Polytechnique Fédérale de Lausanne (EPFL), Brain Mind InstituteLaboratory of Molecular and Chemical Biology of Neurodegeneration1015LausanneSwitzerland
| | - Christian F. W. Becker
- University of ViennaFaculty of ChemistryInstitute of Biological ChemistryWähringer Str. 381090ViennaAustria
| |
Collapse
|