1
|
Ni Y, Yin YN, Yu WB, Liu YQ, Zhao J, Yan YC, Zhao WB, Tang YL, Sun YM, Liu FT, Ran P, Wu JJ, Ding C, Wang J. Assessing Plasma APLP1 for the Progression of Parkinson's Disease: Insights from HSPD and PPMI Cohorts. Mov Disord 2025; 40:969-974. [PMID: 39968922 DOI: 10.1002/mds.30154] [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: 09/28/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Amyloid precursor-like protein 1 (APLP1) is involved in pathological α-synuclein transmission, but its role in Parkinson's disease (PD) progression has not been explored. OBJECTIVE This study investigates APLP1 as a potential predictor for motor and cognitive deterioration in PD. METHODS Plasma APLP1 levels were measured in PD patients from the Huashan Hospital for Parkinson's Disease (HSPD) and Parkinson's Disease Progression Markers Initiative (PPMI) cohorts. A total of 916 participants were recruited in the HSPD cohort, and 171 participants were in the PPMI cohort. Longitudinal analysis examined the association between baseline APLP1 levels and PD progression. RESULTS A significant increase in APLP1 levels was observed in PD patients compared to healthy controls. Longitudinal analysis showed that patients with elevated baseline APLP1 levels experienced faster motor deterioration in HSPD cohort (HR = 3.627, P < 0.0001). CONCLUSIONS The data indicate that APLP1 is associated with the progression of PD, potentially offering a measurable indicator of disease progression. © 2025 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- You Ni
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya-Nan Yin
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Wen-Bo Yu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Qi Liu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jue Zhao
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Chen Yan
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wan-Bing Zhao
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Lin Tang
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Min Sun
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Feng-Tao Liu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Peng Ran
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Jian-Jun Wu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Ding
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Fan X, Xie Y, Cao S, Zhu L, Wang X. VPS35-Retromer: Multifunctional Roles in Various Biological Processes - A Focus on Neurodegenerative Diseases and Cancer. J Inflamm Res 2025; 18:4665-4680. [PMID: 40195959 PMCID: PMC11975009 DOI: 10.2147/jir.s510768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/23/2025] [Indexed: 04/09/2025] Open
Abstract
The Vacuolar Protein Sorting 35 (VPS35)-Retromer complex plays a pivotal role in intracellular protein trafficking and recycling. As an integral component of the Retromer complex, VPS35 selectively recognizes and retrogradely transports membrane protein receptors to the trans-Golgi network, thereby preventing the degradation of transmembrane proteins by lysosomes after they have fulfilled their physiological functions, and facilitating their continued activity. VPS35 regulates autophagy, mitophagy, mitochondrial homeostasis, and various other biological processes, including epidermal regeneration, neuronal iron homeostasis, and synaptic function. Studies have shown that mutations or dysfunctions in VPS35 disrupt the normal operation of Retromer, impair neuronal health and survival, and contribute to the onset of neurodegenerative diseases such as Parkinson's and Alzheimer's diseases. Additionally, VPS35 modulates tumor growth and metastasis in cancers such as liver and breast cancer through the regulation of multiple signaling pathways. Targeting VPS35 might be a potential therapy in clinic treatment of neurodegenerative diseases and cancers.
Collapse
Affiliation(s)
- Xiaoyang Fan
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, People’s Republic of China
| | - Yuqi Xie
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, People’s Republic of China
| | - Sitong Cao
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, People’s Republic of China
| | - Li Zhu
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, People’s Republic of China
| | - Xueting Wang
- Institute of Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, People’s Republic of China
| |
Collapse
|
3
|
Lourbopoulos A, Müller SA, Jocher G, Wick M, Plesnila N, Lichtenthaler SF. An Improved Method for Sampling and Quantitative Protein Analytics of Cerebrospinal Fluid of Individual Mice. Mol Cell Proteomics 2025; 24:100958. [PMID: 40157722 PMCID: PMC12090247 DOI: 10.1016/j.mcpro.2025.100958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025] Open
Abstract
The mouse is the species most commonly used in preclinical research, but protein analytics of murine cerebrospinal fluid (CSF) remains challenging because of low sample volumes (often <10 μl) and frequent contaminations with blood. We developed an improved CSF sampling method that allows routine collection of larger volumes (20-30 μl) of pure CSF from individual mice, enabling multiple protein analytical assays from a single sample. Based on cell counts and hemoglobin ELISAs, we provide an easy quality control workflow for obtaining cell- and blood-free murine CSF. Through mass spectrometry-based proteomics using an absolutely quantified external standard, we estimated concentrations for hundreds of mouse CSF proteins. While repeated CSF sampling from the same mouse was possible, it induced CSF proteome changes. Applying the improved method, we found that the mouse CSF proteome remains largely stable over time in wild-type mice, but that amyloid pathology in the 5xFAD mouse model of Alzheimer's disease massively changes the CSF proteome. Neurofilament light chain and TREM2, markers of neurodegeneration and activated microglia, respectively, were strongly upregulated and validated using immunoassays. In conclusion, our refined murine CSF collection method overcomes previous limitations, allowing multiple quantitative protein analyses for applications in biomedicine.
Collapse
Affiliation(s)
- Athanasios Lourbopoulos
- Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Georg Jocher
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Manfred Wick
- Institute of Laboratory Medicine, University Hospital Ludwig Maximilian University, Munich, Germany
| | - Nikolaus Plesnila
- Laboratory of Experimental Stroke Research, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
4
|
Mishra AK, Tripathi MK, Kumar D, Gupta SP. Neurons Specialize in Presynaptic Autophagy: A Perspective to Ameliorate Neurodegeneration. Mol Neurobiol 2025; 62:2626-2640. [PMID: 39141193 DOI: 10.1007/s12035-024-04399-8] [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: 05/06/2024] [Accepted: 07/24/2024] [Indexed: 08/15/2024]
Abstract
The efficient and prolonged neurotransmission is reliant on the coordinated action of numerous synaptic proteins in the presynaptic compartment that remodels synaptic vesicles for neurotransmitter packaging and facilitates their exocytosis. Once a cycle of neurotransmission is completed, membranes and associated proteins are endocytosed into the cytoplasm for recycling or degradation. Both exocytosis and endocytosis are closely regulated in a timely and spatially constrained manner. Recent research demonstrated the impact of dysfunctional synaptic vesicle retrieval in causing retrograde degeneration of midbrain neurons and has highlighted the importance of such endocytic proteins, including auxilin, synaptojanin1 (SJ1), and endophilin A (EndoA) in neurodegenerative diseases. Additionally, the role of other associated proteins, including leucine-rich repeat kinase 2 (LRRK2), adaptor proteins, and retromer proteins, is being investigated for their roles in regulating synaptic vesicle recycling. Research suggests that the degradation of defective vesicles via presynaptic autophagy, followed by their recycling, not only revitalizes them in the active zone but also contributes to strengthening synaptic plasticity. The presynaptic autophagy rejuvenating terminals and maintaining neuroplasticity is unique in autophagosome formation. It involves several synaptic proteins to support autophagosome construction in tiny compartments and their retrograde trafficking toward the cell bodies. Despite having a comprehensive understanding of ATG proteins in autophagy, we still lack a framework to explain how autophagy is triggered and potentiated in compact presynaptic compartments. Here, we reviewed synaptic proteins' involvement in forming presynaptic autophagosomes and in retrograde trafficking of terminal cargos. The review also discusses the status of endocytic proteins and endocytosis-regulating proteins in neurodegenerative diseases and strategies to combat neurodegeneration.
Collapse
Affiliation(s)
- Abhishek Kumar Mishra
- Department of Zoology, Government Shaheed Gendsingh College, Charama, Uttar Bastar Kanker, 494 337, Chhattisgarh, India.
| | - Manish Kumar Tripathi
- School of Pharmacy, Faculty of Medicine, Institute for Drug Research, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Dipak Kumar
- Department of Zoology, Munger University, Munger, Bihar, India
| | - Satya Prakash Gupta
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221 005, India
| |
Collapse
|
5
|
Liu Y, Xia X, Zheng M, Shi B. Bio-Nano Toolbox for Precision Alzheimer's Disease Gene Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314354. [PMID: 38778446 DOI: 10.1002/adma.202314354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is the most burdensome aging-associated neurodegenerative disorder, and its treatment encounters numerous failures during drug development. Although there are newly approved in-market β-amyloid targeting antibody solutions, pathological heterogeneity among patient populations still challenges the treatment outcome. Emerging advances in gene therapies offer opportunities for more precise personalized medicine; while, major obstacles including the pathological heterogeneity among patient populations, the puzzled mechanism for druggable target development, and the precision delivery of functional therapeutic elements across the blood-brain barrier remain and limit the use of gene therapy for central neuronal diseases. Aiming for "precision delivery" challenges, nanomedicine provides versatile platforms that may overcome the targeted delivery challenges for AD gene therapy. In this perspective, to picture a toolbox for AD gene therapy strategy development, the most recent advances from benchtop to clinics are highlighted, possibly available gene therapy targets, tools, and delivery platforms are outlined, their challenges as well as rational design elements are addressed, and perspectives in this promising research field are discussed.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xue Xia
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| |
Collapse
|
6
|
Parra Bravo C, Giani AM, Madero-Perez J, Zhao Z, Wan Y, Samelson AJ, Wong MY, Evangelisti A, Cordes E, Fan L, Ye P, Zhu D, Pozner T, Mercedes M, Patel T, Yarahmady A, Carling GK, Sterky FH, Lee VMY, Lee EB, DeTure M, Dickson DW, Sharma M, Mok SA, Luo W, Zhao M, Kampmann M, Gong S, Gan L. Human iPSC 4R tauopathy model uncovers modifiers of tau propagation. Cell 2024; 187:2446-2464.e22. [PMID: 38582079 PMCID: PMC11365117 DOI: 10.1016/j.cell.2024.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 01/12/2024] [Accepted: 03/12/2024] [Indexed: 04/08/2024]
Abstract
Tauopathies are age-associated neurodegenerative diseases whose mechanistic underpinnings remain elusive, partially due to a lack of appropriate human models. Here, we engineered human induced pluripotent stem cell (hiPSC)-derived neuronal lines to express 4R Tau and 4R Tau carrying the P301S MAPT mutation when differentiated into neurons. 4R-P301S neurons display progressive Tau inclusions upon seeding with Tau fibrils and recapitulate features of tauopathy phenotypes including shared transcriptomic signatures, autophagic body accumulation, and reduced neuronal activity. A CRISPRi screen of genes associated with Tau pathobiology identified over 500 genetic modifiers of seeding-induced Tau propagation, including retromer VPS29 and genes in the UFMylation cascade. In progressive supranuclear palsy (PSP) and Alzheimer's Disease (AD) brains, the UFMylation cascade is altered in neurofibrillary-tangle-bearing neurons. Inhibiting the UFMylation cascade in vitro and in vivo suppressed seeding-induced Tau propagation. This model provides a robust platform to identify novel therapeutic strategies for 4R tauopathy.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10021, USA
| | - Alice Maria Giani
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jesus Madero-Perez
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Zeping Zhao
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yuansong Wan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Avi J Samelson
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Alessandro Evangelisti
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ethan Cordes
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Pearly Ye
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Daphne Zhu
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Tatyana Pozner
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Maria Mercedes
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Tark Patel
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Allan Yarahmady
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Gillian K Carling
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Fredrik H Sterky
- Department of Laboratory Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; Department of Clinical Chemistry, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Virginia M Y Lee
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Edward B Lee
- Institute of Aging, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mingrui Zhao
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
7
|
Alessi DR, Cullen PJ, Cookson M, Merchant KM, Small SA. Retromer-dependent lysosomal stress in Parkinson's disease. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220376. [PMID: 38368937 PMCID: PMC10874697 DOI: 10.1098/rstb.2022.0376] [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: 03/07/2023] [Accepted: 11/27/2023] [Indexed: 02/20/2024] Open
Abstract
While causative mutations in complex disorders are rare, they can be used to extract a biological pathway whose pathogenicity can generalize to common forms of the disease. Here we begin by relying on the biological consequences of mutations in LRRK2 and VPS35, genetic causes of autosomal-dominant Parkinson's disease, to hypothesize that 'Retromer-dependent lysosomal stress' represents a pathway that can generalize to idiopathic Parkinson's disease. Next, we outline a series of studies that can test this hypothesis, including the development of biomarkers of pathway dysfunction. If validated, the hypothesis can suggest a unified mechanism of disease and might inform future diagnostic and therapeutic investigations. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
- Dario R. Alessi
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Peter J. Cullen
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, Bristol BS8 1TD, UK
| | - Mark Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kalpana M. Merchant
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60208, USA
| | - Scott A. Small
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| |
Collapse
|
8
|
Herman M, Randall GW, Spiegel JL, Maldonado DJ, Simoes S. Endo-lysosomal dysfunction in neurodegenerative diseases: opinion on current progress and future direction in the use of exosomes as biomarkers. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220387. [PMID: 38368936 PMCID: PMC10874701 DOI: 10.1098/rstb.2022.0387] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/27/2023] [Indexed: 02/20/2024] Open
Abstract
Over the past two decades, increased research has highlighted the connection between endosomal trafficking defects and neurodegeneration. The endo-lysosomal network is an important, complex cellular system specialized in the transport of proteins, lipids, and other metabolites, essential for cell homeostasis. Disruption of this pathway is linked to a wide range of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease and frontotemporal dementia. Furthermore, there is strong evidence that defects in this pathway create opportunities for diagnostic and therapeutic intervention. In this Opinion piece, we concisely address the role of endo-lysosomal dysfunction in five neurodegenerative diseases and discuss how future research can investigate this intracellular pathway, including extracellular vesicles with a specific focus on exosomes for the identification of novel disease biomarkers. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
- Mathieu Herman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Grace W. Randall
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Julia L. Spiegel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Delphina J. Maldonado
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sabrina Simoes
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
9
|
Zhang Y, Wu C, Jiang W, Cao Y, Chen D. VGLUT2 and APP family: unraveling the neurobiochemical mechanisms of neurostimulation therapy to STZ-induced diabetes and neuropathy. Front Endocrinol (Lausanne) 2024; 15:1336854. [PMID: 38370359 PMCID: PMC10869491 DOI: 10.3389/fendo.2024.1336854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Diabetic Peripheral Neuropathy (DPN) poses an escalating threat to public health, profoundly impacting well-being and quality of life. Despite its rising prevalence, the pathogenesis of DPN remains enigmatic, and existing clinical interventions fall short of achieving meaningful reversals of the condition. Notably, neurostimulation techniques have shown promising efficacy in alleviating DPN symptoms, underscoring the imperative to elucidate the neurobiochemical mechanisms underlying DPN. This study employs an integrated multi-omics approach to explore DPN and its response to neurostimulation therapy. Our investigation unveiled a distinctive pattern of vesicular glutamate transporter 2 (VGLUT2) expression in DPN, rigorously confirmed through qPCR and Western blot analyses in DPN C57 mouse model induced by intraperitoneal Streptozotocin (STZ) injection. Additionally, combining microarray and qPCR methodologies, we revealed and substantiated variations in the expression of the Amyloid Precursor Protein (APP) family in STZ-induced DPN mice. Analyzing the transcriptomic dataset generated from neurostimulation therapy for DPN, we intricately explored the differential expression patterns of VGLUT2 and APPs. Through correlation analysis, protein-protein interaction predictions, and functional enrichment analyses, we predicted the key biological processes involving VGLUT2 and the APP family in the pathogenesis of DPN and during neurostimulation therapy. This comprehensive study not only advances our understanding of the pathogenesis of DPN but also provides a theoretical foundation for innovative strategies in neurostimulation therapy for DPN. The integration of multi-omics data facilitates a holistic view of the molecular intricacies of DPN, paving the way for more targeted and effective therapeutic interventions.
Collapse
Affiliation(s)
- Yitong Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Chenxuan Wu
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Wenqi Jiang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dongtai Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
10
|
Marks JD, Ayuso VE, Carlomagno Y, Yue M, Todd TW, Hao Y, Li Z, McEachin ZT, Shantaraman A, Duong DM, Daughrity LM, Jansen-West K, Shao W, Calliari A, Bejarano JG, DeTure M, Rawlinson B, Casey MC, Lilley MT, Donahue MH, Jawahar VM, Boeve BF, Petersen RC, Knopman DS, Oskarsson B, Graff-Radford NR, Wszolek ZK, Dickson DW, Josephs KA, Qi YA, Seyfried NT, Ward ME, Zhang YJ, Prudencio M, Petrucelli L, Cook CN. TMEM106B core deposition associates with TDP-43 pathology and is increased in risk SNP carriers for frontotemporal dementia. Sci Transl Med 2024; 16:eadf9735. [PMID: 38232138 PMCID: PMC10841341 DOI: 10.1126/scitranslmed.adf9735] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Genetic variation at the transmembrane protein 106B gene (TMEM106B) has been linked to risk of frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) through an unknown mechanism. We found that presence of the TMEM106B rs3173615 protective genotype was associated with longer survival after symptom onset in a postmortem FTLD-TDP cohort, suggesting a slower disease course. The seminal discovery that filaments derived from TMEM106B is a common feature in aging and, across a range of neurodegenerative disorders, suggests that genetic variants in TMEM106B could modulate disease risk and progression through modulating TMEM106B aggregation. To explore this possibility and assess the pathological relevance of TMEM106B accumulation, we generated a new antibody targeting the TMEM106B filament core sequence. Analysis of postmortem samples revealed that the TMEM106B rs3173615 risk allele was associated with higher TMEM106B core accumulation in patients with FTLD-TDP. In contrast, minimal TMEM106B core deposition was detected in carriers of the protective allele. Although the abundance of monomeric full-length TMEM106B was unchanged, carriers of the protective genotype exhibited an increase in dimeric full-length TMEM106B. Increased TMEM106B core deposition was also associated with enhanced TDP-43 dysfunction, and interactome data suggested a role for TMEM106B core filaments in impaired RNA transport, local translation, and endolysosomal function in FTLD-TDP. Overall, these findings suggest that prevention of TMEM106B core accumulation is central to the mechanism by which the TMEM106B protective haplotype reduces disease risk and slows progression.
Collapse
Affiliation(s)
- Jordan D. Marks
- Medical Scientist Training Program, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Virginia Estades Ayuso
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yari Carlomagno
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Mei Yue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tiffany W. Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ying Hao
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ziyi Li
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zachary T. McEachin
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
- Department for Human Genetics, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Anantharaman Shantaraman
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Duc M. Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
| | | | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wei Shao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Anna Calliari
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Bailey Rawlinson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Meredith T. Lilley
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Megan H. Donahue
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | | | | - Björn Oskarsson
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | - Dennis W. Dickson
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Yue A. Qi
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicholas T. Seyfried
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Michael E. Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong-Jie Zhang
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Mercedes Prudencio
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Leonard Petrucelli
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Casey N. Cook
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
11
|
Wilson KA, Bar S, Dammer EB, Carrera EM, Hodge BA, Hilsabeck TAU, Bons J, Brownridge GW, Beck JN, Rose J, Granath-Panelo M, Nelson CS, Qi G, Gerencser AA, Lan J, Afenjar A, Chawla G, Brem RB, Campeau PM, Bellen HJ, Schilling B, Seyfried NT, Ellerby LM, Kapahi P. OXR1 maintains the retromer to delay brain aging under dietary restriction. Nat Commun 2024; 15:467. [PMID: 38212606 PMCID: PMC10784588 DOI: 10.1038/s41467-023-44343-3] [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: 08/02/2023] [Accepted: 12/07/2023] [Indexed: 01/13/2024] Open
Abstract
Dietary restriction (DR) delays aging, but the mechanism remains unclear. We identified polymorphisms in mtd, the fly homolog of OXR1, which influenced lifespan and mtd expression in response to DR. Knockdown in adulthood inhibited DR-mediated lifespan extension in female flies. We found that mtd/OXR1 expression declines with age and it interacts with the retromer, which regulates trafficking of proteins and lipids. Loss of mtd/OXR1 destabilized the retromer, causing improper protein trafficking and endolysosomal defects. Overexpression of retromer genes or pharmacological restabilization with R55 rescued lifespan and neurodegeneration in mtd-deficient flies and endolysosomal defects in fibroblasts from patients with lethal loss-of-function of OXR1 variants. Multi-omic analyses in flies and humans showed that decreased Mtd/OXR1 is associated with aging and neurological diseases. mtd/OXR1 overexpression rescued age-related visual decline and tauopathy in a fly model. Hence, OXR1 plays a conserved role in preserving retromer function and is critical for neuronal health and longevity.
Collapse
Affiliation(s)
- Kenneth A Wilson
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Sudipta Bar
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | - Brian A Hodge
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Tyler A U Hilsabeck
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Jennifer N Beck
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Jacob Rose
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | | | - Grace Qi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Jianfeng Lan
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Guanxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Afilliated Hospital of Guilin Medican University, Guilin, 541001, Guanxi, China
| | - Alexandra Afenjar
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire, Paris, 75012, France
- Département de Génétique et Embryologie Médicale, CRMR des Malformations et Maladies Congénitales du Cervelet, GRC ConCer-LD, Sorbonne Universités, Hôpital Trousseau, Paris, 75012, France
| | - Geetanjali Chawla
- RNA Biology Laboratory, Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institute of Eminence, NH91, Tehsil Dadri, G. B. Nagar, 201314, Uttar Pradesh, India
| | - Rachel B Brem
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, 111 Koshland Hall, Berkeley, CA, 94720, USA
| | - Philippe M Campeau
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC, H3T 1J4, Canada
| | - Hugo J Bellen
- Departments of Molecular and Human Genetics and Neuroscience, Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lisa M Ellerby
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
12
|
Ishida K, Yamada K. A Method to Collect Cerebrospinal Fluid from Mouse Cisterna Magna to Determine Extracellular Tau Levels. Methods Mol Biol 2024; 2754:343-349. [PMID: 38512675 DOI: 10.1007/978-1-0716-3629-9_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Despite being a cytoplasmic protein abundant in neurons, tau is detectable in various extracellular fluids. In addition to being passively released from dying/degenerating neurons, tau is also actively released from living neurons in a neuronal activity-dependent mechanism. In vivo, tau released from neurons first appears in brain interstitial fluid (ISF) and subsequently drains into cerebrospinal fluid (CSF) by glymphatic system. Changes in CSF tau levels alter during the course of AD pathogenesis and are considered to predict the disease-progression of AD. A method to collect CSF from various mouse models of AD will serve as a valuable tool to investigate the dynamics of physiological/pathological tau released from neurons. In this chapter, we describe and characterize a method that reliably collects a relatively large volume of CSF from anesthetized mice.
Collapse
Affiliation(s)
- Kazuhisa Ishida
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
13
|
Aljassabi A, Zieneldien T, Kim J, Regmi D, Cao C. Alzheimer's Disease Immunotherapy: Current Strategies and Future Prospects. J Alzheimers Dis 2024; 98:755-772. [PMID: 38489183 DOI: 10.3233/jad-231163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Alzheimer's disease (AD) is an extremely complex and heterogeneous pathology influenced by many factors contributing to its onset and progression, including aging, amyloid-beta (Aβ) plaques, tau fibril accumulation, inflammation, etc. Despite promising advances in drug development, there is no cure for AD. Although there have been substantial advancements in understanding the pathogenesis of AD, there have been over 200 unsuccessful clinical trials in the past decade. In recent years, immunotherapies have been at the forefront of these efforts. Immunotherapy alludes to the immunological field that strives to identify disease treatments via the enhancement, suppression, or induction of immune responses. Interestingly, immunotherapy in AD is a relatively new approach for non-infectious disease. At present, antibody therapy (passive immunotherapy) that targets anti-Aβ aimed to prevent the fibrillization of Aβ peptides and disrupt pre-existing fibrils is a predominant AD immunotherapy due to the continuous failure of active immunotherapy for AD. The most rational and safe strategies will be those targeting the toxic molecule without triggering an abnormal immune response, offering therapeutic advantages, thus making clinical trial design more efficient. This review offers a concise overview of immunotherapeutic strategies, including active and passive immunotherapy for AD. Our review encompasses approved methods and those presently under investigation in clinical trials, while elucidating the recent challenges, complications, successes, and potential treatments. Thus, immunotherapies targeting Aβ throughout the disease progression using a mutant oligomer-Aβ stimulated dendritic cell vaccine may offer a promising therapy in AD.
Collapse
Affiliation(s)
- Ali Aljassabi
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Tarek Zieneldien
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Janice Kim
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Deepika Regmi
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| |
Collapse
|
14
|
Zhao L, Deng H, Yang Q, Tang Y, Zhao J, Li P, Zhang S, Yong X, Li T, Billadeau DD, Jia D. FAM91A1-TBC1D23 complex structure reveals human genetic variations susceptible for PCH. Proc Natl Acad Sci U S A 2023; 120:e2309910120. [PMID: 37903274 PMCID: PMC10636324 DOI: 10.1073/pnas.2309910120] [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: 06/19/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
Pontocerebellar hypoplasia (PCH) is a group of rare neurodevelopmental disorders with limited diagnostic and therapeutic options. Mutations in WDR11, a subunit of the FAM91A1 complex, have been found in patients with PCH-like symptoms; however, definitive evidence that the mutations are causal is still lacking. Here, we show that depletion of FAM91A1 results in developmental defects in zebrafish similar to that of TBC1D23, an established PCH gene. FAM91A1 and TBC1D23 directly interact with each other and cooperate to regulate endosome-to-Golgi trafficking of KIAA0319L, a protein known to regulate axonal growth. Crystal structure of the FAM91A1-TBC1D23 complex reveals that TBC1D23 binds to a conserved surface on FAM91A1 by assuming a Z-shaped conformation. More importantly, the interaction between FAM91A1 and TBC1D23 can be used to predict the risk of certain TBC1D23-associated mutations to PCH. Collectively, our study provides a molecular basis for the interaction between TBC1D23 and FAM91A1 and suggests that disrupted endosomal trafficking underlies multiple PCH subtypes.
Collapse
Affiliation(s)
- Lin Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Huaqing Deng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Qing Yang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Yingying Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Jia Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Ping Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Sitao Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Tianxing Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| | - Daniel D. Billadeau
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN55905
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu610041, China
| |
Collapse
|
15
|
Chen X, Wang M, Niu J, Ma J, Qian J, Ni L, Cheng P, You H, Chen J. Plasma Aβ 42:Aβ 40 ratio as a biomarker for cognitive impairment in haemodialysis patients: a multicentre study. Clin Kidney J 2023; 16:2129-2140. [PMID: 37915896 PMCID: PMC10616443 DOI: 10.1093/ckj/sfad173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Indexed: 11/03/2023] Open
Abstract
Background Mild cognitive impairment (MCI) and dementia are more prevalent in patients undergoing haemodialysis (HD). Although the cerebrospinal fluid amyloid beta (Aβ) and tau (τ) have proven to be valid biomarkers for the diagnosis of Alzheimer's disease (AD) in the general population, the roles of plasma Aβ and τ for the diagnosis of cognitive impairment in HD patients remain unknown. Methods We conducted a cross-sectional study including patients receiving HD in three hospitals in Shanghai. All patients completed the Montreal Cognitive Assessment-Basic (MoCA-B). To validate the effectiveness of the MoCA-B score for screening MCI, a subset group underwent neuropsychological batteries. Serum proteomes were compared in HD patients with normal cognitive function and dementia. Plasma Aβ42, Aβ40 and total τ were measured using a single molecule array. Results A total of 311 HD patients were enrolled (mean age 63 years, 55% male). The best cut-off score of MoCA-B for differentiating MCI and normal cognition was 24, with an area under the curve of 0.94. Serum proteomics revealed that neurodegenerative pathways related to AD were enriched in HD patients with dementia. The plasma Aβ42:Aβ40 ratio was significantly reduced in patients with MCI and dementia and was independently associated with cognitive function after adjusting for age, sex and education levels. Conclusions We validated the MoCA-B as an optimal cognitive function screening instrument for MCI in HD patients. The plasma Aβ42:Aβ40 ratio was a potential biomarker in distinguishing normal cognition, MCI and dementia in HD populations.
Collapse
Affiliation(s)
- Xujiao Chen
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mengjing Wang
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianying Niu
- Division of Nephrology, Fifth People's Hospital Fudan University, Shanghai, China
| | - Jun Ma
- Division of Nephrology, Jingan District Centre Hospital of Shanghai, Shanghai, China
| | - Jing Qian
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Ni
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Cheng
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Huaizhou You
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Chen
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Young JE, Holstege H, Andersen OM, Petsko GA, Small SA. On the causal role of retromer-dependent endosomal recycling in Alzheimer's disease. Nat Cell Biol 2023; 25:1394-1397. [PMID: 37803174 PMCID: PMC10788784 DOI: 10.1038/s41556-023-01245-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Recent findings ranging from genetics to structural biology, together with studies in human neurons, animal models and patient brains, implicate the retromer-dependent endosomal recycling pathway as both causal and common in Alzheimer’s disease.
Collapse
Affiliation(s)
- Jessica E Young
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Henne Holstege
- Genomics of Neurodegenerative Diseases and Aging, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gregory A Petsko
- Department of Neurology and the Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham & Women's Hospital, Boston, MA, USA
| | - Scott A Small
- Department of Neurology, Columbia University, New York, NY, USA.
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| |
Collapse
|
17
|
Richards CM, McRae SA, Ranger AL, Klegeris A. Extracellular histones as damage-associated molecular patterns in neuroinflammatory responses. Rev Neurosci 2023; 34:533-558. [PMID: 36368030 DOI: 10.1515/revneuro-2022-0091] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 07/20/2023]
Abstract
The four core histones H2A, H2B, H3, H4, and the linker histone H1 primarily bind DNA and regulate gene expression within the nucleus. Evidence collected mainly from the peripheral tissues illustrates that histones can be released into the extracellular space by activated or damaged cells. In this article, we first summarize the innate immune-modulatory properties of extracellular histones and histone-containing complexes, such as nucleosomes, and neutrophil extracellular traps (NETs), described in peripheral tissues. There, histones act as damage-associated molecular patterns (DAMPs), which are a class of endogenous molecules that trigger immune responses by interacting directly with the cellular membranes and activating pattern recognition receptors (PRRs), such as toll-like receptors (TLR) 2, 4, 9 and the receptor for advanced glycation end-products (RAGE). We then focus on the available evidence implicating extracellular histones as DAMPs of the central nervous system (CNS). It is becoming evident that histones are present in the brain parenchyma after crossing the blood-brain barrier (BBB) or being released by several types of brain cells, including neurons, microglia, and astrocytes. However, studies on the DAMP-like effects of histones on CNS cells are limited. For example, TLR4 is the only known molecular target of CNS extracellular histones and their interactions with other PRRs expressed by brain cells have not been observed. Nevertheless, extracellular histones are implicated in the pathogenesis of a variety of neurological disorders characterized by sterile neuroinflammation; therefore, detailed studies on the role these proteins and their complexes play in these pathologies could identify novel therapeutic targets.
Collapse
Affiliation(s)
- Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| | - Seamus A McRae
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| | - Athena L Ranger
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna V1V 1V7, BC, Canada
| |
Collapse
|
18
|
Bravo CP, Giani AM, Perez JM, Zhao Z, Samelson A, Wong MY, Evangelisti A, Fan L, Pozner T, Mercedes M, Ye P, Patel T, Yarahmady A, Carling G, Lee VMY, Sharma M, Mok SA, Luo W, Zhao M, Kampmann M, Gong S, Gan L. Human iPSC 4R tauopathy model uncovers modifiers of tau propagation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.19.544278. [PMID: 37745431 PMCID: PMC10516028 DOI: 10.1101/2023.06.19.544278] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Tauopathies are age-associated neurodegenerative diseases whose mechanistic underpinnings remain elusive, partially due to lack of appropriate human models. Current human induced pluripotent stem cell (hiPSC)-derived neurons express very low levels of 4-repeat (4R)-tau isoforms that are normally expressed in adult brain. Here, we engineered new iPSC lines to express 4R-tau and 4R-tau carrying the P301S MAPT mutation when differentiated into neurons. 4R-P301S neurons display progressive Tau inclusions upon seeding with Tau fibrils and recapitulate features of tauopathy phenotypes, including shared transcriptomic signatures, autophagic body accumulation, and impaired neuronal activity. A CRISPRi screen of genes associated with Tau pathobiology identified over 500 genetic modifiers of Tau-seeding-induced Tau propagation, including retromer VPS29 and the UFMylation cascade as top modifiers. In AD brains, the UFMylation cascade is altered in neurofibrillary-tangle-bearing neurons. Inhibiting the UFMylation cascade suppressed seeding-induced Tau propagation. This model provides a powerful platform to identify novel therapeutic strategies for 4R tauopathy.
Collapse
|
19
|
Daly JL, Danson CM, Lewis PA, Zhao L, Riccardo S, Di Filippo L, Cacchiarelli D, Lee D, Cross SJ, Heesom KJ, Xiong WC, Ballabio A, Edgar JR, Cullen PJ. Multi-omic approach characterises the neuroprotective role of retromer in regulating lysosomal health. Nat Commun 2023; 14:3086. [PMID: 37248224 PMCID: PMC10227043 DOI: 10.1038/s41467-023-38719-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/05/2023] [Indexed: 05/31/2023] Open
Abstract
Retromer controls cellular homeostasis through regulating integral membrane protein sorting and transport and by controlling maturation of the endo-lysosomal network. Retromer dysfunction, which is linked to neurodegenerative disorders including Parkinson's and Alzheimer's diseases, manifests in complex cellular phenotypes, though the precise nature of this dysfunction, and its relation to neurodegeneration, remain unclear. Here, we perform an integrated multi-omics approach to provide precise insight into the impact of Retromer dysfunction on endo-lysosomal health and homeostasis within a human neuroglioma cell model. We quantify widespread changes to the lysosomal proteome, indicative of broad lysosomal dysfunction and inefficient autophagic lysosome reformation, coupled with a reconfigured cell surface proteome and secretome reflective of increased lysosomal exocytosis. Through this global proteomic approach and parallel transcriptomic analysis, we provide a holistic view of Retromer function in regulating lysosomal homeostasis and emphasise its role in neuroprotection.
Collapse
Affiliation(s)
- James L Daly
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, BS8 1TD, UK.
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy's Hospital, King's College London, SE1 9RT, London, UK.
| | - Chris M Danson
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Philip A Lewis
- Bristol Proteomics Facility, School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Lu Zhao
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Sara Riccardo
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Lucio Di Filippo
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- School for Advanced Studies, University of Naples "Federico II", Naples, Italy
| | - Daehoon Lee
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Stephen J Cross
- Wolfson Bioimaging Facility, Faculty of Biomedical Sciences, University of Bristol, Bristol, UK
| | - Kate J Heesom
- Bristol Proteomics Facility, School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Wen-Cheng Xiong
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- School for Advanced Studies, University of Naples "Federico II", Naples, Italy
- Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - James R Edgar
- Department of Pathology, Cambridge University, Tennis Court Road, Cambridge, UK
| | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
20
|
Lau V, Ramer L, Tremblay MÈ. An aging, pathology burden, and glial senescence build-up hypothesis for late onset Alzheimer's disease. Nat Commun 2023; 14:1670. [PMID: 36966157 PMCID: PMC10039917 DOI: 10.1038/s41467-023-37304-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 03/07/2023] [Indexed: 03/27/2023] Open
Abstract
Alzheimer's disease (AD) predominantly occurs as a late onset (LOAD) form involving neurodegeneration and cognitive decline with progressive memory loss. Risk factors that include aging promote accumulation of AD pathologies, such as amyloid-beta and tau aggregates, as well as inflammation and oxidative stress. Homeostatic glial states regulate and suppress pathology buildup; inflammatory states exacerbate pathology by releasing pro-inflammatory cytokines. Multiple stresses likely induce glial senescence, which could decrease supportive functions and reinforce inflammation. In this perspective, we hypothesize that aging first drives AD pathology burden, whereafter AD pathology putatively induces glial senescence in LOAD. We hypothesize that increasing glial senescence, particularly local senescent microglia accumulation, sustains and drives perpetuating buildup and spread of AD pathologies, glial aging, and further senescence. We predict that increasing glial senescence, particularly local senescent microglia accumulation, also transitions individuals from healthy cognition into mild cognitive impairment and LOAD diagnosis. These pathophysiological underpinnings may centrally contribute to LOAD onset, but require further mechanistic investigation.
Collapse
Affiliation(s)
- Victor Lau
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Leanne Ramer
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, BC, Canada.
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada.
- The Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| |
Collapse
|
21
|
Erdinger S, Amrein I, Back M, Ludewig S, Korte M, von Engelhardt J, Wolfer DP, Müller UC. Lack of APLP1 leads to subtle alterations in neuronal morphology but does not affect learning and memory. Front Mol Neurosci 2022; 15:1028836. [DOI: 10.3389/fnmol.2022.1028836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
The amyloid precursor protein APP plays a crucial role in Alzheimer pathogenesis. Its physiological functions, however, are only beginning to be unraveled. APP belongs to a small gene family, including besides APP the closely related amyloid precursor-like proteins APLP1 and APLP2, that all constitute synaptic adhesion proteins. While APP and APLP2 are ubiquitously expressed, APLP1 is specific for the nervous system. Previous genetic studies, including combined knockouts of several family members, pointed towards a unique role for APLP1, as only APP/APLP1 double knockouts were viable. We now examined brain and neuronal morphology in APLP1 single knockout (KO) animals, that have to date not been studied in detail. Here, we report that APLP1-KO mice show normal spine density in hippocampal CA1 pyramidal cells and subtle alterations in dendritic complexity. Extracellular field recordings revealed normal basal synaptic transmission and no alterations in synaptic plasticity (LTP). Further, behavioral studies revealed in APLP1-KO mice a small deficit in motor function and reduced diurnal locomotor activity, while learning and memory were not affected by the loss of APLP1. In summary, our study indicates that APP family members serve both distinct and overlapping functions that need to be considered for therapeutic treatments of Alzheimer’s disease.
Collapse
|
22
|
Li H, Wei M, Ye T, Liu Y, Qi D, Cheng X. Identification of the molecular subgroups in Alzheimer's disease by transcriptomic data. Front Neurol 2022; 13:901179. [PMID: 36204002 PMCID: PMC9530954 DOI: 10.3389/fneur.2022.901179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAlzheimer's disease (AD) is a heterogeneous pathological disease with genetic background accompanied by aging. This inconsistency is present among molecular subtypes, which has led to diagnostic ambiguity and failure in drug development. We precisely distinguished patients of AD at the transcriptome level.MethodsWe collected 1,240 AD brain tissue samples collected from the GEO dataset. Consensus clustering was used to identify molecular subtypes, and the clinical characteristics were focused on. To reveal transcriptome differences among subgroups, we certificated specific upregulated genes and annotated the biological function. According to RANK METRIC SCORE in GSEA, TOP10 was defined as the hub gene. In addition, the systematic correlation between the hub gene and “A/T/N” was analyzed. Finally, we used external data sets to verify the diagnostic value of hub genes.ResultsWe identified three molecular subtypes of AD from 743 AD samples, among which subtypes I and III had high-risk factors, and subtype II had protective factors. All three subgroups had higher neuritis plaque density, and subgroups I and III had higher clinical dementia scores and neurofibrillary tangles than subgroup II. Our results confirmed a positive association between neurofibrillary tangles and dementia, but not neuritis plaques. Subgroup I genes clustered in viral infection, hypoxia injury, and angiogenesis. Subgroup II showed heterogeneity in synaptic pathology, and we found several essential beneficial synaptic proteins. Due to presenilin one amplification, Subgroup III was a risk subgroup suspected of familial AD, involving abnormal neurogenic signals, glial cell differentiation, and proliferation. Among the three subgroups, the highest combined diagnostic value of the hub genes were 0.95, 0.92, and 0.83, respectively, indicating that the hub genes had sound typing and diagnostic ability.ConclusionThe transcriptome classification of AD cases played out the pathological heterogeneity of different subgroups. It throws daylight on the personalized diagnosis and treatment of AD.
Collapse
Affiliation(s)
- He Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Meiqi Wei
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyuan Ye
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiduan Liu
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongmei Qi
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaorui Cheng
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Xiaorui Cheng
| |
Collapse
|
23
|
A genetically modified minipig model for Alzheimer’s disease with SORL1 haploinsufficiency. Cell Rep Med 2022; 3:100740. [PMID: 36099918 PMCID: PMC9512670 DOI: 10.1016/j.xcrm.2022.100740] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/20/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022]
Abstract
The established causal genes in Alzheimer’s disease (AD), APP, PSEN1, and PSEN2, are functionally characterized using biomarkers, capturing an in vivo profile reflecting the disease’s initial preclinical phase. Mutations in SORL1, encoding the endosome recycling receptor SORLA, are found in 2%–3% of individuals with early-onset AD, and SORL1 haploinsufficiency appears to be causal for AD. To test whether SORL1 can function as an AD causal gene, we use CRISPR-Cas9-based gene editing to develop a model of SORL1 haploinsufficiency in Göttingen minipigs, taking advantage of porcine models for biomarker investigations. SORL1 haploinsufficiency in young adult minipigs is found to phenocopy the preclinical in vivo profile of AD observed with APP, PSEN1, and PSEN2, resulting in elevated levels of β-amyloid (Aβ) and tau preceding amyloid plaque formation and neurodegeneration, as observed in humans. Our study provides functional support for the theory that SORL1 haploinsufficiency leads to endosome cytopathology with biofluid hallmarks of autosomal dominant AD. Minipig model of Alzheimer’s disease by CRISPR knockout of the causal gene SORL1 Young SORL1 het minipigs phenocopy a preclinical CSF biomarker profile of individuals with AD SORL1 haploinsufficiency causes enlarged endosomes similar to neuronal AD pathology A minipig model bridging the translational gap between AD mouse models and affected individuals
Collapse
|
24
|
Asadzadeh J, Ruchti E, Jiao W, Limoni G, MacLachlan C, Small SA, Knott G, Santa-Maria I, McCabe BD. Retromer deficiency in Tauopathy models enhances the truncation and toxicity of Tau. Nat Commun 2022; 13:5049. [PMID: 36030267 PMCID: PMC9420134 DOI: 10.1038/s41467-022-32683-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Alteration of the levels, localization or post-translational processing of the microtubule associated protein Tau is associated with many neurodegenerative disorders. Here we develop adult-onset models for human Tau (hTau) toxicity in Drosophila that enable age-dependent quantitative measurement of central nervous system synapse loss and axonal degeneration, in addition to effects upon lifespan, to facilitate evaluation of factors that may contribute to Tau-dependent neurodegeneration. Using these models, we interrogate the interaction of hTau with the retromer complex, an evolutionarily conserved cargo-sorting protein assembly, whose reduced activity has been associated with both Parkinson’s and late onset Alzheimer’s disease. We reveal that reduction of retromer activity induces a potent enhancement of hTau toxicity upon synapse loss, axon retraction and lifespan through a specific increase in the production of a C-terminal truncated isoform of hTau. Our data establish a molecular and subcellular mechanism necessary and sufficient for the depletion of retromer activity to exacerbate Tau-dependent neurodegeneration. Tau and the Retromer complex are both linked to Parkinson’s and Alzheimer’s disease. Using Drosophila neurodegeneration models, this study finds that low retromer activity induces a specific increase of a highly toxic truncated form of human Tau.
Collapse
Affiliation(s)
- Jamshid Asadzadeh
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Greta Limoni
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Catherine MacLachlan
- BioEM Facility, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Scott A Small
- Department of Neurology, Columbia University, New York, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
| | - Graham Knott
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.,BioEM Facility, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA.,Department of Pathology & Cell Biology, Columbia University, New York, USA.,Facultad Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.
| |
Collapse
|
25
|
Pérez-Torres EJ, Utkina-Sosunova I, Mishra V, Barbuti P, De Planell-Saguer M, Dermentzaki G, Geiger H, Basile AO, Robine N, Fagegaltier D, Politi KA, Rinchetti P, Jackson-Lewis V, NYGC ALS Consortium, Harms M, Phatnani H, Lotti F, Przedborski S. Retromer dysfunction in amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A 2022; 119:e2118755119. [PMID: 35749364 PMCID: PMC9245686 DOI: 10.1073/pnas.2118755119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/03/2022] [Indexed: 12/26/2022] Open
Abstract
Retromer is a heteropentameric complex that plays a specialized role in endosomal protein sorting and trafficking. Here, we report a reduction in the retromer proteins-vacuolar protein sorting 35 (VPS35), VPS26A, and VPS29-in patients with amyotrophic lateral sclerosis (ALS) and in the ALS model provided by transgenic (Tg) mice expressing the mutant superoxide dismutase-1 G93A. These changes are accompanied by a reduction of levels of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit GluA1, a proxy of retromer function, in spinal cords from Tg SOD1G93A mice. Correction of the retromer deficit by a viral vector expressing VPS35 exacerbates the paralytic phenotype in Tg SOD1G93A mice. Conversely, lowering Vps35 levels in Tg SOD1G93A mice ameliorates the disease phenotype. In light of these findings, we propose that mild alterations in retromer inversely modulate neurodegeneration propensity in ALS.
Collapse
Affiliation(s)
- Eduardo J. Pérez-Torres
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
| | - Irina Utkina-Sosunova
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
| | - Vartika Mishra
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
| | - Peter Barbuti
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
| | - Mariangels De Planell-Saguer
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
| | - Georgia Dermentzaki
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
| | - Heather Geiger
- Computational Biology, New York Genome Center, New York, NY 10013
| | - Anna O. Basile
- Computational Biology, New York Genome Center, New York, NY 10013
| | - Nicolas Robine
- Computational Biology, New York Genome Center, New York, NY 10013
| | - Delphine Fagegaltier
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013
| | - Kristin A. Politi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
| | - Paola Rinchetti
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
| | - Vernice Jackson-Lewis
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
| | | | - Matthew Harms
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY 10032
| | - Hemali Phatnani
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013
| | - Francesco Lotti
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
| | - Serge Przedborski
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neuroscience, Columbia University, New York, NY 10027
| |
Collapse
|
26
|
Yoshida S, Hasegawa T. Beware of Misdelivery: Multifaceted Role of Retromer Transport in Neurodegenerative Diseases. Front Aging Neurosci 2022; 14:897688. [PMID: 35601613 PMCID: PMC9120357 DOI: 10.3389/fnagi.2022.897688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Retromer is a highly integrated multimeric protein complex that mediates retrograde cargo sorting from endosomal compartments. In concert with its accessory proteins, the retromer drives packaged cargoes to tubular and vesicular structures, thereby transferring them to the trans-Golgi network or to the plasma membrane. In addition to the endosomal trafficking, the retromer machinery participates in mitochondrial dynamics and autophagic processes and thus contributes to cellular homeostasis. The retromer components and their associated molecules are expressed in different types of cells including neurons and glial cells, and accumulating evidence from genetic and biochemical studies suggests that retromer dysfunction is profoundly involved in the pathogenesis of neurodegenerative diseases including Alzheimer’s Disease and Parkinson’s disease. Moreover, targeting retromer components could alleviate the neurodegenerative process, suggesting that the retromer complex may serve as a promising therapeutic target. In this review, we will provide the latest insight into the regulatory mechanisms of retromer and discuss how its dysfunction influences the pathological process leading to neurodegeneration.
Collapse
Affiliation(s)
- Shun Yoshida
- Division of Neurology, Department of Neuroscience and Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurology, National Hospital Organization Yonezawa Hospital, Yonezawa, Japan
| | - Takafumi Hasegawa
- Division of Neurology, Department of Neuroscience and Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Japan
- *Correspondence: Takafumi Hasegawa,
| |
Collapse
|
27
|
Mishra S, Knupp A, Szabo MP, Williams CA, Kinoshita C, Hailey DW, Wang Y, Andersen OM, Young JE. The Alzheimer's gene SORL1 is a regulator of endosomal traffic and recycling in human neurons. Cell Mol Life Sci 2022; 79:162. [PMID: 35226190 PMCID: PMC8885486 DOI: 10.1007/s00018-022-04182-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Loss of the Sortilin-related receptor 1 (SORL1) gene seems to act as a causal event for Alzheimer's disease (AD). Recent studies have established that loss of SORL1, as well as mutations in autosomal dominant AD genes APP and PSEN1/2, pathogenically converge by swelling early endosomes, AD's cytopathological hallmark. Acting together with the retromer trafficking complex, SORL1 has been shown to regulate the recycling of the amyloid precursor protein (APP) out of the endosome, contributing to endosomal swelling and to APP misprocessing. We hypothesized that SORL1 plays a broader role in neuronal endosomal recycling and used human induced pluripotent stem cell-derived neurons (hiPSC-Ns) to test this hypothesis. We examined endosomal recycling of three transmembrane proteins linked to AD pathophysiology: APP, the BDNF receptor Tropomyosin-related kinase B (TRKB), and the glutamate receptor subunit AMPA1 (GLUA1). METHODS We used isogenic hiPSCs engineered to have SORL1 depleted or to have enhanced SORL1 expression. We differentiated neurons from these cell lines and mapped the trafficking of APP, TRKB and GLUA1 within the endosomal network using confocal microscopy. We also performed cell surface recycling and lysosomal degradation assays to assess the functionality of the endosomal network in both SORL1-depleted and -overexpressing neurons. The functional impact of GLUA1 recycling was determined by measuring synaptic activity. Finally, we analyzed alterations in gene expression in SORL1-depleted neurons using RNA sequencing. RESULTS We find that as with APP, endosomal trafficking of GLUA1 and TRKB is impaired by loss of SORL1. We show that trafficking of all three cargoes to late endosomes and lysosomes is affected by manipulating SORL1 expression. We also show that depletion of SORL1 significantly impacts the endosomal recycling pathway for APP and GLUA1 at the level of the recycling endosome and trafficking to the cell surface. This has a functional effect on neuronal activity as shown by multi-electrode array (MEA). Conversely, increased SORL1 expression enhances endosomal recycling for APP and GLUA1. Our unbiased transcriptomic data further support SORL1's role in endosomal recycling. We observe altered expression networks that regulate cell surface trafficking and neurotrophic signaling in SORL1-depleted neurons. CONCLUSION Collectively, and together with other recent observations, these findings suggest that one role for SORL1 is to contribute to endosomal degradation and recycling pathways in neurons, a conclusion that has both pathogenic and therapeutic implications for Alzheimer's disease.
Collapse
Affiliation(s)
- Swati Mishra
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
| | - Allison Knupp
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
| | - Marcell P. Szabo
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
| | - Charles A. Williams
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
| | - Dale W. Hailey
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA 98195 USA
| | - Olav M. Andersen
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
| |
Collapse
|
28
|
Qureshi YH, Berman DE, Marsh SE, Klein RL, Patel VM, Simoes S, Kannan S, Petsko GA, Stevens B, Small SA. The neuronal retromer can regulate both neuronal and microglial phenotypes of Alzheimer's disease. Cell Rep 2022; 38:110262. [PMID: 35045281 PMCID: PMC8830374 DOI: 10.1016/j.celrep.2021.110262] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/14/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023] Open
Abstract
Disruption of retromer-dependent endosomal trafficking is considered pathogenic in late-onset Alzheimer's disease (AD). Here, to investigate this disruption in the intact brain, we turn to a genetic mouse model where the retromer core protein VPS35 is depleted in hippocampal neurons, and then we replete VPS35 using an optimized viral vector protocol. The VPS35 depletion-repletion studies strengthen the causal link between the neuronal retromer and AD-associated neuronal phenotypes, including the acceleration of amyloid precursor protein cleavage and the loss of synaptic glutamate receptors. Moreover, the studies show that the neuronal retromer can regulate a distinct, dystrophic, microglia morphology, phenotypic of hippocampal microglia in AD. Finally, the neuronal and, in part, the microglia responses to VPS35 depletion were found to occur independent of tau. Showing that the neuronal retromer can regulate AD-associated pathologies in two of AD's principal cell types strengthens the link, and clarifies the mechanism, between endosomal trafficking and late-onset sporadic AD.
Collapse
Affiliation(s)
- Yasir H Qureshi
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Diego E Berman
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Samuel E Marsh
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ronald L Klein
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Vivek M Patel
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Sabrina Simoes
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Suvarnambiga Kannan
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Gregory A Petsko
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Beth Stevens
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| | - Scott A Small
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| |
Collapse
|
29
|
Wang B, Sun X, Wang J, Deng X, Lin Y, Liu F, Dong R, Lin X, Bi Y. Potential Value of Cerebrospinal Fluid Progranulin in the Identification of Postoperative Delirium in Geriatrics Patients Undergoing Knee Replacement: The Perioperative Neurocognitive Disorder and Biomarker LifestylE Study. Front Aging Neurosci 2022; 13:772795. [PMID: 35069175 PMCID: PMC8770335 DOI: 10.3389/fnagi.2021.772795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Objective: The aim of this study was to investigate whether progranulin (PGRN) levels in cerebrospinal fluid (CSF) were associated with postoperative delirium (POD) in geriatric patients undergoing knee replacement. Method: A total of 600 Han Chinese patients aged 65–90 years and who underwent unilateral total knee arthroplasty were included in the Perioperative Neurocognitive Disorder And Biomarker LifestylE (PNDABLE) study from June 2020 to November 2020. All participants were assessed using the Confusion Assessment Method and the Memorial Delirium Assessment Scale on postoperative days 1–7 (or before discharge) by an anesthesiologist. CSF PGRN and CSF biomarkers of POD were measured by ELISA. We analyzed the risk and protective factors of POD using the multivariate logistic regression, and the associations between CSF PGRN and CSF biomarkers of POD using multiple linear regression. We also explored whether the influence of CSF PGRN on POD was mediated by POD core pathology in linear regression models. Results: Postoperative delirium incidence was 9.7% (53/545). There were significant differences in preoperative CSF PGRN between patients with POD and non-POD (NPOD). As for CSF biomarkers, CSF Aβ40, T-tau, and P-tau were risk factors for POD, while CSF PGRN, Aβ42, and Aβ42/Aβ40 were protective factors for POD, as shown by the multivariate logistic regression analysis. CSF PGRN was positively associated with CSF Aβ42 and was negatively associated with CSF Aβ40, T-tau, and P-tau in patients with POD. We found that the AUC was 0.795 (95% CI = 0.706, 0.867) for PGRN between POD and NPOD groups. We found the influence of CSF PGRN on POD was mediated by POD core pathology. The effect was considered partial mediation with the proportion of mediation varying from 44.92 to 62.07%. Conclusion: Cerebrospinal fluid PGRN may be a reasonably good prognostic factor for POD development. Overall, amyloid pathology and tau protein might partially mediate the influence of PGRN on POD. Clinical Trial Registration:www.clinicaltrials.gov, identifier ChiCTR2000033439.
Collapse
Affiliation(s)
- Bin Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiujie Sun
- Department of Nursing, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Jiahan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiyuan Deng
- Department of Anesthesiology, Dalian Medical University, Dalian, China
| | - Yanan Lin
- Department of Anesthesiology, Weifang Medical University, Weifang, China
| | - Fanghao Liu
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Rui Dong
- Department of Anesthesiology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Xu Lin
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Yanlin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
- *Correspondence: Yanlin Bi,
| |
Collapse
|
30
|
Deng Z, Dong Y, Zhou X, Lu JH, Yue Z. Pharmacological modulation of autophagy for Alzheimer’s disease therapy: Opportunities and obstacles. Acta Pharm Sin B 2021; 12:1688-1706. [PMID: 35847516 PMCID: PMC9279633 DOI: 10.1016/j.apsb.2021.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent and deleterious neurodegenerative disorder characterized by an irreversible and progressive impairment of cognitive abilities as well as the formation of amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. By far, the precise mechanisms of AD are not fully understood and no interventions are available to effectively slow down progression of the disease. Autophagy is a conserved degradation pathway that is crucial to maintain cellular homeostasis by targeting damaged organelles, pathogens, and disease-prone protein aggregates to lysosome for degradation. Emerging evidence suggests dysfunctional autophagy clearance pathway as a potential cellular mechanism underlying the pathogenesis of AD in affected neurons. Here we summarize the current evidence for autophagy dysfunction in the pathophysiology of AD and discuss the role of autophagy in the regulation of AD-related protein degradation and neuroinflammation in neurons and glial cells. Finally, we review the autophagy modulators reported in the treatment of AD models and discuss the obstacles and opportunities for potential clinical application of the novel autophagy activators for AD therapy.
Collapse
Affiliation(s)
- Zhiqiang Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Yu Dong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Xiaoting Zhou
- Department of Neurology, the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
- Corresponding authors.
| | - Zhenyu Yue
- Department of Neurology, the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Corresponding authors.
| |
Collapse
|
31
|
Simoes S, Guo J, Buitrago L, Qureshi YH, Feng X, Kothiya M, Cortes E, Patel V, Kannan S, Kim YH, Chang KT, Hussaini SA, Moreno H, Di Paolo G, Andersen OM, Small SA. Alzheimer's vulnerable brain region relies on a distinct retromer core dedicated to endosomal recycling. Cell Rep 2021; 37:110182. [PMID: 34965419 PMCID: PMC8792909 DOI: 10.1016/j.celrep.2021.110182] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 11/15/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
Whether and how the pathogenic disruptions in endosomal trafficking observed in Alzheimer’s disease (AD) are linked to its anatomical vulnerability remain unknown. Here, we began addressing these questions by showing that neurons are enriched with a second retromer core, organized around VPS26b, differentially dedicated to endosomal recycling. Next, by imaging mouse models, we show that the trans-entorhinal cortex, a region most vulnerable to AD, is most susceptible to VPS26b depletion—a finding validated by electrophysiology, immunocytochemistry, and behavior. VPS26b was then found enriched in the trans-entorhinal cortex of human brains, where both VPS26b and the retromer-related receptor SORL1 were found deficient in AD. Finally, by regulating glutamate receptor and SORL1 recycling, we show that VPS26b can mediate regionally selective synaptic dysfunction and SORL1 deficiency. Together with the trans-entorhinal’s unique network properties, hypothesized to impose a heavy demand on endosomal recycling, these results suggest a general mechanism that can explain AD’s regional vulnerability. Trans-entorhinal cortex neurons are most vulnerable to Alzheimer’s disease. Simoes et al. explain this vulnerability by showing that these neurons are dependent on a distinct VPS26b-retromer core differentially dedicated to endosomal recycling. VPS26b is highly expressed in these neurons, where they regulate synaptic function, GluA1/SORL1 recycling, and disease-associated pathologies
Collapse
Affiliation(s)
- Sabrina Simoes
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA.
| | - Jia Guo
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Luna Buitrago
- The Robert F. Furchgott Center for Neural and Behavioral Science, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Yasir H Qureshi
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Xinyang Feng
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Milankumar Kothiya
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Etty Cortes
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Vivek Patel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Suvarnambiga Kannan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, South Korea
| | - Kyu-Tae Chang
- National Primate Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, South Korea
| | - S Abid Hussaini
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA; Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Herman Moreno
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Gilbert Di Paolo
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA; Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høgh-Guldbergs Gade 10, 8000 AarhusC, Denmark
| | - Scott A Small
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
32
|
Yan Y, Aierken A, Wang C, Song D, Ni J, Wang Z, Quan Z, Qing H. A potential biomarker of preclinical Alzheimer's disease: The olfactory dysfunction and its pathogenesis-based neural circuitry impairments. Neurosci Biobehav Rev 2021; 132:857-869. [PMID: 34810025 DOI: 10.1016/j.neubiorev.2021.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/26/2021] [Accepted: 11/07/2021] [Indexed: 01/24/2023]
Abstract
The olfactory dysfunction can signal and act as a potential biomarker of preclinical AD. However, the precise regulatory mechanism of olfactory function on the neural pathogenesis of AD is still unclear. The impairment of neural networks in olfaction system has been shown to be tightly associated with AD. As key brain regions of the olfactory system, the olfactory bulb (OB) and the piriform cortex (PCx) have a profound influence on the olfactory function. Therefore, this review will explore the mechanism of olfactory dysfunction in preclinical AD in the perspective of abnormal neural networks in the OB and PCx and their associated brain regions, especially from two aspects of aberrant oscillations and synaptic plasticity damages, which help better understand the underlying mechanism of olfactory neural network damages related to AD.
Collapse
Affiliation(s)
- Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Ailikemu Aierken
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Chunjian Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhe Wang
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
33
|
Yu Y, Gao Y, Winblad B, Tjernberg LO, Schedin-Weiss S. A Super-Resolved View of the Alzheimer's Disease-Related Amyloidogenic Pathway in Hippocampal Neurons. J Alzheimers Dis 2021; 83:833-852. [PMID: 34366358 PMCID: PMC8543249 DOI: 10.3233/jad-215008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Processing of the amyloid-β protein precursor (AβPP) is neurophysiologically important due to the resulting fragments that regulate synapse biology, as well as potentially harmful due to generation of the 42 amino acid long amyloid β-peptide (Aβ42), which is a key player in Alzheimer's disease. OBJECTIVE Our aim was to clarify the subcellular locations of the fragments involved in the amyloidogenic pathway in primary neurons with a focus on Aβ42 and its immediate substrate AβPP C-terminal fragment (APP-CTF). To overcome the difficulties of resolving these compartments due to their small size, we used super-resolution microscopy. METHODS Mouse primary hippocampal neurons were immunolabelled and imaged by stimulated emission depletion (STED) microscopy, including three-dimensional three-channel imaging, and quantitative image analyses. RESULTS The first (β-secretase) and second (γ-secretase) cleavages of AβPP were localized to functionally and distally distinct compartments. The β-secretase cleavage was observed in early endosomes in soma, where we were able to show that the liberated N- and C-terminal fragments were sorted into distinct vesicles budding from the early endosomes. Lack of colocalization of Aβ42 and APP-CTF in soma suggested that γ-secretase cleavage occurs in neurites. Indeed, APP-CTF was, in line with Aβ42 in our previous study, enriched in the presynapse but absent from the postsynapse. In contrast, full-length AβPP was not detected in either the pre- or the postsynaptic side of the synapse. Furthermore, we observed that endogenously produced and endocytosed Aβ42 were localized in different compartments. CONCLUSION These findings provide critical super-resolved insight into amyloidogenic AβPP processing in primary neurons.
Collapse
Affiliation(s)
- Yang Yu
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Yang Gao
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
34
|
Small SA, Petsko GA. Endosomal recycling reconciles the Alzheimer's disease paradox. Sci Transl Med 2021; 12:12/572/eabb1717. [PMID: 33268506 DOI: 10.1126/scitranslmed.abb1717] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022]
Abstract
A hub-and-spoke model with endosomal recycling as the hub can reconcile the pathogenic contribution of amyloid precursor protein to Alzheimer's disease.
Collapse
Affiliation(s)
- Scott A Small
- Alzheimer's Disease Research Center, Department of Neurology, Columbia University, New York, NY 10032, USA.
| | - Gregory A Petsko
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
35
|
Kang YJ, Diep YN, Tran M, Cho H. Therapeutic Targeting Strategies for Early- to Late-Staged Alzheimer's Disease. Int J Mol Sci 2020; 21:E9591. [PMID: 33339351 PMCID: PMC7766709 DOI: 10.3390/ijms21249591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, typically showing progressive neurodegeneration in aging brains. The key signatures of the AD progression are the deposition of amyloid-beta (Aβ) peptides, the formation of tau tangles, and the induction of detrimental neuroinflammation leading to neuronal loss. However, conventional pharmacotherapeutic options are merely relying on the alleviation of symptoms that are limited to mild to moderate AD patients. Moreover, some of these medicines discontinued to use due to either the insignificant effectiveness in improving the cognitive impairment or the adverse side effects worsening essential bodily functions. One of the reasons for the failure is the lack of knowledge on the underlying mechanisms that can accurately explain the major causes of the AD progression correlating to the severity of AD. Therefore, there is an urgent need for the better understanding of AD pathogenesis and the development of the disease-modifying treatments, particularly for severe and late-onset AD, which have not been covered thoroughly. Here, we review the underlying mechanisms of AD progression, which have been employed for the currently established therapeutic strategies. We believe this will further spur the discovery of a novel disease-modifying treatment for mild to severe, as well as early- to late-onset, AD.
Collapse
Affiliation(s)
- You Jung Kang
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, University of North Carolina, Charlotte, NC 28223, USA;
- Department of Biological Sciences, Center for Biomedical Engineering and Science, University of North Carolina, Charlotte, NC 28223, USA
| | - Yen N. Diep
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea; (Y.N.D.); (M.T.)
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea; (Y.N.D.); (M.T.)
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea; (Y.N.D.); (M.T.)
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea
| |
Collapse
|