1
|
Ali I, Ali MM, Liu Q, Hu L. Unraveling Clinical Glycoproteome by Integrating Affinity Enrichment with Nanopore Sequencing. Chembiochem 2024; 25:e202400419. [PMID: 39234982 DOI: 10.1002/cbic.202400419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/28/2024] [Indexed: 09/06/2024]
Abstract
This prospect explores the integration of enrichment strategies with nanopore detection to advance clinical glycoproteomics. Glycoproteins, crucial for understanding biological processes, pose challenges due to their low abundance and structural diversity. Enrichment techniques using lectin affinity, boronate affinity, and hydrazide chemistry and especially molecular imprinted polymers may selectively and specifically isolate glycoproteins from complex samples, while nanopore technology enables label-free, real-time, and single-molecule analysis. This approach holds promise for disease-related glycosylation studies, biomarker discovery, personalized medicine, and streamlined clinical analysis. Standardization, optimization, and data analysis remain challenges, requiring interdisciplinary collaborations and technological advancements. Overall, this integration may offer transformative potential for clinical glycoproteomics and innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Irshad Ali
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China
| | - Muhammad Mujahid Ali
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, 130012, China
- Department of Biochemistry, Purdue University, West Lafayette, IN 47906, US
| | - Quanjun Liu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China
| | - Lianghai Hu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, 130012, China
| |
Collapse
|
2
|
Morales MM, Pratt MR. The post-translational modification O-GlcNAc is a sensor and regulator of metabolism. Open Biol 2024; 14:240209. [PMID: 39474868 PMCID: PMC11523104 DOI: 10.1098/rsob.240209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Cells must rapidly adapt to changes in nutrient conditions through responsive signalling cascades to maintain homeostasis. One of these adaptive pathways results in the post-translational modification of proteins by O-GlcNAc. O-GlcNAc modifies thousands of nuclear and cytoplasmic proteins in response to nutrient availability through the hexosamine biosynthetic pathway. O-GlcNAc is highly dynamic and can be added and removed from proteins multiple times throughout their life cycle, setting it up to be an ideal regulator of cellular processes in response to metabolic changes. Here, we describe the link between cellular metabolism and O-GlcNAc, and we explore O-GlcNAc's role in regulating cellular processes in response to nutrient levels. Specifically, we discuss the mechanisms of elevated O-GlcNAc levels in contributing to diabetes and cancer, as well as the role of decreased O-GlcNAc levels in neurodegeneration. These studies form a foundational understanding of aberrant O-GlcNAc in human disease and provide an opportunity to further improve disease identification and treatment.
Collapse
Affiliation(s)
- Murielle M. Morales
- Department of Biological Sciences, University of Southern California, Los Angeles, CA90089, USA
| | - Matthew R. Pratt
- Department of Chemistry, University of Southern California, Los Angeles, CA90089, USA
| |
Collapse
|
3
|
Mittal S, Jena MK, Pathak B. Integration of Artificial Intelligence and Quantum Transport toward Stereoselective Identification of Carbohydrate Isomers. ACS CENTRAL SCIENCE 2024; 10:1689-1702. [PMID: 39345811 PMCID: PMC11428302 DOI: 10.1021/acscentsci.4c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/24/2024] [Accepted: 07/26/2024] [Indexed: 10/01/2024]
Abstract
Detection of stereoisomers of carbohydrates with molecular resolution, a challenging goal analysts desire to achieve, is key to the full development of glycosciences. Despite the promise that analytical techniques made, including widely used nuclear magnetic resonance and mass spectrometry, high throughput de novo carbohydrate sequencing remains an unsolved issue. Notably, while next-generation sequencing technologies are readily available for DNA and proteins, they are conspicuously absent for carbohydrates due to the immense stereochemical and structural complexity inherent in these molecules. In this work, we report a novel computational technique that employs quantum tunneling coupled with artificial intelligence to detect complex carbohydrate anomers and stereoisomers with excellent sensitivity. The quantum tunneling footprints of carbohydrate isomers show high distinguishability with an in-depth analysis of underlying chemistry. Our findings open up a new route for carbohydrate sensing, which can be seamlessly integrated with next-generation sequencing technology for real-time analysis.
Collapse
Affiliation(s)
- Sneha Mittal
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh 453552, India
| | - Milan Kumar Jena
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh 453552, India
| | - Biswarup Pathak
- Department of Chemistry, Indian Institute of Technology (IIT) Indore, Indore, Madhya Pradesh 453552, India
| |
Collapse
|
4
|
Sheng L, Bhalla R. Biomarkers and Target-Specific Small-Molecule Drugs in Alzheimer's Diagnostic and Therapeutic Research: From Amyloidosis to Tauopathy. Neurochem Res 2024; 49:2273-2302. [PMID: 38844706 PMCID: PMC11310295 DOI: 10.1007/s11064-024-04178-w] [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: 01/16/2024] [Revised: 03/31/2024] [Accepted: 05/22/2024] [Indexed: 08/09/2024]
Abstract
Alzheimer's disease (AD) is the most common type of human dementia and is responsible for over 60% of diagnosed dementia cases worldwide. Abnormal deposition of β-amyloid and the accumulation of neurofibrillary tangles have been recognised as the two pathological hallmarks targeted by AD diagnostic imaging as well as therapeutics. With the progression of pathological studies, the two hallmarks and their related pathways have remained the focus of researchers who seek for AD diagnostic and therapeutic strategies in the past decades. In this work, we reviewed the development of the AD biomarkers and their corresponding target-specific small molecule drugs for both diagnostic and therapeutic applications, underlining their success, failure, and future possibilities.
Collapse
Affiliation(s)
- Li Sheng
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
5
|
Wang H, Vant JW, Zhang A, Sanchez RG, Wu Y, Micou ML, Luczak V, Whiddon Z, Carlson NM, Yu SB, Jabbo M, Yoon S, Abushawish AA, Ghassemian M, Masubuchi T, Gan Q, Watanabe S, Griffis ER, Hammarlund M, Singharoy A, Pekkurnaz G. Organization of a functional glycolytic metabolon on mitochondria for metabolic efficiency. Nat Metab 2024; 6:1712-1735. [PMID: 39261628 DOI: 10.1038/s42255-024-01121-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
Glucose, the primary cellular energy source, is metabolized through glycolysis initiated by the rate-limiting enzyme hexokinase (HK). In energy-demanding tissues like the brain, HK1 is the dominant isoform, primarily localized on mitochondria, and is crucial for efficient glycolysis-oxidative phosphorylation coupling and optimal energy generation. This study unveils a unique mechanism regulating HK1 activity, glycolysis and the dynamics of mitochondrial coupling, mediated by the metabolic sensor enzyme O-GlcNAc transferase (OGT). OGT catalyses reversible O-GlcNAcylation, a post-translational modification influenced by glucose flux. Elevated OGT activity induces dynamic O-GlcNAcylation of the regulatory domain of HK1, subsequently promoting the assembly of the glycolytic metabolon on the outer mitochondrial membrane. This modification enhances the mitochondrial association with HK1, orchestrating glycolytic and mitochondrial ATP production. Mutation in HK1's O-GlcNAcylation site reduces ATP generation in multiple cell types, specifically affecting metabolic efficiency in neurons. This study reveals a previously unappreciated pathway that links neuronal metabolism and mitochondrial function through OGT and the formation of the glycolytic metabolon, providing potential strategies for tackling metabolic and neurological disorders.
Collapse
Affiliation(s)
- Haoming Wang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - John W Vant
- Biodesign Institute, The School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Andrew Zhang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Richard G Sanchez
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Youjun Wu
- Department of Genetics and Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Mary L Micou
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Thomas Jefferson University, Philadelphia, PA, USA
| | - Vincent Luczak
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Neurocrine Biosciences, San Diego, CA, USA
| | - Zachary Whiddon
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Natasha M Carlson
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Seungyoon B Yu
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Mirna Jabbo
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Seokjun Yoon
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- University of Southern California, Los Angeles, CA, USA
| | - Ahmed A Abushawish
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, La Jolla, CA, USA
| | - Takeya Masubuchi
- Cell and Developmental Biology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Quan Gan
- Department of Cell Biology, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Shigeki Watanabe
- Department of Cell Biology, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Eric R Griffis
- Nikon Imaging Center, University of California San Diego, La Jolla, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Marc Hammarlund
- Department of Genetics and Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Abhishek Singharoy
- Biodesign Institute, The School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Gulcin Pekkurnaz
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Yu SB, Wang H, Sanchez RG, Carlson NM, Nguyen K, Zhang A, Papich ZD, Abushawish AA, Whiddon Z, Matysik W, Zhang J, Whisenant TC, Ghassemian M, Koberstein JN, Stewart ML, Myers SA, Pekkurnaz G. Neuronal activity-driven O-GlcNAcylation promotes mitochondrial plasticity. Dev Cell 2024; 59:2143-2157.e9. [PMID: 38843836 PMCID: PMC11338717 DOI: 10.1016/j.devcel.2024.05.008] [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: 01/12/2023] [Revised: 02/15/2024] [Accepted: 05/09/2024] [Indexed: 06/18/2024]
Abstract
Neuronal activity is an energy-intensive process that is largely sustained by instantaneous fuel utilization and ATP synthesis. However, how neurons couple ATP synthesis rate to fuel availability is largely unknown. Here, we demonstrate that the metabolic sensor enzyme O-linked N-acetyl glucosamine (O-GlcNAc) transferase regulates neuronal activity-driven mitochondrial bioenergetics in hippocampal and cortical neurons. We show that neuronal activity upregulates O-GlcNAcylation in mitochondria. Mitochondrial O-GlcNAcylation is promoted by activity-driven glucose consumption, which allows neurons to compensate for high energy expenditure based on fuel availability. To determine the proteins that are responsible for these adjustments, we mapped the mitochondrial O-GlcNAcome of neurons. Finally, we determine that neurons fail to meet activity-driven metabolic demand when O-GlcNAcylation dynamics are prevented. Our findings suggest that O-GlcNAcylation provides a fuel-dependent feedforward control mechanism in neurons to optimize mitochondrial performance based on neuronal activity. This mechanism thereby couples neuronal metabolism to mitochondrial bioenergetics and plays a key role in sustaining energy homeostasis.
Collapse
Affiliation(s)
- Seungyoon B Yu
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Haoming Wang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Richard G Sanchez
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Natasha M Carlson
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Khanh Nguyen
- Laboratory for Immunochemical Circuits, Center of Autoimmunity and Inflammation, and Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92093, USA
| | - Andrew Zhang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Zachary D Papich
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ahmed A Abushawish
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Zachary Whiddon
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Weronika Matysik
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jie Zhang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Thomas C Whisenant
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, La Jolla, CA 92093, USA
| | - John N Koberstein
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Melissa L Stewart
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samuel A Myers
- Laboratory for Immunochemical Circuits, Center of Autoimmunity and Inflammation, and Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92093, USA; Department of Pharmacology, Program in Immunology, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Gulcin Pekkurnaz
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
7
|
Alghusen IM, Carman MS, Wilkins HM, Strope TA, Gimore C, Fedosyuk H, Shawa J, Ephrame SJ, Denson AR, Wang X, Swerdlow RH, Slawson C. O-GlcNAc impacts mitophagy via the PINK1-dependent pathway. Front Aging Neurosci 2024; 16:1387931. [PMID: 39175808 PMCID: PMC11339348 DOI: 10.3389/fnagi.2024.1387931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024] Open
Abstract
Background The accumulation of dysfunctional mitochondria is an early feature of Alzheimer's disease (AD). The impaired turnover of damaged mitochondria increases reactive oxygen species production and lowers ATP generation, leading to cellular toxicity and neurodegeneration. Interestingly, AD exhibits a disruption in the global post-translational modification β-N-acetylglucosamine (O-GlcNAc). O-GlcNAc is a ubiquitous single sugar modification found in the nuclear, cytoplasmic, and mitochondrial proteins. Cells maintain a homeostatic level of O-GlcNAc by cycling the addition and removal of the sugar by O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA), respectively. Methods We used patient-derived induced pluripotent stem cells, a transgenic mouse model of AD, SH-SY5Y neuroblastoma cell lines to examine the effect of sustained O-GlcNAcase inhibition by Thiamet-G (TMG) or OGT deficiency on mitophagy using biochemical analyses. Results Here, we established an essential role for O-GlcNAc in regulating mitophagy (mitochondria-selective autophagy). Stimulating mitophagy using urolithin A (UA) decreases cellular O-GlcNAc and elevates mitochondrial O-GlcNAc. Sustained elevation in O-GlcNAcylation via pharmacologically inhibiting OGA using Thiamet-G (TMG) increases the mitochondrial level of mitophagy protein PTEN-induced kinase 1 (PINK1) and autophagy-related protein light chain 3 (LC3). Moreover, we detected O-GlcNAc on PINK1 and TMG increases its O-GlcNAcylation level. Conversely, decreasing cellular O-GlcNAcylation by knocking down OGT decreases both PINK1 protein expression and LC3 protein expression. Mitochondria isolated from CAMKII-OGT-KO mice also had decreased PINK1 and LC3. Moreover, human brain organoids treated with TMG showed significant elevation in LC3 compared to control. However, TMG-treated AD organoids showed no changes in LC3 expression. Conclusion Collectively, these data demonstrate that O-GlcNAc plays a crucial role in the activation and progression of mitophagy, and this activation is disrupted in AD.
Collapse
Affiliation(s)
- Ibtihal M. Alghusen
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Marisa S. Carman
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Heather M. Wilkins
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Taylor A. Strope
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Caleb Gimore
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Halyna Fedosyuk
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jad Shawa
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sophiya John Ephrame
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Aspin R. Denson
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Xiaowan Wang
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Russell H. Swerdlow
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Chad Slawson
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
8
|
Kim DY, Kim SM, Han IO. Chronic rapid eye movement sleep deprivation aggravates the pathogenesis of Alzheimer's disease by decreasing brain O-GlcNAc cycling in mice. J Neuroinflammation 2024; 21:180. [PMID: 39044290 PMCID: PMC11264383 DOI: 10.1186/s12974-024-03179-4] [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: 03/02/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
This study investigated the role of O-GlcNAc cycling in Alzheimer's disease-related changes in brain pathophysiology induced by chronic REM sleep deprivation (CSD) in mice. CSD increased amyloid beta (Aβ) and p-Tau accumulation and impaired learning and memory (L/M) function. CSD decreased dendritic length and spine density. CSD also increased the intensity of postsynaptic density protein-95 (PSD-95) staining. All of these Alzheimer's disease (AD) pathogenic changes were effectively reversed through glucosamine (GlcN) treatment by enhancing O-GlcNAcylation. Interestingly, the lelvel of O-GlcNAcylated-Tau (O-Tau) exhibited an opposite trend compared to p-Tau, as it was elevated by CSD and suppressed by GlcN treatment. CSD increased neuroinflammation, as indicated by elevated levels of glial fibrillary acidic protein and IBA-1-positive glial cells in the brain, which were suppressed by GlcN treatment. CSD promoted the phosphorylation of GSK3β and led to an upregulation in the expression of endoplasmic reticulum (ER) stress regulatory proteins and genes. These alterations were effectively suppressed by GlcN treatment. Minocycline not only suppressed neuroinflammation induced by CSD, but it also rescued the decrease in O-GlcNAc levels caused by CSD. Minocycline also reduced AD neuropathy without affecting CSD-induced ER stress. Notably, overexpressing O-GlcNAc transferase in the dentate gyrus region of the mouse brain rescued CSD-induced cognitive dysfunction, neuropathy, neuroinflammation, and ER stress responses. Collectively, our findings reveal that dysregulation of O-GlcNAc cycling underlies CSD-induced AD pathology and demonstrate that restoration of OGlcNAcylation protects against CSD-induced neurodegeneration.
Collapse
Affiliation(s)
- Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Sang-Min Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Korea.
| |
Collapse
|
9
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [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: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
10
|
Kang Y, Zhang Q, Xu S, Yu Y. The alteration and role of glycoconjugates in Alzheimer's disease. Front Aging Neurosci 2024; 16:1398641. [PMID: 38946780 PMCID: PMC11212478 DOI: 10.3389/fnagi.2024.1398641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by abnormal protein deposition. With an alarming 30 million people affected worldwide, AD poses a significant public health concern. While inhibiting key enzymes such as β-site amyloid precursor protein-cleaving enzyme 1 and γ-secretase or enhancing amyloid-β clearance, has been considered the reasonable strategy for AD treatment, their efficacy has been compromised by ineffectiveness. Furthermore, our understanding of AD pathogenesis remains incomplete. Normal aging is associated with a decline in glucose uptake in the brain, a process exacerbated in patients with AD, leading to significant impairment of a critical post-translational modification: glycosylation. Glycosylation, a finely regulated mechanism of intracellular secondary protein processing, plays a pivotal role in regulating essential functions such as synaptogenesis, neurogenesis, axon guidance, as well as learning and memory within the central nervous system. Advanced glycomic analysis has unveiled that abnormal glycosylation of key AD-related proteins closely correlates with the onset and progression of the disease. In this context, we aimed to delve into the intricate role and underlying mechanisms of glycosylation in the etiopathology and pathogenesis of AD. By highlighting the potential of targeting glycosylation as a promising and alternative therapeutic avenue for managing AD, we strive to contribute to the advancement of treatment strategies for this debilitating condition.
Collapse
Affiliation(s)
- Yue Kang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qian Zhang
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Silu Xu
- Department of Pharmacy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Yu
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
11
|
Murray M, Davidson L, Ferenbach AT, Lefeber D, van Aalten DMF. Neuroectoderm phenotypes in a human stem cell model of O-GlcNAc transferase associated with intellectual disability. Mol Genet Metab 2024; 142:108492. [PMID: 38759397 DOI: 10.1016/j.ymgme.2024.108492] [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: 02/07/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
Pathogenic variants in the O-GlcNAc transferase gene (OGT) have been associated with a congenital disorder of glycosylation (OGT-CDG), presenting with intellectual disability which may be of neuroectodermal origin. To test the hypothesis that pathology is linked to defects in differentiation during early embryogenesis, we developed an OGT-CDG induced pluripotent stem cell line together with isogenic control generated by CRISPR/Cas9 gene-editing. Although the OGT-CDG variant leads to a significant decrease in OGT and O-GlcNAcase protein levels, there were no changes in differentiation potential or stemness. However, differentiation into ectoderm resulted in significant differences in O-GlcNAc homeostasis. Further differentiation to neuronal stem cells revealed differences in morphology between patient and control lines, accompanied by disruption of the O-GlcNAc pathway. This suggests a critical role for O-GlcNAcylation in early neuroectoderm architecture, with robust compensatory mechanisms in the earliest stages of stem cell differentiation.
Collapse
Affiliation(s)
- Marta Murray
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Lindsay Davidson
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Andrew T Ferenbach
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, DK, Denmark
| | - Dirk Lefeber
- Department of Neurology, Department of Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, NL, the Netherlands
| | - Daan M F van Aalten
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK; Department of Molecular Biology and Genetics, Aarhus University, Aarhus, DK, Denmark.
| |
Collapse
|
12
|
Balana AT, Mahul-Mellier AL, Nguyen BA, Horvath M, Javed A, Hard ER, Jasiqi Y, Singh P, Afrin S, Pedretti R, Singh V, Lee VMY, Luk KC, Saelices L, Lashuel HA, Pratt MR. O-GlcNAc forces an α-synuclein amyloid strain with notably diminished seeding and pathology. Nat Chem Biol 2024; 20:646-655. [PMID: 38347213 PMCID: PMC11062923 DOI: 10.1038/s41589-024-01551-2] [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: 03/13/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024]
Abstract
Amyloid-forming proteins such α-synuclein and tau, which are implicated in Alzheimer's and Parkinson's disease, can form different fibril structures or strains with distinct toxic properties, seeding activities and pathology. Understanding the determinants contributing to the formation of different amyloid features could open new avenues for developing disease-specific diagnostics and therapies. Here we report that O-GlcNAc modification of α-synuclein monomers results in the formation of amyloid fibril with distinct core structure, as revealed by cryogenic electron microscopy, and diminished seeding activity in seeding-based neuronal and rodent models of Parkinson's disease. Although the mechanisms underpinning the seeding neutralization activity of the O-GlcNAc-modified fibrils remain unclear, our in vitro mechanistic studies indicate that heat shock proteins interactions with O-GlcNAc fibril inhibit their seeding activity, suggesting that the O-GlcNAc modification may alter the interactome of the α-synuclein fibrils in ways that lead to reduce seeding activity in vivo. Our results show that posttranslational modifications, such as O-GlcNAc modification, of α-synuclein are key determinants of α-synuclein amyloid strains and pathogenicity.
Collapse
Affiliation(s)
- Aaron T Balana
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Anne-Laure Mahul-Mellier
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Binh A Nguyen
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Mian Horvath
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Afraah Javed
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Eldon R Hard
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Yllza Jasiqi
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Preeti Singh
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shumaila Afrin
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rose Pedretti
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Virender Singh
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Virginia M-Y Lee
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelvin C Luk
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorena Saelices
- Center for Alzheimer's and Neurodegenerative Diseases, Department of Biophysics, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Matthew R Pratt
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA.
- Department Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Peng Y, Wang C, Ma W, Chen Q, Xu G, Kong Y, Ma L, Ding W, Zhang W. Deficiency of polypeptide N-acetylgalactosamine transferase 9 contributes to a risk for Parkinson's disease via mitochondrial dysfunctions. Int J Biol Macromol 2024; 263:130347. [PMID: 38401583 DOI: 10.1016/j.ijbiomac.2024.130347] [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: 09/14/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
Polypeptide N-acetylgalactosamine transferase 9 (GALNT9) catalyzes the initial step of mucin-type O-glycosylation via linking N-acetylgalactosamine (GalNAc) to serine/threonine in a protein. To unravel the association of GALNT9 with Parkinson's disease (PD), a progressive neurodegenerative disorder, GALNT9 levels were evaluated in the patients with PD and mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, and statistically analyzed based on the GEO datasets of GSE114918 and GSE216281. Glycoproteins with exposing GalNAc were purified using lectin affinity chromatography and identified by LC-MS/MS. The influence of GALNT9 on cells was evaluated via introducing a GALNT9-specific siRNA into SH-SY5Y cells. Consequently, GALNT9 deficiency was found to occur under PD conditions. GALNT9 silencing contributed to a causative factor in PD pathogenesis via reducing the levels of intracellular dopamine, tyrosine hydroxylase and soluble α-synuclein, and promoting α-synuclein aggregates. MS identification revealed 14 glycoproteins. 5 glycoproteins, including ACO2, ATP5B, CKB, CKMT1A, ALDOC, were associated with energy metabolism. GALNT9 silencing resulted in mitochondrial dysfunctions via increasing ROS accumulation, mitochondrial membrane depolarization, mPTPs opening, Ca2+ releasing and activation of the CytC-related apoptotic pathway. The dysfunctional mitochondria then triggered mitophagy, possibly intermediated by adenine nucleotide translocase 1. Our study suggests that GALNT9 is potentially developed into an auxiliary diagnostic index and therapeutic target of PD.
Collapse
Affiliation(s)
- Yuanwen Peng
- Department of Epidemiology, Dalian Medical University, Dalian 116044, China
| | - Cui Wang
- Department of Neurology, Dalian Municipal Central Hospital, Dalian 116033, China
| | - Wei Ma
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Qianhui Chen
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Guannan Xu
- Department of Epidemiology, Dalian Medical University, Dalian 116044, China
| | - Ying Kong
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Li Ma
- Department of Epidemiology, Dalian Medical University, Dalian 116044, China
| | - Wenyong Ding
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Wenli Zhang
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
14
|
Authier F, Ondruskova N, Ferenbach AT, McNeilly AD, van Aalten DMF. Neurodevelopmental defects in a mouse model of O-GlcNAc transferase intellectual disability. Dis Model Mech 2024; 17:dmm050671. [PMID: 38566589 PMCID: PMC11095632 DOI: 10.1242/dmm.050671] [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: 12/21/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
The addition of O-linked β-N-acetylglucosamine (O-GlcNAc) to proteins (referred to as O-GlcNAcylation) is a modification that is crucial for vertebrate development. O-GlcNAcylation is catalyzed by O-GlcNAc transferase (OGT) and reversed by O-GlcNAcase (OGA). Missense variants of OGT have recently been shown to segregate with an X-linked syndromic form of intellectual disability, OGT-linked congenital disorder of glycosylation (OGT-CDG). Although the existence of OGT-CDG suggests that O-GlcNAcylation is crucial for neurodevelopment and/or cognitive function, the underlying pathophysiologic mechanisms remain unknown. Here we report a mouse line that carries a catalytically impaired OGT-CDG variant. These mice show altered O-GlcNAc homeostasis with decreased global O-GlcNAcylation and reduced levels of OGT and OGA in the brain. Phenotypic characterization of the mice revealed lower body weight associated with reduced body fat mass, short stature and microcephaly. This mouse model will serve as an important tool to study genotype-phenotype correlations in OGT-CDG in vivo and for the development of possible treatment avenues for this disorder.
Collapse
Affiliation(s)
- Florence Authier
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Nina Ondruskova
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, 128 08 Praha 2, Czech Republic
| | - Andrew T. Ferenbach
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Alison D. McNeilly
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Daan M. F. van Aalten
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
15
|
Xue Q, Ji S, Xu H, Yu S. O-GlcNAcylation: a pro-survival response to acute stress in the cardiovascular and central nervous systems. Eur J Med Res 2024; 29:174. [PMID: 38491477 PMCID: PMC10943874 DOI: 10.1186/s40001-024-01773-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
O-GlcNAcylation is a unique monosaccharide modification that is ubiquitously present in numerous nucleoplasmic and mitochondrial proteins. The hexosamine biosynthesis pathway (HBP), which is a key branch of glycolysis, provides the unique sugar donor UDP-GlcNAc for the O-GlcNAc modification. Thus, HBP/O-GlcNAcylation can act as a nutrient sensor to perceive changes in nutrient levels and trigger O-GlcNAc modifications of functional proteins in cellular (patho-)physiology, thereby regulating diverse metabolic processes. An imbalance in O-GlcNAcylation has been shown to be a pathogenic contributor to dysfunction in metabolic diseases, including type 2 diabetes, cancer, and neurodegeneration. However, under acute stress conditions, protein O-GlcNAc modification exhibits rapid and transient upregulation, which is strongly correlated with stress tolerance and cell survival. In this context, we discuss the metabolic, pharmacological and genetic modulation of HBP/O-GlcNAc modification in the biological system, the beneficial role of O-GlcNAcylation in regulating stress tolerance for cardioprotection, and neuroprotection, which is a novel and rapidly growing field. Current evidence suggests that transient activation of the O-GlcNAc modification represents a potent pro-survival signalling pathway and may provide a promising strategy for stress-related disorder therapy.
Collapse
Affiliation(s)
- Qiu Xue
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China
- Department of General Surgery, Nantong Tumor Hospital, Nantong Fifth People's Hospital, Affiliated Tumor Hospital of Nantong University, 30 Tongyang North Road, Nantong, 226361, China
| | - Shengtao Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Hui Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, 399 Century Avenue, Nantong, 226001, China
| | - Shu Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China.
| |
Collapse
|
16
|
Pratt MR, Vocadlo DJ. Understanding and exploiting the roles of O-GlcNAc in neurodegenerative diseases. J Biol Chem 2023; 299:105411. [PMID: 37918804 PMCID: PMC10687168 DOI: 10.1016/j.jbc.2023.105411] [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: 08/16/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
O-GlcNAc is a common modification found on nuclear and cytoplasmic proteins. Determining the catalytic mechanism of the enzyme O-GlcNAcase (OGA), which removes O-GlcNAc from proteins, enabled the creation of potent and selective inhibitors of this regulatory enzyme. Such inhibitors have served as important tools in helping to uncover the cellular and organismal physiological roles of this modification. In addition, OGA inhibitors have been important for defining the augmentation of O-GlcNAc as a promising disease-modifying approach to combat several neurodegenerative diseases including both Alzheimer's disease and Parkinson's disease. These studies have led to development and optimization of OGA inhibitors for clinical application. These compounds have been shown to be well tolerated in early clinical studies and are steadily advancing into the clinic. Despite these advances, the mechanisms by which O-GlcNAc protects against these various types of neurodegeneration are a topic of continuing interest since improved insight may enable the creation of more targeted strategies to modulate O-GlcNAc for therapeutic benefit. Relevant pathways on which O-GlcNAc has been found to exert beneficial effects include autophagy, necroptosis, and processing of the amyloid precursor protein. More recently, the development and application of chemical methods enabling the synthesis of homogenous proteins have clarified the biochemical effects of O-GlcNAc on protein aggregation and uncovered new roles for O-GlcNAc in heat shock response. Here, we discuss the features of O-GlcNAc in neurodegenerative diseases, the application of inhibitors to identify the roles of this modification, and the biochemical effects of O-GlcNAc on proteins and pathways associated with neurodegeneration.
Collapse
Affiliation(s)
- Matthew R Pratt
- Department of Chemistry and Department of Biological Sciences, University of Southern California, Los Angeles, California, USA.
| | - 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.
| |
Collapse
|
17
|
Park J, Kim DY, Hwang GS, Han IO. Repeated sleep deprivation decreases the flux into hexosamine biosynthetic pathway/O-GlcNAc cycling and aggravates Alzheimer's disease neuropathology in adult zebrafish. J Neuroinflammation 2023; 20:257. [PMID: 37946213 PMCID: PMC10634120 DOI: 10.1186/s12974-023-02944-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
This study investigated chronic and repeated sleep deprivation (RSD)-induced neuronal changes in hexosamine biosynthetic pathway/O-linked N-acetylglucosamine (HBP/O-GlcNAc) cycling of glucose metabolism and further explored the role of altered O-GlcNAc cycling in promoting neurodegeneration using an adult zebrafish model. RSD-triggered degenerative changes in the brain led to impairment of memory, neuroinflammation and amyloid beta (Aβ) accumulation. Metabolite profiling of RSD zebrafish brain revealed a significant decrease in glucose, indicating a potential association between RSD-induced neurodegeneration and dysregulated glucose metabolism. While RSD had no impact on overall O-GlcNAcylation levels in the hippocampus region, changes were observed in two O-GlcNAcylation-regulating enzymes, specifically, a decrease in O-GlcNAc transferase (OGT) and an increase in O-GlcNAcase (OGA). Glucosamine (GlcN) treatment induced an increase in O-GlcNAcylation and recovery of the OGT level that was decreased in the RSD group. In addition, GlcN reversed cognitive impairment by RSD. GlcN reduced neuroinflammation and attenuated Aβ accumulation induced by RSD. Repeated treatment of zebrafish with diazo-5-oxo-l-norleucine (DON), an inhibitor of HBP metabolism, resulted in cognitive dysfunction, neuroinflammation and Aβ accumulation, similar to the effects of RSD. The pathological changes induced by DON were restored to normal upon treatment with GlcN. Both the SD and DON-treated groups exhibited a common decrease in glutamate and γ-aminobutyric acid compared to the control group. Overexpression of OGT in zebrafish brain rescued RSD-induced neuronal dysfunction and neurodegeneration. RSD induced a decrease in O-GlcNAcylation of amyloid precursor protein and increase in β-secretase activity, which were reversed by GlcN treatment. Based on the collective findings, we propose that dysregulation of HBP and O-GlcNAc cycling in brain plays a crucial role in RSD-mediated progression of neurodegeneration and Alzheimer's disease pathogenesis. Targeting of this pathway may, therefore, offer an effective regulatory approach for treatment of sleep-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Jiwon Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea
| | - Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, Korea
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea.
- Department of Physiology and Biophysics, College of Medicine, Inha University, 100 Inha Ro, Nam-Gu, Incheon, 22212, Korea.
| |
Collapse
|
18
|
Shoob S, Buchbinder N, Shinikamin O, Gold O, Baeloha H, Langberg T, Zarhin D, Shapira I, Braun G, Habib N, Slutsky I. Deep brain stimulation of thalamic nucleus reuniens promotes neuronal and cognitive resilience in an Alzheimer's disease mouse model. Nat Commun 2023; 14:7002. [PMID: 37919286 PMCID: PMC10622498 DOI: 10.1038/s41467-023-42721-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
The mechanisms that confer cognitive resilience to Alzheimer's Disease (AD) are not fully understood. Here, we describe a neural circuit mechanism underlying this resilience in a familial AD mouse model. In the prodromal disease stage, interictal epileptiform spikes (IESs) emerge during anesthesia in the CA1 and mPFC regions, leading to working memory disruptions. These IESs are driven by inputs from the thalamic nucleus reuniens (nRE). Indeed, tonic deep brain stimulation of the nRE (tDBS-nRE) effectively suppresses IESs and restores firing rate homeostasis under anesthesia, preventing further impairments in nRE-CA1 synaptic facilitation and working memory. Notably, applying tDBS-nRE during the prodromal phase in young APP/PS1 mice mitigates age-dependent memory decline. The IES rate during anesthesia in young APP/PS1 mice correlates with later working memory impairments. These findings highlight the nRE as a central hub of functional resilience and underscore the clinical promise of DBS in conferring resilience to AD pathology by restoring circuit-level homeostasis.
Collapse
Affiliation(s)
- Shiri Shoob
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Nadav Buchbinder
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Ortal Shinikamin
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Or Gold
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Halit Baeloha
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Tomer Langberg
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Daniel Zarhin
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Gabriella Braun
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Naomi Habib
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
19
|
Kim DY, Park J, Han IO. Hexosamine biosynthetic pathway and O-GlcNAc cycling of glucose metabolism in brain function and disease. Am J Physiol Cell Physiol 2023; 325:C981-C998. [PMID: 37602414 DOI: 10.1152/ajpcell.00191.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023]
Abstract
Impaired brain glucose metabolism is considered a hallmark of brain dysfunction and neurodegeneration. Disruption of the hexosamine biosynthetic pathway (HBP) and subsequent O-linked N-acetylglucosamine (O-GlcNAc) cycling has been identified as an emerging link between altered glucose metabolism and defects in the brain. Myriads of cytosolic and nuclear proteins in the nervous system are modified at serine or threonine residues with a single N-acetylglucosamine (O-GlcNAc) molecule by O-GlcNAc transferase (OGT), which can be removed by β-N-acetylglucosaminidase (O-GlcNAcase, OGA). Homeostatic regulation of O-GlcNAc cycling is important for the maintenance of normal brain activity. Although significant evidence linking dysregulated HBP metabolism and aberrant O-GlcNAc cycling to induction or progression of neuronal diseases has been obtained, the issue of whether altered O-GlcNAcylation is causal in brain pathogenesis remains uncertain. Elucidation of the specific functions and regulatory mechanisms of individual O-GlcNAcylated neuronal proteins in both normal and diseased states may facilitate the identification of novel therapeutic targets for various neuronal disorders. The information presented in this review highlights the importance of HBP/O-GlcNAcylation in the neuronal system and summarizes the roles and potential mechanisms of O-GlcNAcylated neuronal proteins in maintaining normal brain function and initiation and progression of neurological diseases.
Collapse
Affiliation(s)
- Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Jiwon Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| |
Collapse
|
20
|
Wang H, Vant J, Wu Y, Sanchez R, Micou ML, Zhang A, Luczak V, Yu SB, Jabbo M, Yoon S, Abushawish AA, Ghassemian M, Griffis E, Hammarlund M, Singharoy A, Pekkurnaz G. Functional Organization of Glycolytic Metabolon on Mitochondria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554955. [PMID: 37662343 PMCID: PMC10473731 DOI: 10.1101/2023.08.26.554955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Glucose, the primary cellular energy source, is metabolized through glycolysis initiated by the rate-limiting enzyme Hexokinase (HK). In energy-demanding tissues like the brain, HK1 is the dominant isoform, primarily localized on mitochondria, crucial for efficient glycolysis-oxidative phosphorylation coupling and optimal energy generation. This study unveils a unique mechanism regulating HK1 activity, glycolysis, and the dynamics of mitochondrial coupling, mediated by the metabolic sensor enzyme O-GlcNAc transferase (OGT). OGT catalyzes reversible O-GlcNAcylation, a post-translational modification, influenced by glucose flux. Elevated OGT activity induces dynamic O-GlcNAcylation of HK1's regulatory domain, subsequently promoting the assembly of the glycolytic metabolon on the outer mitochondrial membrane. This modification enhances HK1's mitochondrial association, orchestrating glycolytic and mitochondrial ATP production. Mutations in HK1's O-GlcNAcylation site reduce ATP generation, affecting synaptic functions in neurons. The study uncovers a novel pathway that bridges neuronal metabolism and mitochondrial function via OGT and the formation of the glycolytic metabolon, offering new prospects for tackling metabolic and neurological disorders.
Collapse
|
21
|
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
|
22
|
Ephrame SJ, Cork GK, Marshall V, Johnston MA, Shawa J, Alghusen I, Qiang A, Denson AR, Carman MS, Fedosyuk H, Swerdlow RH, Slawson C. O-GlcNAcylation regulates extracellular signal-regulated kinase (ERK) activation in Alzheimer's disease. Front Aging Neurosci 2023; 15:1155630. [PMID: 37469955 PMCID: PMC10352608 DOI: 10.3389/fnagi.2023.1155630] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/13/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction Aberrant activation of Extracellular Signal-Regulated Kinase (ERK) signaling is associated with Alzheimer's disease (AD) pathogenesis. For example, enhanced ERK signal activation mediated by Apolipoprotein E4 (APOE4), which is a critical genetic risk factor for AD, increases the transcription of amyloid precursor protein (APP). We hypothesize that O-linked N-acetylglucosamine (O-GlcNAc) regulates the phosphorylation and activation of ERK. O-GlcNAc is a single sugar post-translational modification that dynamically cycles on and off proteins in response to nutrient changes by the action of the enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. However, O-GlcNAc quickly returns to a baseline level after stimulus removal (called O-GlcNAc homeostasis). Methods We did a serum reactivation time-course followed by western blot in SH-SY5Y neuroblastoma cells after long-term O-GlcNAcase (OGA) inhibition by Thiamet-G (TMG) treatment, O-GlcNAc transferase (OGT) knock-down (KD) and OGA KD. Brain tissues of C57BL6/J mice and 5XFAD Alzheimer's disease mice intra-peritoneally injected with TMG for 1 month and C57BL6/J mice intra-peritoneally injected with TMG for 6 months were also used for western blot. Results We found that ERK1/2 phosphorylation at Thr 202/Tyr204 and Thr183/Tyr185 (p-ERK) are amplified and hence ERK1/2 are activated after long-term OGA inhibition in SH-SY5Y cells. In addition to pharmacological treatment, genetic disruption of O-GlcNAc by OGT KD and OGA KD also increased p-ERK in SH-SY5Y cells suggesting O-GlcNAc homeostasis controls ERK signaling. To determine how O-GlcNAc regulates p-ERK, we probed the expression of phosphorylated mitogen-activated protein kinase-kinase (p-MEK) which phosphorylates and activates ERK and Dual specificity phosphatase-4 (DUSP4) which dephosphorylates and inactivates ERK in SH-SY5Y cells. p-MEK increases in TMG treated and OGT KD cells whereas total DUSP4 decreases in OGT KD and OGA KD cells with serum reactivation time course. Next, we probed the role of OGA inhibition in regulating ERK activation using mice brain-tissue samples. Interestingly, 6-month intra-peritoneal TMG injection in C57BL/6J mice showed an increase in amplitude of p-ERK and APP protein levels, indicating long-term OGA inhibition potentially contributes to AD progression. Furthermore, 1-month TMG injection was sufficient to increase the amplitude of p-ERK in 5XFAD AD mice brains suggesting AD phenotype contributes to the acceleration of ERK activation mediated by OGA inhibition. Conclusion Together, these results indicate that disruptions to O-GlcNAc homeostasis amplify ERK signal activation in AD.
Collapse
Affiliation(s)
- Sophiya John Ephrame
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Gentry K. Cork
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Victoria Marshall
- School of Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Margaret A. Johnston
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jenna Shawa
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ibtihal Alghusen
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Amy Qiang
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Aspin R. Denson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Marisa S. Carman
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Halyna Fedosyuk
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Russell H. Swerdlow
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Chad Slawson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
23
|
Salman Sajid M, Saleem S, Jabeen F, Waqas Ishaq M, Najam-Ul-Haq M, Ressom HW. Mapping the low abundant plasma glycoproteome using Ranachrome-5 immobilized magnetic terpolymer as improved HILIC sorbent. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1227:123846. [PMID: 37567067 PMCID: PMC10528939 DOI: 10.1016/j.jchromb.2023.123846] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
HILIC (hydrophilic interaction liquid chromatography) materials enrich glycopeptides. The non-specific interactions because of support material and inadequate hydrophilicity render loss of less abundant glycopeptides in SPE-based enrichments. In this work, magnetic terpolymer (Fe3O4@MAA/DVB/1,2-Epoxy-5-hexene) is functionalized with Ranachrome-5 to generate enhanced hydrophilicity. Amine, carboxylic, and amide groups of ranachrome-5 provide zwitterionic chemistry. Material's magnetic core contributes to ease of operation while higher surface area 97.0711 m2 g-1 immobilizes better quantities of Ranachrome-5. Homogeneous morphology, nano-size, and super hydrophilicity enhance enrichment. Ranachrome-5 functionalized polymeric core-shell beads enrich 25, 18 and 16 N-linked glycopeptides via SPE strategy from tryptic digests of model glycoproteins i.e., immunoglobulin G (IgG), horseradish peroxidase (HRP) and chicken avidin, respectively. Zwitterionic chemistry of ranachrome-5 helps in achieving higher selectivity (1:250, HRP / Bovine Serum Albumin), and lower detection limit (100 attomole, HRP digest) with complete glycosylation profile of each standard digest. High binding capacity (137.1 mg/g) and reuse of affinity material up to seven cycles reduce the cost and amount of affinity material for complex sample analysis. A recovery of 91.76% and relative standard deviation (RSD) values less than 1 define the application of HILIC beads for complex samples like plasma. 508 N-linked intact low abundant glycopeptides corresponding to 50 glycoproteins are identified from depleted human plasma samples via nano-Liquid Chromatography-Tandem Mass Spectrometry (nLC-MS/MS). Using Single Nucleotide Variances (BioMuta) for low abundant plasma glycoproteins, the potential association of proteins to four cancers, i.e., breast, lung, uterine, and melanoma is evaluated. Via the bottom-up approach, HILIC beads can analyze clinically important low-abundant glycoproteins.
Collapse
Affiliation(s)
- Muhammad Salman Sajid
- Department of Oncology, Genomics and Epigenomics Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shafaq Saleem
- Department of Oncology, Genomics and Epigenomics Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Fahmida Jabeen
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan
| | | | - Muhammad Najam-Ul-Haq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Habtom W Ressom
- Department of Oncology, Genomics and Epigenomics Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
24
|
Dong X, Shu L, Zhang J, Yang X, Cheng X, Zhao X, Qu W, Zhu Q, Shou Y, Peng G, Sun B, Yi W, Shu Q, Li X. Ogt-mediated O-GlcNAcylation inhibits astrocytes activation through modulating NF-κB signaling pathway. J Neuroinflammation 2023; 20:146. [PMID: 37349834 DOI: 10.1186/s12974-023-02824-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 06/05/2023] [Indexed: 06/24/2023] Open
Abstract
Previous studies have shown that Ogt-mediated O-GlcNAcylation is essential for neuronal development and function. However, the function of O-GlcNAc transferase (Ogt) and O-GlcNAcylation in astrocytes remains largely unknown. Here we show that Ogt deficiency induces inflammatory activation of astrocytes in vivo and in vitro, and impairs cognitive function of mice. The restoration of O-GlcNAcylation via GlcNAc supplementation inhibits the activation of astrocytes, inflammation and improves the impaired cognitive function of Ogt deficient mice. Mechanistically, Ogt interacts with NF-κB p65 and catalyzes the O-GlcNAcylation of NF-κB p65 in astrocytes. Ogt deficiency induces the activation of NF-κB signaling pathway by promoting Gsk3β binding. Moreover, Ogt depletion induces the activation of astrocytes derived from human induced pluripotent stem cells. The restoration of O-GlcNAcylation inhibits the activation of astrocytes, inflammation and reduces Aβ plaque of AD mice in vitro and in vivo. Collectively, our study reveals a critical function of Ogt-mediated O-GlcNAcylation in astrocytes through regulating NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaoxue Dong
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Liqi Shu
- Department of Neurology, The Warren Alpert Medical School of Brown University, Providence, RI, 02908, USA
| | - Jinyu Zhang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xu Yang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xuejun Cheng
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Xingsen Zhao
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Wenzheng Qu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Qiang Zhu
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yikai Shou
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Guoping Peng
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Binggui Sun
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Wen Yi
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Qiang Shu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
| | - Xuekun Li
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, 310052, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029, China.
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou, 310029, China.
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
25
|
Zhang J, Wei K, Qu W, Wang M, Zhu Q, Dong X, Huang X, Yi W, Xu S, Li X. Ogt Deficiency Induces Abnormal Cerebellar Function and Behavioral Deficits of Adult Mice through Modulating RhoA/ROCK Signaling. J Neurosci 2023; 43:4559-4579. [PMID: 37225434 PMCID: PMC10286951 DOI: 10.1523/jneurosci.1962-22.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
Previous studies have shown the essential roles of O-GlcNAc transferase (Ogt) and O-GlcNAcylation in neuronal development, function and neurologic diseases. However, the function of Ogt and O-GlcNAcylation in the adult cerebellum has not been well elucidated. Here, we have found that cerebellum has the highest level of O-GlcNAcylation relative to cortex and hippocampus of adult male mice. Specific deletion of Ogt in granule neuron precursors (GNPs) induces abnormal morphology and decreased size of the cerebellum in adult male Ogt deficient [conditional knock-out (cKO)] mice. Adult male cKO mice show the reduced density and aberrant distribution of cerebellar granule cells (CGCs), the disrupted arrangement of Bergman glia (BG) and Purkinje cells. In addition, adult male cKO mice exhibit aberrant synaptic connection, impaired motor coordination, and learning and memory abilities. Mechanistically, we have identified G-protein subunit α12 (Gα12) is modified by Ogt-mediated O-GlcNAcylation. O-GlcNAcylation of Gα12 facilitates its binding to Rho guanine nucleotide exchange factor 12 (Arhgef12) and consequently activates RhoA/ROCK signaling. RhoA/ROCK pathway activator LPA can rescue the developmental deficits of Ogt deficient CGCs. Therefore, our study has revealed the critical function and related mechanisms of Ogt and O-GlcNAcylation in the cerebellum of adult male mice.SIGNIFICANCE STATEMENT Cerebellar function are regulated by diverse mechanisms. To unveil novel mechanisms is critical for understanding the cerebellar function and the clinical therapy of cerebellum-related diseases. In the present study, we have shown that O-GlcNAc transferase gene (Ogt) deletion induces abnormal cerebellar morphology, synaptic connection, and behavioral deficits of adult male mice. Mechanistically, Ogt catalyzes O-GlcNAcylation of Gα12, which promotes the binding to Arhgef12, and regulates RhoA/ROCK signaling pathway. Our study has uncovered the important roles of Ogt and O-GlcNAcylation in regulating cerebellar function and cerebellum-related behavior. Our results suggest that Ogt and O-GlcNAcylation could be potential targets for some cerebellum-related diseases.
Collapse
Affiliation(s)
- Jinyu Zhang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Kaiyan Wei
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Wenzheng Qu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Mengxuan Wang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Qiang Zhu
- MOE Key Laboratory of Biosystems Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310002, China
| | - Xiaoxue Dong
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Xiaoli Huang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Wen Yi
- MOE Key Laboratory of Biosystems Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310002, China
| | - Shunliang Xu
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Xuekun Li
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou 310052, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310029, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou 310052, China
- Binjiang Institute of Zhejiang University, Hangzhou 310053, China
- Zhejiang University Cancer Center, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
26
|
Victor AK, Hedgecock T, Donaldson M, Johnson D, Rand CM, Weese-Mayer DE, Reiter LT. Analysis and comparisons of gene expression changes in patient- derived neurons from ROHHAD, CCHS, and PWS. Front Pediatr 2023; 11:1090084. [PMID: 37234859 PMCID: PMC10206321 DOI: 10.3389/fped.2023.1090084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Background Rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation (ROHHAD) syndrome is an ultra-rare neurocristopathy with no known genetic or environmental etiology. Rapid-onset obesity over a 3-12 month period with onset between ages 1.5-7 years of age is followed by an unfolding constellation of symptoms including severe hypoventilation that can lead to cardiorespiratory arrest in previously healthy children if not identified early and intervention provided. Congenital Central Hypoventilation syndrome (CCHS) and Prader-Willi syndrome (PWS) have overlapping clinical features with ROHHAD and known genetic etiologies. Here we compare patient neurons from three pediatric syndromes (ROHHAD, CCHS, and PWS) and neurotypical control subjects to identify molecular overlap that may explain the clinical similarities. Methods Dental pulp stem cells (DPSC) from neurotypical control, ROHHAD, and CCHS subjects were differentiated into neuronal cultures for RNA sequencing (RNAseq). Differential expression analysis identified transcripts variably regulated in ROHHAD and CCHS vs. neurotypical control neurons. In addition, we used previously published PWS transcript data to compare both groups to PWS patient-derived DPSC neurons. Enrichment analysis was performed on RNAseq data and downstream protein expression analysis was performed using immunoblotting. Results We identified three transcripts differentially regulated in all three syndromes vs. neurotypical control subjects. Gene ontology analysis on the ROHHAD dataset revealed enrichments in several molecular pathways that may contribute to disease pathology. Importantly, we found 58 transcripts differentially expressed in both ROHHAD and CCHS patient neurons vs. control neurons. Finally, we validated transcript level changes in expression of ADORA2A, a gene encoding for an adenosine receptor, at the protein level in CCHS neurons and found variable, although significant, changes in ROHHAD neurons. Conclusions The molecular overlap between CCHS and ROHHAD neurons suggests that the clinical phenotypes in these syndromes likely arise from or affect similar transcriptional pathways. Further, gene ontology analysis identified enrichments in ATPase transmembrane transporters, acetylglucosaminyltransferases, and phagocytic vesicle membrane proteins that may contribute to the ROHHAD phenotype. Finally, our data imply that the rapid-onset obesity seen in both ROHHAD and PWS likely arise from different molecular mechanisms. The data presented here describes important preliminary findings that warrant further validation.
Collapse
Affiliation(s)
- A. Kaitlyn Victor
- IPBS Program, Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Tayler Hedgecock
- IPBS Program, Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Martin Donaldson
- Department of Pediatric Dentistry and Community Oral Health, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Daniel Johnson
- Molecular Bioinformatics Core, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Casey M. Rand
- Department of Pediatrics, Division of Autonomic Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago and Stanley Manne Children’s Research Institute, Chicago, IL, United States
| | - Debra E. Weese-Mayer
- Department of Pediatrics, Division of Autonomic Medicine, Ann & Robert H. Lurie Children’s Hospital of Chicago and Stanley Manne Children’s Research Institute, Chicago, IL, United States
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lawrence T. Reiter
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
27
|
Pradeep P, Kang H, Lee B. Glycosylation and behavioral symptoms in neurological disorders. Transl Psychiatry 2023; 13:154. [PMID: 37156804 PMCID: PMC10167254 DOI: 10.1038/s41398-023-02446-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023] Open
Abstract
Glycosylation, the addition of glycans or carbohydrates to proteins, lipids, or other glycans, is a complex post-translational modification that plays a crucial role in cellular function. It is estimated that at least half of all mammalian proteins undergo glycosylation, underscoring its importance in the functioning of cells. This is reflected in the fact that a significant portion of the human genome, around 2%, is devoted to encoding enzymes involved in glycosylation. Changes in glycosylation have been linked to various neurological disorders, including Alzheimer's disease, Parkinson's disease, autism spectrum disorder, and schizophrenia. Despite its widespread occurrence, the role of glycosylation in the central nervous system remains largely unknown, particularly with regard to its impact on behavioral abnormalities in brain diseases. This review focuses on examining the role of three types of glycosylation: N-glycosylation, O-glycosylation, and O-GlcNAcylation, in the manifestation of behavioral and neurological symptoms in neurodevelopmental, neurodegenerative, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Prajitha Pradeep
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea
- IBS School, University of Science and Technology (UST), Daejeon, 34113, South Korea
| | - Hyeyeon Kang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea
| | - Boyoung Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea.
- IBS School, University of Science and Technology (UST), Daejeon, 34113, South Korea.
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea.
| |
Collapse
|
28
|
Li A, Fan J, Jia Y, Tang X, Chen J, Shen C. Phenotype and metabolism alterations in PCB-degrading Rhodococcus biphenylivorans TG9 T under acid stress. J Environ Sci (China) 2023; 127:441-452. [PMID: 36522076 DOI: 10.1016/j.jes.2022.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 06/17/2023]
Abstract
Environmental acidification impairs microorganism diversity and their functions on substance transformation. Rhodococcus is a ubiquitously distributed genus for contaminant detoxification in the environment, and it can also adapt a certain range of pH. This work interpreted the acid responses from both phenotype and metabolism in strain Rhodococcus biphenylivorans TG9T (TG9) induced at pH 3. The phenotype alterations were described with the number of culturable and viable cells, intracellular ATP concentrations, cell shape and entocyte, degradation efficiency of polychlorinated biphenyl (PCB) 31 and biphenyl. The number of culturable cells maintained rather stable within the first 10 days, even though the other phenotypes had noticeable alterations, indicating that TG9 possesses certain capacities to survive under acid stress. The metabolism responses were interpreted based on transcription analyses with four treatments including log phase (LP), acid-induced (PER), early recovery after removing acid (RE) and later recovery (REL). With the overview on the expression regulations among the 4 treatments, the RE sample presented more upregulated and less downregulated genes, suggesting that its metabolism was somehow more active after recovering from acid stress. In addition, the response mechanism was interpreted on 10 individual metabolism pathways mainly covering protein modification, antioxidation, antipermeability, H+ consumption, neutralization and extrusion. Furthermore, the transcription variations were verified with RT-qPCR on 8 genes with 24-hr, 48-hr and 72-hr acid treatment. Taken together, TG9 possesses comprehensive metabolism strategies defending against acid stress. Consequently, a model was built to provide an integrate insight to understand the acid resistance/tolerance metabolisms in microorganisms.
Collapse
Affiliation(s)
- Aili Li
- Department of Environmental Engineering, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahui Fan
- Department of Environmental Engineering, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yangyang Jia
- Department of Environmental Engineering, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xianjin Tang
- MOE Key Lab of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jingwen Chen
- Department of Environmental Engineering, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chaofeng Shen
- Department of Environmental Engineering, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
29
|
Paton SE, Menard C. Glutamate shall not pass: a mechanistic role for astrocytic O-GlcNAc transferase in stress and depression. J Clin Invest 2023; 133:168662. [PMID: 37009895 PMCID: PMC10065068 DOI: 10.1172/jci168662] [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: 04/04/2023] Open
Abstract
Major depressive disorder, characterized by aberrant glutamatergic signaling in the prefrontal cortex (PFC), is a leading cause of disability worldwide. Depression is highly comorbid with metabolic disorders, but a mechanistic link is elusive. In this issue of the JCI, Fan and coauthors report that elevated posttranslational modification with the glucose metabolite N-acetylglucosamine (GlcNAc) by O-GlcNAc transferase (OGT) contributed to stress-induced establishment of depression-like behaviors in mice. This effect was specific to medial PFC (mPFC) astrocytes, with glutamate transporter-1 (GLT-1) identified as an OGT target. Specifically, O-GlcNAcylation of GLT-1 resulted in diminished glutamate clearance from excitatory synapses. Further, astrocytic OGT knockdown restored stress-induced deficits in glutamatergic signaling, promoting resilience. These findings provide a mechanistic link between metabolism and depression and have relevance for antidepressant targets.
Collapse
|
30
|
Balana AT, Mahul-Mellier AL, Nguyen BA, Horvath M, Javed A, Hard ER, Jasiqi Y, Singh P, Afrin S, Pedretti R, Singh V, Lee VMY, Luk KC, Saelices L, Lashuel HA, Pratt MR. O-GlcNAc modification forces the formation of an α-Synuclein amyloid-strain with notably diminished seeding activity and pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531573. [PMID: 36945566 PMCID: PMC10028859 DOI: 10.1101/2023.03.07.531573] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The process of amyloid fibril formation remains one of the primary targets for developing diagnostics and treatments for several neurodegenerative diseases (NDDs). Amyloid-forming proteins such α-Synuclein and Tau, which are implicated in the pathogenesis of Alzheimer's and Parkinson's disease, can form different types of fibril structure, or strains, that exhibit distinct structures, toxic properties, seeding activities, and pathology spreading patterns in the brain. Therefore, understanding the molecular and structural determinants contributing to the formation of different amyloid strains or their distinct features could open new avenues for developing disease-specific diagnostics and therapies. In this work, we report that O-GlcNAc modification of α-Synuclein monomers results in the formation of amyloid fibril with distinct core structure, as revealed by Cryo-EM, and diminished seeding activity in seeding-based neuronal and rodent models of Parkinson's disease. Although the mechanisms underpinning the seeding neutralization activity of the O-GlcNAc modified fibrils remain unclear, our in vitro mechanistic studies indicate that heat shock proteins interactions with O-GlcNAc fibril inhibit their seeding activity, suggesting that the O-GlcNAc modification may alter the interactome of the α-Synuclein fibrils in ways that lead to reduce seeding activity in vivo. Our results show that post-translational modifications, such as O-GlcNAc modification, of α-Synuclein are key determinants of α-Synuclein amyloid strains and pathogenicity. These findings have significant implications for how we investigate and target amyloids in the brain and could possibly explain the lack of correlation between amyloid burden and neurodegeneration or cognitive decline in some subtypes of NDDs.
Collapse
Affiliation(s)
- Aaron T. Balana
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, United States
| | - Anne-Laure Mahul-Mellier
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland CH-1015
| | - Binh A Nguyen
- Center for Alzheimer’s and Neurodegenerative Disease, Department of Biophysics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX-75390
| | - Mian Horvath
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Afraah Javed
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, United States
| | - Eldon R. Hard
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, United States
| | - Yllza Jasiqi
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland CH-1015
| | - Preeti Singh
- Center for Alzheimer’s and Neurodegenerative Disease, Department of Biophysics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX-75390
| | - Shumaila Afrin
- Center for Alzheimer’s and Neurodegenerative Disease, Department of Biophysics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX-75390
| | - Rose Pedretti
- Center for Alzheimer’s and Neurodegenerative Disease, Department of Biophysics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX-75390
| | - Virender Singh
- Center for Alzheimer’s and Neurodegenerative Disease, Department of Biophysics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX-75390
| | - Virginia M.-Y. Lee
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelvin C. Luk
- The Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lorena Saelices
- Center for Alzheimer’s and Neurodegenerative Disease, Department of Biophysics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX-75390
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland CH-1015
| | - Matthew R. Pratt
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, United States
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, United States
| |
Collapse
|
31
|
Bogetofte H, Ryan BJ, Jensen P, Schmidt SI, Vergoossen DLE, Barnkob MB, Kiani LN, Chughtai U, Heon-Roberts R, Caiazza MC, McGuinness W, Márquez-Gómez R, Vowles J, Bunn FS, Brandes J, Kilfeather P, Connor JP, Fernandes HJR, Caffrey TM, Meyer M, Cowley SA, Larsen MR, Wade-Martins R. Post-translational proteomics platform identifies neurite outgrowth impairments in Parkinson's disease GBA-N370S dopamine neurons. Cell Rep 2023; 42:112180. [PMID: 36870058 DOI: 10.1016/j.celrep.2023.112180] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/04/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Variants at the GBA locus, encoding glucocerebrosidase, are the strongest common genetic risk factor for Parkinson's disease (PD). To understand GBA-related disease mechanisms, we use a multi-part-enrichment proteomics and post-translational modification (PTM) workflow, identifying large numbers of dysregulated proteins and PTMs in heterozygous GBA-N370S PD patient induced pluripotent stem cell (iPSC) dopamine neurons. Alterations in glycosylation status show disturbances in the autophagy-lysosomal pathway, which concur with upstream perturbations in mammalian target of rapamycin (mTOR) activation in GBA-PD neurons. Several native and modified proteins encoded by PD-associated genes are dysregulated in GBA-PD neurons. Integrated pathway analysis reveals impaired neuritogenesis in GBA-PD neurons and identify tau as a key pathway mediator. Functional assays confirm neurite outgrowth deficits and identify impaired mitochondrial movement in GBA-PD neurons. Furthermore, pharmacological rescue of glucocerebrosidase activity in GBA-PD neurons improves the neurite outgrowth deficit. Overall, this study demonstrates the potential of PTMomics to elucidate neurodegeneration-associated pathways and potential drug targets in complex disease models.
Collapse
Affiliation(s)
- Helle Bogetofte
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Brent J Ryan
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK.
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Sissel I Schmidt
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Dana L E Vergoossen
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Mike B Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Department of Clinical Immunology, Odense University Hospital, University of Southern Denmark, 5000 Odense C, Denmark
| | - Lisa N Kiani
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Uroosa Chughtai
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Rachel Heon-Roberts
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Maria Claudia Caiazza
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - William McGuinness
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Ricardo Márquez-Gómez
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Fiona S Bunn
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Janine Brandes
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Peter Kilfeather
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Jack P Connor
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Hugo J R Fernandes
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | - Tara M Caffrey
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark; Department of Neurology, Odense University Hospital, 5000 Odense C, Denmark
| | - Sally A Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
32
|
Yu SB, Sanchez RG, Papich ZD, Whisenant TC, Ghassemian M, Koberstein JN, Stewart ML, Pekkurnaz G. Neuronal activity-driven O-GlcNAcylation promotes mitochondrial plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523512. [PMID: 36711626 PMCID: PMC9882081 DOI: 10.1101/2023.01.11.523512] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Neuronal activity is an energy-intensive process that is largely sustained by instantaneous fuel utilization and ATP synthesis. However, how neurons couple ATP synthesis rate to fuel availability is largely unknown. Here, we demonstrate that the metabolic sensor enzyme O-GlcNAc transferase regulates neuronal activity-driven mitochondrial bioenergetics. We show that neuronal activity upregulates O-GlcNAcylation mainly in mitochondria. Mitochondrial O-GlcNAcylation is promoted by activity-driven fuel consumption, which allows neurons to compensate for high energy expenditure based on fuel availability. To determine the proteins that are responsible for these adjustments, we mapped the mitochondrial O-GlcNAcome of neurons. Finally, we determine that neurons fail to meet activity-driven metabolic demand when O-GlcNAcylation dynamics are prevented. Our findings suggest that O-GlcNAcylation provides a fuel-dependent feedforward control mechanism in neurons to optimize mitochondrial performance based on neuronal activity. This mechanism thereby couples neuronal metabolism to mitochondrial bioenergetics and plays a key role in sustaining energy homeostasis.
Collapse
|
33
|
Huang Y, Wang J, Liu F, Wang C, Xiao Z, Zhou W. Liuwei Dihuang formula ameliorates chronic stress-induced emotional and cognitive impairments in mice by elevating hippocampal O-GlcNAc modification. Front Neurosci 2023; 17:1134176. [PMID: 37152609 PMCID: PMC10157057 DOI: 10.3389/fnins.2023.1134176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
A substantial body of evidence has indicated that intracerebral O-linked N-acetyl-β-D-glucosamine (O-GlcNAc), a generalized post-translational modification, was emerging as an effective regulator of stress-induced emotional and cognitive impairments. Our previous studies showed that the Liuwei Dihuang formula (LW) significantly improved the emotional and cognitive dysfunctions in various types of stress mouse models. In the current study, we sought to determine the effects of LW on intracerebral O-GlcNAc levels in chronic unpredictable mild stress (CUMS) mice. The dynamic behavioral tests showed that anxiety- and depression-like behaviors and object recognition memory of CUMS mice were improved in a dose-dependent manner after LW treatment. Moreover, linear discriminate analysis (LEfSe) of genera abundance revealed a significant difference in microbiome among the study groups. LW showed a great impact on the relative abundance of these gut microbiota in CUMS mice and reinstated them to control mouse levels. We found that LW potentially altered the Uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) biosynthesis process, and the abundance of O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT) in CUMS mice, which was inferred using PICRUSt analysis. We further verified advantageous changes in hippocampal O-GlcNAc modification of CUMS mice following LW administration, as well as changes in the levels of OGA and OGT. In summary, LW intervention increased the levels of hippocampal O-GlcNAc modification and ameliorated the emotional and cognitive impairments induced by chronic stress in CUMS mice. LW therefore could be considered a potential prophylactic and therapeutic agent for chronic stress.
Collapse
Affiliation(s)
- Yan Huang
- Nanjing University of Chinese Medicine, Nanjing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Jianhui Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Feng Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Chenran Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- *Correspondence: Zhiyong Xiao,
| | - Wenxia Zhou
- Nanjing University of Chinese Medicine, Nanjing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
- Wenxia Zhou,
| |
Collapse
|
34
|
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
|
35
|
Fahie KMM, Papanicolaou KN, Zachara NE. Integration of O-GlcNAc into Stress Response Pathways. Cells 2022; 11:3509. [PMID: 36359905 PMCID: PMC9654274 DOI: 10.3390/cells11213509] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
The modification of nuclear, mitochondrial, and cytosolic proteins by O-linked βN-acetylglucosamine (O-GlcNAc) has emerged as a dynamic and essential post-translational modification of mammalian proteins. O-GlcNAc is cycled on and off over 5000 proteins in response to diverse stimuli impacting protein function and, in turn, epigenetics and transcription, translation and proteostasis, metabolism, cell structure, and signal transduction. Environmental and physiological injury lead to complex changes in O-GlcNAcylation that impact cell and tissue survival in models of heat shock, osmotic stress, oxidative stress, and hypoxia/reoxygenation injury, as well as ischemic reperfusion injury. Numerous mechanisms that appear to underpin O-GlcNAc-mediated survival include changes in chaperone levels, impacts on the unfolded protein response and integrated stress response, improvements in mitochondrial function, and reduced protein aggregation. Here, we discuss the points at which O-GlcNAc is integrated into the cellular stress response, focusing on the roles it plays in the cardiovascular system and in neurodegeneration.
Collapse
Affiliation(s)
- Kamau M. M. Fahie
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kyriakos N. Papanicolaou
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Natasha E. Zachara
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
36
|
Lockridge A, Hanover JA. A nexus of lipid and O-Glcnac metabolism in physiology and disease. Front Endocrinol (Lausanne) 2022; 13:943576. [PMID: 36111295 PMCID: PMC9468787 DOI: 10.3389/fendo.2022.943576] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although traditionally considered a glucose metabolism-associated modification, the O-linked β-N-Acetylglucosamine (O-GlcNAc) regulatory system interacts extensively with lipids and is required to maintain lipid homeostasis. The enzymes of O-GlcNAc cycling have molecular properties consistent with those expected of broad-spectrum environmental sensors. By direct protein-protein interactions and catalytic modification, O-GlcNAc cycling enzymes may provide both acute and long-term adaptation to stress and other environmental stimuli such as nutrient availability. Depending on the cell type, hyperlipidemia potentiates or depresses O-GlcNAc levels, sometimes biphasically, through a diversity of unique mechanisms that target UDP-GlcNAc synthesis and the availability, activity and substrate selectivity of the glycosylation enzymes, O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA). At the same time, OGT activity in multiple tissues has been implicated in the homeostatic regulation of systemic lipid uptake, storage and release. Hyperlipidemic patterns of O-GlcNAcylation in these cells are consistent with both transient physiological adaptation and feedback uninhibited obesogenic and metabolic dysregulation. In this review, we summarize the numerous interconnections between lipid and O-GlcNAc metabolism. These links provide insights into how the O-GlcNAc regulatory system may contribute to lipid-associated diseases including obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Amber Lockridge
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
37
|
Hypoxia-triggered O-GlcNAcylation in the brain drives the glutamate-glutamine cycle and reduces sensitivity to sevoflurane in mice. Br J Anaesth 2022; 129:703-715. [PMID: 36031420 DOI: 10.1016/j.bja.2022.06.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 06/11/2022] [Accepted: 06/29/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Hypersensitivity to general anaesthetics predicts adverse postoperative outcomes in patients. Hypoxia exerts extensive pathophysiological effects on the brain; however, whether hypoxia influences sevoflurane sensitivity and its underlying mechanisms remain poorly understood. METHODS Mice were acclimated to hypoxia (oxygen 10% for 8 h day-1) for 28 days and anaesthetised with sevoflurane; the effective concentrations for 50% of the animals (EC50) showing loss of righting reflex (LORR) and loss of tail-pinch withdrawal response (LTWR) were determined. Positron emission tomography-computed tomography, O-glycoproteomics, seahorse analysis, carbon-13 tracing, site-specific mutagenesis, and electrophysiological techniques were performed to explore the underlying mechanisms. RESULTS Compared with the control group, the hypoxia-acclimated mice required higher concentrations of sevoflurane to present LORR and LTWR (EC50LORR: 1.61 [0.03]% vs 1.46 [0.04]%, P<0.01; EC50LTWR: 2.46 [0.14]% vs 2.22 [0.06]%, P<0.01). Hypoxia-induced reduction in sevoflurane sensitivity was correlated with elevation of protein O-linked N-acetylglucosamine (O-GlcNAc) modification in brain, especially in the thalamus, and could be abolished by 6-diazo-5-oxo-l-norleucine, a glutamine fructose-6-phosphate amidotransferase inhibitor, and mimicked by thiamet-G, a selective O-GlcNAcase inhibitor. Mechanistically, O-GlcNAcylation drives de novo synthesis of glutamine from glucose in astrocytes and promotes the glutamate-glutamine cycle, partially via glycolytic flux and activation of glutamine synthetase. CONCLUSIONS Intermittent hypoxia exposure decreased mouse sensitivity to sevoflurane anaesthesia through enhanced O-GlcNAc-dependent modulation of the glutamate-glutamine cycle in the brain.
Collapse
|
38
|
Sajid MS, Saleem S, Jabeen F, Najam-Ul-Haq M, Ressom HW. Terpolymeric platform with enhanced hydrophilicity via cysteic acid for serum intact glycopeptide analysis. Mikrochim Acta 2022; 189:277. [PMID: 35829791 DOI: 10.1007/s00604-022-05343-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/15/2022] [Indexed: 11/29/2022]
Abstract
A new polymeric (methyl methacrylate/ethylene glycol dimethacrylate/1,2-epoxy-5-hexene) base/matrix has been fabricated and decorated with zwitterionic hydrophilic cysteic acid (Cya) for the enrichment of intact N-glycopeptides from standards and biological samples. Terpolymer-Cya provides good enrichment efficiency, improved hydrophilicity, and selectivity by virtue of better surface area (2.09 × 102 m2/g) provided by terpolymer and the zwitterionic property offered by cysteic acid. Cysteic acid-functionalized polymeric hydrophilic interaction liquid chromatography (HILIC) sorbent enriches 35 and 24 N-linked glycopeptides via SPE (solid phase extraction) mode from tryptic digests of model glycoproteins, i.e., immunoglobulin G (IgG) and horseradish peroxidase (HRP), respectively. Zwitterionic chemistry of cysteine helps in achieving higher selectivity with BSA digest (1:200), and lower detection limit down to 100 attomoles with a complete glycosylation profile of each standard digest. The recovery of 81% and good reproducibility define the application of terpolymer-Cya for complex samples like a serum. Analysis of human serum provides a profile of 807 intact N-linked glycopeptides via nano-liquid chromatography-tandem mass spectrometry (nLC-MS/MS). To the best of our knowledge, this is the highest number of glycopeptides enriched by any HILIC sorbent. Selected glycoproteins are evaluated in link to various cancers including the breast, lung, uterine, and melanoma using single-nucleotide variances (BioMuta). This study represents the complete idea of using an in-house developed strategy as a successful tool to help analyze, relate, and answer glycoprotein-based clinical issues regarding cancers.
Collapse
Affiliation(s)
- Muhammad Salman Sajid
- Division of Analytical Chemistry, Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan
- Department of Oncology, Genomics and Epigenomics Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Shafaq Saleem
- Department of Chemistry, The Women University, Kutchery Campus, L.M.Q. Road, Multan, 66000, Pakistan
| | - Fahmida Jabeen
- Division of Analytical Chemistry, Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muhammad Najam-Ul-Haq
- Division of Analytical Chemistry, Institute of Chemical Sciences, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Habtom W Ressom
- Department of Oncology, Genomics and Epigenomics Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057, USA.
| |
Collapse
|
39
|
Zhao W, Liu Y, Xu L, He Y, Cai Z, Yu J, Zhang W, Xing C, Zhuang C, Qu Z. Targeting Necroptosis as a Promising Therapy for Alzheimer's Disease. ACS Chem Neurosci 2022; 13:1697-1713. [PMID: 35607807 DOI: 10.1021/acschemneuro.2c00172] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is an irreversible and progressive neurodegenerative disorder featured by memory loss and cognitive default. However, there has been no effective therapeutic approach to prevent the development of AD and the available therapies are only to alleviate some symptoms with limited efficacy and severe side effects. Necroptosis is a new kind of cell death, being regarded as a genetically programmed and regulated pattern of necrosis. Increasing evidence reveals that necroptosis is tightly related to the occurrence and development of AD. This review aims to summarize the potential role of necroptosis in AD progression and the therapeutic capacity of targeting necroptosis for AD patients.
Collapse
Affiliation(s)
- Wenli Zhao
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Yue Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Lijuan Xu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yuan He
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200070, China
| | - Zhenyu Cai
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200070, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Wannian Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Zhuo Qu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| |
Collapse
|
40
|
Burt RA, Alghusen IM, John Ephrame S, Villar MT, Artigues A, Slawson C. Mapping the O-GlcNAc Modified Proteome: Applications for Health and Disease. Front Mol Biosci 2022; 9:920727. [PMID: 35664676 PMCID: PMC9161079 DOI: 10.3389/fmolb.2022.920727] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/02/2022] [Indexed: 01/03/2023] Open
Abstract
O-GlcNAc is a pleotropic, enigmatic post-translational modification (PTM). This PTM modifies thousands of proteins differentially across tissue types and regulates diverse cellular signaling processes. O-GlcNAc is implicated in numerous diseases, and the advent of O-GlcNAc perturbation as a novel class of therapeutic underscores the importance of identifying and quantifying the O-GlcNAc modified proteome. Here, we review recent advances in mass spectrometry-based proteomics that will be critical in elucidating the role of this unique glycosylation system in health and disease.
Collapse
Affiliation(s)
- Rajan A. Burt
- University of Kansas Medical Center, Medical Scientist Training Program (MSTP), Kansas, KS, United States
| | - Ibtihal M. Alghusen
- Department Biochemistry, University of Kansas Medical Center, Kansas, KS, United States
| | - Sophiya John Ephrame
- Department Biochemistry, University of Kansas Medical Center, Kansas, KS, United States
| | - Maria T. Villar
- Department Biochemistry, University of Kansas Medical Center, Kansas, KS, United States
| | - Antonio Artigues
- Department Biochemistry, University of Kansas Medical Center, Kansas, KS, United States
| | - Chad Slawson
- University of Kansas Medical Center, Medical Scientist Training Program (MSTP), Kansas, KS, United States
- Department Biochemistry, University of Kansas Medical Center, Kansas, KS, United States
| |
Collapse
|
41
|
Gao AYL, Lourdin-De Filippis E, Orlowski J, McKinney RA. Roles of Endomembrane Alkali Cation/Proton Exchangers in Synaptic Function and Neurodevelopmental Disorders. Front Physiol 2022; 13:892196. [PMID: 35547574 PMCID: PMC9081726 DOI: 10.3389/fphys.2022.892196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/30/2022] [Indexed: 12/25/2022] Open
Abstract
Endomembrane alkali cation (Na+, K+)/proton (H+) exchangers (eNHEs) are increasingly associated with neurological disorders. These eNHEs play integral roles in regulating the luminal pH, processing, and trafficking of cargo along the secretory (Golgi and post-Golgi vesicles) and endocytic (early, recycling, and late endosomes) pathways, essential regulatory processes vital for neuronal development and plasticity. Given the complex morphology and compartmentalization of multipolar neurons, the contribution of eNHEs in maintaining optimal pH homeostasis and cargo trafficking is especially significant during periods of structural and functional development and remodeling. While the importance of eNHEs has been demonstrated in a variety of non-neuronal cell types, their involvement in neuronal function is less well understood. In this review, we will discuss their emerging roles in excitatory synaptic function, particularly as it pertains to cellular learning and remodeling. We will also explore their connections to neurodevelopmental conditions, including intellectual disability, autism, and attention deficit hyperactivity disorders.
Collapse
Affiliation(s)
- Andy Y L Gao
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | | | - John Orlowski
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - R Anne McKinney
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| |
Collapse
|
42
|
Permanne B, Sand A, Ousson S, Nény M, Hantson J, Schubert R, Wiessner C, Quattropani A, Beher D. O-GlcNAcase Inhibitor ASN90 is a Multimodal Drug Candidate for Tau and α-Synuclein Proteinopathies. ACS Chem Neurosci 2022; 13:1296-1314. [PMID: 35357812 PMCID: PMC9026285 DOI: 10.1021/acschemneuro.2c00057] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Neurodegenerative proteinopathies are characterized by the intracellular formation of insoluble and toxic protein aggregates in the brain that are closely linked to disease progression. In Alzheimer's disease and in rare tauopathies, aggregation of the microtubule-associated tau protein leads to the formation of neurofibrillary tangles (NFT). In Parkinson's disease (PD) and other α-synucleinopathies, intracellular Lewy bodies containing aggregates of α-synuclein constitute the pathological hallmark. Inhibition of the glycoside hydrolase O-GlcNAcase (OGA) prevents the removal of O-linked N-acetyl-d-glucosamine (O-GlcNAc) moieties from intracellular proteins and has emerged as an attractive therapeutic approach to prevent the formation of tau pathology. Like tau, α-synuclein is known to be modified with O-GlcNAc moieties and in vitro these have been shown to prevent its aggregation and toxicity. Here, we report the preclinical discovery and development of a novel small molecule OGA inhibitor, ASN90. Consistent with the substantial exposure of the drug and demonstrating target engagement in the brain, the clinical OGA inhibitor ASN90 promoted the O-GlcNAcylation of tau and α-synuclein in brains of transgenic mice after daily oral dosing. Across human tauopathy mouse models, oral administration of ASN90 prevented the development of tau pathology (NFT formation), functional deficits in motor behavior and breathing, and increased survival. In addition, ASN90 slowed the progression of motor impairment and reduced astrogliosis in a frequently utilized α-synuclein-dependent preclinical rodent model of PD. These findings provide a strong rationale for the development of OGA inhibitors as disease-modifying agents in both tauopathies and α-synucleinopathies. Since tau and α-synuclein pathologies frequently co-exist in neurodegenerative diseases, OGA inhibitors represent unique, multimodal drug candidates for further clinical development.
Collapse
Affiliation(s)
- Bruno Permanne
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Astrid Sand
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Solenne Ousson
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Maud Nény
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Jennifer Hantson
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Ryan Schubert
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Christoph Wiessner
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Anna Quattropani
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Dirk Beher
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| |
Collapse
|
43
|
Nelson AR. Peripheral Pathways to Neurovascular Unit Dysfunction, Cognitive Impairment, and Alzheimer’s Disease. Front Aging Neurosci 2022; 14:858429. [PMID: 35517047 PMCID: PMC9062225 DOI: 10.3389/fnagi.2022.858429] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. It was first described more than a century ago, and scientists are acquiring new data and learning novel information about the disease every day. Although there are nuances and details continuously being unraveled, many key players were identified in the early 1900’s by Dr. Oskar Fischer and Dr. Alois Alzheimer, including amyloid-beta (Aβ), tau, vascular abnormalities, gliosis, and a possible role of infections. More recently, there has been growing interest in and appreciation for neurovascular unit dysfunction that occurs early in mild cognitive impairment (MCI) before and independent of Aβ and tau brain accumulation. In the last decade, evidence that Aβ and tau oligomers are antimicrobial peptides generated in response to infection has expanded our knowledge and challenged preconceived notions. The concept that pathogenic germs cause infections generating an innate immune response (e.g., Aβ and tau produced by peripheral organs) that is associated with incident dementia is worthwhile considering in the context of sporadic AD with an unknown root cause. Therefore, the peripheral amyloid hypothesis to cognitive impairment and AD is proposed and remains to be vetted by future research. Meanwhile, humans remain complex variable organisms with individual risk factors that define their immune status, neurovascular function, and neuronal plasticity. In this focused review, the idea that infections and organ dysfunction contribute to Alzheimer’s disease, through the generation of peripheral amyloids and/or neurovascular unit dysfunction will be explored and discussed. Ultimately, many questions remain to be answered and critical areas of future exploration are highlighted.
Collapse
|
44
|
Glycomic and Glycoproteomic Techniques in Neurodegenerative Disorders and Neurotrauma: Towards Personalized Markers. Cells 2022; 11:cells11030581. [PMID: 35159390 PMCID: PMC8834236 DOI: 10.3390/cells11030581] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/22/2022] [Accepted: 02/03/2022] [Indexed: 12/16/2022] Open
Abstract
The proteome represents all the proteins expressed by a genome, a cell, a tissue, or an organism at any given time under defined physiological or pathological circumstances. Proteomic analysis has provided unparalleled opportunities for the discovery of expression patterns of proteins in a biological system, yielding precise and inclusive data about the system. Advances in the proteomics field opened the door to wider knowledge of the mechanisms underlying various post-translational modifications (PTMs) of proteins, including glycosylation. As of yet, the role of most of these PTMs remains unidentified. In this state-of-the-art review, we present a synopsis of glycosylation processes and the pathophysiological conditions that might ensue secondary to glycosylation shortcomings. The dynamics of protein glycosylation, a crucial mechanism that allows gene and pathway regulation, is described. We also explain how-at a biomolecular level-mutations in glycosylation-related genes may lead to neuropsychiatric manifestations and neurodegenerative disorders. We then analyze the shortcomings of glycoproteomic studies, putting into perspective their downfalls and the different advanced enrichment techniques that emanated to overcome some of these challenges. Furthermore, we summarize studies tackling the association between glycosylation and neuropsychiatric disorders and explore glycoproteomic changes in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington disease, multiple sclerosis, and amyotrophic lateral sclerosis. We finally conclude with the role of glycomics in the area of traumatic brain injury (TBI) and provide perspectives on the clinical application of glycoproteomics as potential diagnostic tools and their application in personalized medicine.
Collapse
|
45
|
Tools, tactics and objectives to interrogate cellular roles of O-GlcNAc in disease. Nat Chem Biol 2022; 18:8-17. [PMID: 34934185 PMCID: PMC8712397 DOI: 10.1038/s41589-021-00903-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/21/2021] [Indexed: 12/15/2022]
Abstract
The vast array of cell types of multicellular organisms must individually fine-tune their internal metabolism. One important metabolic and stress regulatory mechanism is the dynamic attachment/removal of glucose-derived sugar N-acetylglucosamine on proteins (O-GlcNAcylation). The number of proteins modified by O-GlcNAc is bewildering, with at least 7,000 sites in human cells. The outstanding challenge is determining how key O-GlcNAc sites regulate a target pathway amidst thousands of potential global sites. Innovative solutions are required to address this challenge in cell models and disease therapy. This Perspective shares critical suggestions for the O-GlcNAc field gleaned from the international O-GlcNAc community. Further, we summarize critical tools and tactics to enable newcomers to O-GlcNAc biology to drive innovation at the interface of metabolism and disease. The growing pace of O-GlcNAc research makes this a timely juncture to involve a wide array of scientists and new toolmakers to selectively approach the regulatory roles of O-GlcNAc in disease.
Collapse
|
46
|
Xie J, Kittur FS, Li PA, Hung CY. Rethinking the necessity of low glucose intervention for cerebral ischemia/reperfusion injury. Neural Regen Res 2021; 17:1397-1403. [PMID: 34916409 PMCID: PMC8771096 DOI: 10.4103/1673-5374.330592] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Glucose is the essential and almost exclusive metabolic fuel for the brain. Ischemic stroke caused by a blockage in one or more cerebral arteries quickly leads to a lack of regional cerebral blood supply resulting in severe glucose deprivation with subsequent induction of cellular homeostasis disturbance and eventual neuronal death. To make up ischemia-mediated adenosine 5′-triphosphate depletion, glucose in the ischemic penumbra area rapidly enters anaerobic metabolism to produce glycolytic adenosine 5′-triphosphate for cell survival. It appears that an increase in glucose in the ischemic brain would exert favorable effects. This notion is supported by in vitro studies, but generally denied by most in vivo studies. Clinical studies to manage increased blood glucose levels after stroke also failed to show any benefits or even brought out harmful effects while elevated admission blood glucose concentrations frequently correlated with poor outcomes. Surprisingly, strict glycaemic control in clinical practice also failed to yield any beneficial outcome. These controversial results from glucose management studies during the past three decades remain a challenging question of whether glucose intervention is needed for ischemic stroke care. This review provides a brief overview of the roles of cerebral glucose under normal and ischemic conditions and the results of managing glucose levels in non-diabetic patients. Moreover, the relationship between blood glucose and cerebral glucose during the ischemia/reperfusion processes and the potential benefits of low glucose supplements for non-diabetic patients are discussed.
Collapse
Affiliation(s)
- Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC, USA
| | - Farooqahmed S Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC, USA
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC, USA
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC, USA
| |
Collapse
|
47
|
Limorenko G, Lashuel HA. Revisiting the grammar of Tau aggregation and pathology formation: how new insights from brain pathology are shaping how we study and target Tauopathies. Chem Soc Rev 2021; 51:513-565. [PMID: 34889934 DOI: 10.1039/d1cs00127b] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Converging evidence continues to point towards Tau aggregation and pathology formation as central events in the pathogenesis of Alzheimer's disease and other Tauopathies. Despite significant advances in understanding the morphological and structural properties of Tau fibrils, many fundamental questions remain about what causes Tau to aggregate in the first place. The exact roles of cofactors, Tau post-translational modifications, and Tau interactome in regulating Tau aggregation, pathology formation, and toxicity remain unknown. Recent studies have put the spotlight on the wide gap between the complexity of Tau structures, aggregation, and pathology formation in the brain and the simplicity of experimental approaches used for modeling these processes in research laboratories. Embracing and deconstructing this complexity is an essential first step to understanding the role of Tau in health and disease. To help deconstruct this complexity and understand its implication for the development of effective Tau targeting diagnostics and therapies, we firstly review how our understanding of Tau aggregation and pathology formation has evolved over the past few decades. Secondly, we present an analysis of new findings and insights from recent studies illustrating the biochemical, structural, and functional heterogeneity of Tau aggregates. Thirdly, we discuss the importance of adopting new experimental approaches that embrace the complexity of Tau aggregation and pathology as an important first step towards developing mechanism- and structure-based therapies that account for the pathological and clinical heterogeneity of Alzheimer's disease and Tauopathies. We believe that this is essential to develop effective diagnostics and therapies to treat these devastating diseases.
Collapse
Affiliation(s)
- Galina Limorenko
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Federal de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
48
|
Glycoconjugate journal special issue on: the glycobiology of Parkinson's disease. Glycoconj J 2021; 39:55-74. [PMID: 34757539 DOI: 10.1007/s10719-021-10024-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/14/2021] [Accepted: 09/24/2021] [Indexed: 10/19/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that affects over 10 million aging people worldwide. This condition is characterized by the degeneration of dopaminergic neurons in the pars compacta region of the substantia nigra (SNpc) and by aggregation of proteins, commonly α-synuclein (SNCA). The formation of Lewy bodies that encapsulate aggregated proteins in lipid vesicles is a hallmark of PD. Glycosylation of proteins and neuroinflammation are involved in the pathogenesis. SNCA has many posttranslational modifications and interacts with components of membranes that affect aggregation. The large membrane lipid dolichol accumulates in the brain upon age and has a significant effect on membrane structure. The replacement of dopamine and dopaminergic neurons are at the forefront of therapeutic development. This review examines the role of membrane lipids, glycolipids, glycoproteins and dopamine in the aggregation of SNCA and development of PD. We discuss the SNCA-dopamine-neuromelanin-dolichol axis and the role of membranes in neuronal stem cells that could be a regenerative therapy for PD patients.
Collapse
|
49
|
Xu X, Zheng C, Lu D, Song CP, Zhang L. Phase separation in plants: New insights into cellular compartmentalization. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2021; 63:1835-1855. [PMID: 34314106 DOI: 10.1111/jipb.13152] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/16/2021] [Indexed: 05/16/2023]
Abstract
A fundamental challenge for cells is how to coordinate various biochemical reactions in space and time. To achieve spatiotemporal control, cells have developed organelles that are surrounded by lipid bilayer membranes. Further, membraneless compartmentalization, a process induced by dynamic physical association of biomolecules through phase transition offers another efficient mechanism for intracellular organization. While our understanding of phase separation was predominantly dependent on yeast and animal models, recent findings have provided compelling evidence for emerging roles of phase separation in plants. In this review, we first provide an overview of the current knowledge of phase separation, including its definition, biophysical principles, molecular features and regulatory mechanisms. Then we summarize plant-specific phase separation phenomena and describe their functions in plant biological processes in great detail. Moreover, we propose that phase separation is an evolutionarily conserved and efficient mechanism for cellular compartmentalization which allows for distinct metabolic processes and signaling pathways, and is especially beneficial for the sessile lifestyle of plants to quickly and efficiently respond to the changing environment.
Collapse
Affiliation(s)
- Xiumei Xu
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Canhui Zheng
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Dandan Lu
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Chun-Peng Song
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Lixin Zhang
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Kaifeng, 475004, China
| |
Collapse
|
50
|
Lee BE, Suh PG, Kim JI. O-GlcNAcylation in health and neurodegenerative diseases. Exp Mol Med 2021; 53:1674-1682. [PMID: 34837015 PMCID: PMC8639716 DOI: 10.1038/s12276-021-00709-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/09/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022] Open
Abstract
O-GlcNAcylation is a posttranslational modification that adds O-linked β-N-acetylglucosamine (O-GlcNAc) to serine or threonine residues of many proteins. This protein modification interacts with key cellular pathways involved in transcription, translation, and proteostasis. Although ubiquitous throughout the body, O-GlcNAc is particularly abundant in the brain, and various proteins commonly found at synapses are O-GlcNAcylated. Recent studies have demonstrated that the modulation of O-GlcNAc in the brain alters synaptic and neuronal functions. Furthermore, altered brain O-GlcNAcylation is associated with either the etiology or pathology of numerous neurodegenerative diseases, while the manipulation of O-GlcNAc exerts neuroprotective effects against these diseases. Although the detailed molecular mechanisms underlying the functional roles of O-GlcNAcylation in the brain remain unclear, O-GlcNAcylation is critical for regulating diverse neural functions, and its levels change during normal and pathological aging. In this review, we will highlight the functional importance of O-GlcNAcylation in the brain and neurodegenerative diseases.
Collapse
Affiliation(s)
- Byeong Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Pann-Ghill Suh
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|