1
|
Amano A, Sanjo N, Araki W, Anraku Y, Nakakido M, Matsubara E, Tomiyama T, Nagata T, Tsumoto K, Kataoka K, Yokota T. Peripheral administration of nanomicelle-encapsulated anti-Aβ oligomer fragment antibody reduces various toxic Aβ species in the brain. J Nanobiotechnology 2023; 21:36. [PMID: 36721182 PMCID: PMC9888736 DOI: 10.1186/s12951-023-01772-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/07/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Although a large amount of evidence has revealed that amyloid β (Aβ), especially Aβ oligomers, protofibrils, and pyroglutamated Aβs, participate primarily in the pathophysiological processes of Alzheimer's disease, most clinical trials of anti-Aβ antibody therapy have never acquired successful efficacy in human clinical trials, partly because peripheral administration of antibody medications was unable to deliver sufficient amounts of the molecules to the brain. Recently, we developed polymeric nanomicelles capable of passing through the blood-brain barrier that function as chaperones to deliver larger amounts of heavy molecules to the brain. Herein, we aimed to evaluate the efficacy of newly developed antibody 6H4 fragments specific to Aβ oligomers encapsulated in polymeric nanomicelles on the development of Alzheimer's disease pathology in Alzheimer's disease model mice at the age of emergence of early Alzheimer's disease pathology. RESULTS During the 10-week administration of 6H4 antibody fragments in polymeric nanomicelles, a significant reduction in the amounts of various toxic Aβ species, such as Aβ oligomers, toxic Aβ conformers, and pyroglutamated Aβs in the brain was observed. In addition, immunohistochemistry indicated inhibition of diameters of Aβ plaques, Aβ-antibody immunoreactive areas, and also plaque core formation. Behavioral analysis of the mice model revealed that the 6H4 fragments-polymeric nanomicelle group was significantly better at maintaining long-term spatial reference memory in the probe and platform tests of the water maze, thereby indicating inhibition of the pathophysiological process of Alzheimer's disease. CONCLUSIONS The results indicated that the strategy of reducing toxic Aβ species in early dementia owing to Alzheimer's disease by providing sufficient antibodies in the brain may modify Alzheimer's disease progression.
Collapse
Affiliation(s)
- Akiko Amano
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Nobuo Sanjo
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Wataru Araki
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Yasutaka Anraku
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.493442.c0000 0004 5936 3316Innovation Center of Nano Medicine, Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| | - Makoto Nakakido
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Etsuro Matsubara
- grid.412334.30000 0001 0665 3553Department of Neurology, Oita University, Oita, Japan
| | - Takami Tomiyama
- grid.258799.80000 0004 0372 2033Department of Translational Neuroscience, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Tetsuya Nagata
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Kouhei Tsumoto
- grid.26999.3d0000 0001 2151 536XDepartment of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XDepartment of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XThe Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazunori Kataoka
- grid.493442.c0000 0004 5936 3316Innovation Center of Nano Medicine, Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| | - Takanori Yokota
- grid.265073.50000 0001 1014 9130Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima Bunkyo-ku, Tokyo, 113-8510 Japan
| |
Collapse
|
2
|
Salasova A, Monti G, Andersen OM, Nykjaer A. Finding memo: versatile interactions of the VPS10p-Domain receptors in Alzheimer’s disease. Mol Neurodegener 2022; 17:74. [PMID: 36397124 PMCID: PMC9673319 DOI: 10.1186/s13024-022-00576-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
The family of VPS10p-Domain (D) receptors comprises five members named SorLA, Sortilin, SorCS1, SorCS2 and SorCS3. While their physiological roles remain incompletely resolved, they have been recognized for their signaling engagements and trafficking abilities, navigating a number of molecules between endosome, Golgi compartments, and the cell surface. Strikingly, recent studies connected all the VPS10p-D receptors to Alzheimer’s disease (AD) development. In addition, they have been also associated with diseases comorbid with AD such as diabetes mellitus and major depressive disorder. This systematic review elaborates on genetic, functional, and mechanistic insights into how dysfunction in VPS10p-D receptors may contribute to AD etiology, AD onset diversity, and AD comorbidities. Starting with their functions in controlling cellular trafficking of amyloid precursor protein and the metabolism of the amyloid beta peptide, we present and exemplify how these receptors, despite being structurally similar, regulate various and distinct cellular events involved in AD. This includes a plethora of signaling crosstalks that impact on neuronal survival, neuronal wiring, neuronal polarity, and synaptic plasticity. Signaling activities of the VPS10p-D receptors are especially linked, but not limited to, the regulation of neuronal fitness and apoptosis via their physical interaction with pro- and mature neurotrophins and their receptors. By compiling the functional versatility of VPS10p-D receptors and their interactions with AD-related pathways, we aim to further propel the AD research towards VPS10p-D receptor family, knowledge that may lead to new diagnostic markers and therapeutic strategies for AD patients.
Collapse
|
3
|
Amyloidogenesis and Neurotrophic Dysfunction in Alzheimer’s Disease: Do They have a Common Regulating Pathway? Cells 2022; 11:cells11203201. [PMID: 36291068 PMCID: PMC9600014 DOI: 10.3390/cells11203201] [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: 09/12/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
The amyloid cascade hypothesis has predominately been used to describe the pathogenesis of Alzheimer’s disease (AD) for decades, as Aβ oligomers are thought to be the prime cause of AD. Meanwhile, the neurotrophic factor hypothesis has also been proposed for decades. Accumulating evidence states that the amyloidogenic process and neurotrophic dysfunction are mutually influenced and may coincidently cause the onset and progress of AD. Meanwhile, there are intracellular regulators participating both in the amyloidogenic process and neurotrophic pathways, which might be the common original causes of amyloidogenesis and neurotrophic dysfunction. In this review, the current understanding regarding the role of neurotrophic dysfunction and the amyloidogenic process in AD pathology is briefly summarized. The mutual influence of these two pathogenesis pathways and their potential common causal pathway are further discussed. Therapeutic strategies targeting the common pathways to simultaneously prevent amyloidogenesis and neurotrophic dysfunction might be anticipated for the disease-modifying treatment of AD.
Collapse
|
4
|
Zhang Y, Gao X, Bai X, Yao S, Chang YZ, Gao G. The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl Neurodegener 2022; 11:39. [PMID: 35996194 PMCID: PMC9395820 DOI: 10.1186/s40035-022-00313-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Furin is an important mammalian proprotein convertase that catalyzes the proteolytic maturation of a variety of prohormones and proproteins in the secretory pathway. In the brain, the substrates of furin include the proproteins of growth factors, receptors and enzymes. Emerging evidence, such as reduced FURIN mRNA expression in the brains of Alzheimer's disease patients or schizophrenia patients, has implicated a crucial role of furin in the pathophysiology of neurodegenerative and neuropsychiatric diseases. Currently, compared to cancer and infectious diseases, the aberrant expression of furin and its pharmaceutical potentials in neurological diseases remain poorly understood. In this article, we provide an overview on the physiological roles of furin and its substrates in the brain, summarize the deregulation of furin expression and its effects in neurodegenerative and neuropsychiatric disorders, and discuss the implications and current approaches that target furin for therapeutic interventions. This review may expedite future studies to clarify the molecular mechanisms of furin deregulation and involvement in the pathogenesis of neurodegenerative and neuropsychiatric diseases, and to develop new diagnosis and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xiaoqin Gao
- Shijiazhuang People's Hospital, Hebei Medical University, Shijiazhuang, 050027, China
| | - Xue Bai
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Shanshan Yao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
5
|
Mitok KA, Keller MP, Attie AD. Sorting through the extensive and confusing roles of sortilin in metabolic disease. J Lipid Res 2022; 63:100243. [PMID: 35724703 PMCID: PMC9356209 DOI: 10.1016/j.jlr.2022.100243] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 01/06/2023] Open
Abstract
Sortilin is a post-Golgi trafficking receptor homologous to the yeast vacuolar protein sorting receptor 10 (VPS10). The VPS10 motif on sortilin is a 10-bladed β-propeller structure capable of binding more than 50 proteins, covering a wide range of biological functions including lipid and lipoprotein metabolism, neuronal growth and death, inflammation, and lysosomal degradation. Sortilin has a complex cellular trafficking itinerary, where it functions as a receptor in the trans-Golgi network, endosomes, secretory vesicles, multivesicular bodies, and at the cell surface. In addition, sortilin is associated with hypercholesterolemia, Alzheimer's disease, prion diseases, Parkinson's disease, and inflammation syndromes. The 1p13.3 locus containing SORT1, the gene encoding sortilin, carries the strongest association with LDL-C of all loci in human genome-wide association studies. However, the mechanism by which sortilin influences LDL-C is unclear. Here, we review the role sortilin plays in cardiovascular and metabolic diseases and describe in detail the large and often contradictory literature on the role of sortilin in the regulation of LDL-C levels.
Collapse
Affiliation(s)
- Kelly A Mitok
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
6
|
Kawarabayashi T, Nakamura T, Sato K, Seino Y, Ichii S, Nakahata N, Takatama M, Westaway D, George-Hyslop PS, Shoji M. Lipid Rafts Act as a Common Platform for Amyloid-β Oligomer-Induced Alzheimer’s Disease Pathology. J Alzheimers Dis 2022; 87:1189-1203. [DOI: 10.3233/jad-215662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Amyloid-β (Aβ) oligomers induce the overproduction of phosphorylated tau and neurodegeneration. These cascades gradually cause cognitive impairment in Alzheimer’s disease (AD). While each pathological event in AD has been studied in detail separately, the spatial and temporal relationships between pathological events in AD remain unclear. Objective: We demonstrated that lipid rafts function as a common platform for the pathological cascades of AD. Methods: Cellular and synaptosomal lipid rafts were prepared from the brains of Aβ amyloid model mice (Tg2576 mice) and double transgenic mice (Tg2576 x TgTauP301L mice) and longitudinally analyzed. Results: Aβ dimers, the cellular prion protein (PrPc), and Aβ dimer/PrPc complexes were detected in the lipid rafts. The levels of Fyn, the phosphorylated NR2B subunit of the N-methyl-D-aspartate receptor, glycogen synthase kinase 3β, total tau, phosphorylated tau, and tau oligomers increased with Aβ dimer accumulation in both the cellular and synaptosomal lipid rafts. Increases in the levels of these molecules were first seen at 6 months of age and corresponded with the early stages of Aβ accumulation in the amyloid model mice. Conclusion: Lipid rafts act as a common platform for the progression of AD pathology. The findings of this study suggest a novel therapeutic approach to AD, involving the modification of lipid raft components and the inhibition of their roles in the sequential pathological events of AD.
Collapse
Affiliation(s)
- Takeshi Kawarabayashi
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Gunma, Japan
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Takumi Nakamura
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kaoru Sato
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yusuke Seino
- Department of Neurology, Hirosaki National Hospital, Hirosaki, Aomori, Japan
| | - Sadanobu Ichii
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Naoko Nakahata
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Speech and Hearing, Hirosaki University of Health and Welfare/JuniorCollege, Hirosaki, Aomori, Japan
| | - Masamitsu Takatama
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Gunma, Japan
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Peter St. George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases and Departments of Medicine, Medical Biophysics, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mikio Shoji
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Gunma, Japan
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
7
|
Eggert S, Kins S, Endres K, Brigadski T. Brothers in arms: proBDNF/BDNF and sAPPα/Aβ-signaling and their common interplay with ADAM10, TrkB, p75NTR, sortilin, and sorLA in the progression of Alzheimer's disease. Biol Chem 2022; 403:43-71. [PMID: 34619027 DOI: 10.1515/hsz-2021-0330] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an important modulator for a variety of functions in the central nervous system (CNS). A wealth of evidence, such as reduced mRNA and protein level in the brain, cerebrospinal fluid (CSF), and blood samples of Alzheimer's disease (AD) patients implicates a crucial role of BDNF in the progression of this disease. Especially, processing and subcellular localization of BDNF and its receptors TrkB and p75 are critical determinants for survival and death in neuronal cells. Similarly, the amyloid precursor protein (APP), a key player in Alzheimer's disease, and its cleavage fragments sAPPα and Aβ are known for their respective roles in neuroprotection and neuronal death. Common features of APP- and BDNF-signaling indicate a causal relationship in their mode of action. However, the interconnections of APP- and BDNF-signaling are not well understood. Therefore, we here discuss dimerization properties, localization, processing by α- and γ-secretase, relevance of the common interaction partners TrkB, p75, sorLA, and sortilin as well as shared signaling pathways of BDNF and sAPPα.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
8
|
Pathak A, Clark S, Bronfman FC, Deppmann CD, Carter BD. Long-distance regressive signaling in neural development and disease. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2021; 10:e382. [PMID: 32391977 PMCID: PMC7655682 DOI: 10.1002/wdev.382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
Nervous system development proceeds via well-orchestrated processes involving a balance between progressive and regressive events including stabilization or elimination of axons, synapses, and even entire neurons. These progressive and regressive events are driven by functionally antagonistic signaling pathways with the dominant pathway eventually determining whether a neural element is retained or removed. Many of these developmental sculpting events are triggered by final target innervation necessitating a long-distance mode of communication. While long-distance progressive signaling has been well characterized, particularly for neurotrophic factors, there remains relatively little known about how regressive events are triggered from a distance. Here we discuss the emergent phenomenon of long-distance regressive signaling pathways. In particular, we will cover (a) progressive and regressive cues known to be employed after target innervation, (b) the mechanisms of long-distance signaling from an endosomal platform, (c) recent evidence that long-distance regressive cues emanate from platforms like death receptors or repulsive axon guidance receptors, and (d) evidence that these pathways are exploited in pathological scenarios. This article is categorized under: Nervous System Development > Vertebrates: General Principles Signaling Pathways > Global Signaling Mechanisms Establishment of Spatial and Temporal Patterns > Cytoplasmic Localization.
Collapse
Affiliation(s)
- Amrita Pathak
- Department of Biochemistry and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Shayla Clark
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | - Francisca C. Bronfman
- Institute of Biomedical Sciences (ICB), Faculty of Medicine, Faculty of Life Science, Universidad Andres Bello, Santiago, Chile
| | - Christopher D. Deppmann
- Departments of Biology, Cell Biology, Biomedical Engineering, and Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Bruce D. Carter
- Department of Biochemistry and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
9
|
Forner S, Martini AC, Prieto GA, Dang CT, Rodriguez-Ortiz CJ, Reyes-Ruiz JM, Trujillo-Estrada L, da Cunha C, Andrews EJ, Phan J, Vu Ha J, Chang AVZD, Levites Y, Cruz PE, Ager R, Medeiros R, Kitazawa M, Glabe CG, Cotman CW, Golde T, Baglietto-Vargas D, LaFerla FM. Intra- and extracellular β-amyloid overexpression via adeno-associated virus-mediated gene transfer impairs memory and synaptic plasticity in the hippocampus. Sci Rep 2019; 9:15936. [PMID: 31685865 PMCID: PMC6828807 DOI: 10.1038/s41598-019-52324-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 10/11/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD), the most common age-related neurodegenerative disorder, is currently conceptualized as a disease of synaptic failure. Synaptic impairments are robust within the AD brain and better correlate with dementia severity when compared with other pathological features of the disease. Nevertheless, the series of events that promote synaptic failure still remain under debate, as potential triggers such as β-amyloid (Aβ) can vary in size, configuration and cellular location, challenging data interpretation in causation studies. Here we present data obtained using adeno-associated viral (AAV) constructs that drive the expression of oligomeric Aβ either intra or extracellularly. We observed that expression of Aβ in both cellular compartments affect learning and memory, reduce the number of synapses and the expression of synaptic-related proteins, and disrupt chemical long-term potentiation (cLTP). Together, these findings indicate that during the progression AD the early accumulation of Aβ inside neurons is sufficient to promote morphological and functional cellular toxicity, a phenomenon that can be exacerbated by the buildup of Aβ in the brain parenchyma. Moreover, our AAV constructs represent a valuable tool in the investigation of the pathological properties of Aβ oligomers both in vivo and in vitro.
Collapse
Affiliation(s)
- Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Alessandra C Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - G Aleph Prieto
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Cindy T Dang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | | | - Jorge Mauricio Reyes-Ruiz
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, 92697, USA
| | - Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Elizabeth J Andrews
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Jordan Vu Ha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Allissa V Z D Chang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Yona Levites
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Pedro E Cruz
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Rahasson Ager
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Rodrigo Medeiros
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Masashi Kitazawa
- Department of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Charles G Glabe
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, 92697, USA
| | - Carl W Cotman
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, USA
- Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Todd Golde
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, USA
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
10
|
Cline EN, Bicca MA, Viola KL, Klein WL. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis 2019; 64:S567-S610. [PMID: 29843241 PMCID: PMC6004937 DOI: 10.3233/jad-179941] [Citation(s) in RCA: 557] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid-β oligomer (AβO) hypothesis was introduced in 1998. It proposed that the brain damage leading to Alzheimer’s disease (AD) was instigated by soluble, ligand-like AβOs. This hypothesis was based on the discovery that fibril-free synthetic preparations of AβOs were potent CNS neurotoxins that rapidly inhibited long-term potentiation and, with time, caused selective nerve cell death (Lambert et al., 1998). The mechanism was attributed to disrupted signaling involving the tyrosine-protein kinase Fyn, mediated by an unknown toxin receptor. Over 4,000 articles concerning AβOs have been published since then, including more than 400 reviews. AβOs have been shown to accumulate in an AD-dependent manner in human and animal model brain tissue and, experimentally, to impair learning and memory and instigate major facets of AD neuropathology, including tau pathology, synapse deterioration and loss, inflammation, and oxidative damage. As reviewed by Hayden and Teplow in 2013, the AβO hypothesis “has all but supplanted the amyloid cascade.” Despite the emerging understanding of the role played by AβOs in AD pathogenesis, AβOs have not yet received the clinical attention given to amyloid plaques, which have been at the core of major attempts at therapeutics and diagnostics but are no longer regarded as the most pathogenic form of Aβ. However, if the momentum of AβO research continues, particularly efforts to elucidate key aspects of structure, a clear path to a successful disease modifying therapy can be envisioned. Ensuring that lessons learned from recent, late-stage clinical failures are applied appropriately throughout therapeutic development will further enable the likelihood of a successful therapy in the near-term.
Collapse
Affiliation(s)
- Erika N Cline
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Maíra Assunção Bicca
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
11
|
Abstract
Most common neurodegenerative diseases feature deposition of protein amyloids and degeneration of brain networks. Amyloids are ordered protein assemblies that can act as templates for their own replication through monomer addition. Evidence suggests that this characteristic may underlie the progression of pathology in neurodegenerative diseases. Many different amyloid proteins, including Aβ, tau, and α-synuclein, exhibit properties similar to those of infectious prion protein in experimental systems: discrete and self-replicating amyloid structures, transcellular propagation of aggregation, and transmissible neuropathology. This review discusses the contribution of prion phenomena and transcellular propagation to the progression of pathology in common neurodegenerative diseases such as Alzheimer's and Parkinson's. It reviews fundamental events such as cell entry, amplification, and transcellular movement. It also discusses amyloid strains, which produce distinct patterns of neuropathology and spread through the nervous system. These concepts may impact the development of new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Jaime Vaquer-Alicea
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| |
Collapse
|
12
|
Xu SY, Jiang J, Pan A, Yan C, Yan XX. Sortilin: a new player in dementia and Alzheimer-type neuropathology. Biochem Cell Biol 2018; 96:491-497. [PMID: 29687731 DOI: 10.1139/bcb-2018-0023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Age-related dementias are now a major mortality factor among most human populations in the world, with Alzheimer's disease (AD) being the leading dementia-causing neurodegenerative disease. The pathogenic mechanism underlying dementia disorders, and AD in particular, remained largely unknown. Efforts to develop drugs targeting the disease's hallmark lesions, such as amyloid plaque and tangle pathologies, have been unsuccessful so far. The vacuolar protein sorting 10p (Vps10p) family plays a critical role in membrane signal transduction and protein sorting and trafficking between intracellular compartments. Data emerging during the past few years point to an involvement of this family in the development of AD. Specifically, the Vps10p member sortilin has been shown to participate in amyloid plaque formation, tau phosphorylation, abnormal protein sorting and apoptosis. In this minireview, we update some latest findings from animal experiments and human brain studies suggesting that abnormal sortilin expression is associated with AD-type neuropathology, warranting further research that might lead to novel targets for the development of AD therapies.
Collapse
Affiliation(s)
- Shu-Yin Xu
- a Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Juan Jiang
- a Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Aihua Pan
- a Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Cai Yan
- a Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.,b Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Xiao-Xin Yan
- a Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
13
|
Andersson CH, Hansson O, Minthon L, Andreasen N, Blennow K, Zetterberg H, Skoog I, Wallin A, Nilsson S, Kettunen P. A Genetic Variant of the Sortilin 1 Gene is Associated with Reduced Risk of Alzheimer's Disease. J Alzheimers Dis 2018; 53:1353-63. [PMID: 27392867 PMCID: PMC5147507 DOI: 10.3233/jad-160319] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder represented by the accumulation of intracellular tau protein and extracellular deposits of amyloid-β (Aβ) in the brain. The gene sortilin 1 (SORT1) has previously been associated with cardiovascular disease in gene association studies. It has also been proposed to be involved in AD pathogenesis through facilitating Aβ clearance by binding apoE/Aβ complexes prior to cellular uptake. However, the neuropathological role of SORT1 in AD is not fully understood. To evaluate the associations between gene variants of SORT1 and risk of AD, we performed genetic analyses in a Swedish case-control cohort. Ten single nucleotide polymorphisms (SNPs), covering the whole SORT1 gene, were selected and genotyped in 620 AD patients and 1107 controls. The SNP rs17646665, located in a non-coding region of the SORT1 gene, remained significantly associated with decreased risk of AD after multiple testing (pc = 0.0061). In addition, other SNPs were found to be nominally associated with risk of AD, as well as altered cognitive function and the CSF biomarker Aβ42, but these associations did not survive correction for multiple testing. The fact that SORT1 has been strongly associated with risk of cardiovascular disease is intriguing as cardiovascular disease is also regarded as a risk factor for AD. Finally, increased knowledge about SORT1 function has a potential to increase our understanding of APOE, the strongest risk factor for AD.
Collapse
Affiliation(s)
- Carl-Henrik Andersson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Lennart Minthon
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niels Andreasen
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Ingmar Skoog
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Sweden
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Amengual J, Guo L, Strong A, Madrigal-Matute J, Wang H, Kaushik S, Brodsky JL, Rader DJ, Cuervo AM, Fisher EA. Autophagy Is Required for Sortilin-Mediated Degradation of Apolipoprotein B100. Circ Res 2018; 122:568-582. [PMID: 29301854 DOI: 10.1161/circresaha.117.311240] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 12/28/2017] [Accepted: 12/29/2017] [Indexed: 12/30/2022]
Abstract
RATIONALE Genome-wide association studies identified single-nucleotide polymorphisms near the SORT1 locus strongly associated with decreased plasma LDL-C (low-density lipoprotein cholesterol) levels and protection from atherosclerotic cardiovascular disease and myocardial infarction. The minor allele of the causal SORT1 single-nucleotide polymorphism locus creates a putative C/EBPα (CCAAT/enhancer-binding protein α)-binding site in the SORT1 promoter, thereby increasing in homozygotes sortilin expression by 12-fold in liver, which is rich in this transcription factor. Our previous studies in mice have showed reductions in plasma LDL-C and its principal protein component, apoB (apolipoprotein B) with increased SORT1 expression, and in vitro studies suggested that sortilin promoted the presecretory lysosomal degradation of apoB associated with the LDL precursor, VLDL (very-low-density lipoprotein). OBJECTIVE To determine directly that SORT1 overexpression results in apoB degradation and to identify the mechanisms by which this reduces apoB and VLDL secretion by the liver, thereby contributing to understanding the clinical phenotype of lower LDL-C levels. METHODS AND RESULTS Pulse-chase studies directly established that SORT1 overexpression results in apoB degradation. As noted above, previous work implicated a role for lysosomes in this degradation. Through in vitro and in vivo studies, we now demonstrate that the sortilin-mediated route of apoB to lysosomes is unconventional and intersects with autophagy. Increased expression of sortilin diverts more apoB away from secretion, with both proteins trafficking to the endosomal compartment in vesicles that fuse with autophagosomes to form amphisomes. The amphisomes then merge with lysosomes. Furthermore, we show that sortilin itself is a regulator of autophagy and that its activity is scaled to the level of apoB synthesis. CONCLUSIONS These results strongly suggest that an unconventional lysosomal targeting process dependent on autophagy degrades apoB that was diverted from the secretory pathway by sortilin and provides a mechanism contributing to the reduced LDL-C found in individuals with SORT1 overexpression.
Collapse
Affiliation(s)
- Jaume Amengual
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Liang Guo
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Alanna Strong
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Julio Madrigal-Matute
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Haizhen Wang
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Susmita Kaushik
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Jeffrey L Brodsky
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Daniel J Rader
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Ana Maria Cuervo
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.)
| | - Edward A Fisher
- From the Division of Cardiology (J.A., L.G., H.W., E.A.F.), Department of Medicine (J.A., L.G., H.W., E.A.F.), and Marc and Ruti Bell Program in Vascular Biology (J.A., E.A.F., L.G, H.W.), NYU School of Medicine; Institute for Translational Medicine and Therapeutics, Cardiovascular Institute, and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (A.S., D.J.R.); Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York (J.M.-M., S.K., A.M.C.); and Department of Biological Sciences, University of Pittsburgh, PA (J.L.B.).
| |
Collapse
|
15
|
Rindflesch TC, Blake CL, Fiszman M, Kilicoglu H, Rosemblat G, Schneider J, Zeiss CJ. Informatics Support for Basic Research in Biomedicine. ILAR J 2017; 58:80-89. [PMID: 28838071 DOI: 10.1093/ilar/ilx004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 01/13/2017] [Indexed: 11/13/2022] Open
Abstract
Informatics methodologies exploit computer-assisted techniques to help biomedical researchers manage large amounts of information. In this paper, we focus on the biomedical research literature (MEDLINE). We first provide an overview of some text mining techniques that offer assistance in research by identifying biomedical entities (e.g., genes, substances, and diseases) and relations between them in text.We then discuss Semantic MEDLINE, an application that integrates PubMed document retrieval, concept and relation identification, and visualization, thus enabling a user to explore concepts and relations from within a set of retrieved citations. Semantic MEDLINE provides a roadmap through content and helps users discern patterns in large numbers of retrieved citations. We illustrate its use with an informatics method we call "discovery browsing," which provides a principled way of navigating through selected aspects of some biomedical research area. The method supports an iterative process that accommodates learning and hypothesis formation in which a user is provided with high level connections before delving into details.As a use case, we examine current developments in basic research on mechanisms of Alzheimer's disease. Out of the nearly 90 000 citations returned by the PubMed query "Alzheimer's disease," discovery browsing led us to 73 citations on sortilin and that disorder. We provide a synopsis of the basic research reported in 15 of these. There is wide-spread consensus among researchers working with a range of animal models and human cells that increased sortilin expression and decreased receptor expression are associated with amyloid beta and/or amyloid precursor protein.
Collapse
Affiliation(s)
- Thomas C Rindflesch
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois, Urbana-Champaign; Center for Informatics in Science and Scholarship. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois Urbana-Champaign, Champaign, Illinois. Yale University School of Medicine, New Haven, Connecticut
| | - Catherine L Blake
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois, Urbana-Champaign; Center for Informatics in Science and Scholarship. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois Urbana-Champaign, Champaign, Illinois. Yale University School of Medicine, New Haven, Connecticut
| | - Marcelo Fiszman
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois, Urbana-Champaign; Center for Informatics in Science and Scholarship. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois Urbana-Champaign, Champaign, Illinois. Yale University School of Medicine, New Haven, Connecticut
| | - Halil Kilicoglu
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois, Urbana-Champaign; Center for Informatics in Science and Scholarship. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois Urbana-Champaign, Champaign, Illinois. Yale University School of Medicine, New Haven, Connecticut
| | - Graciela Rosemblat
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois, Urbana-Champaign; Center for Informatics in Science and Scholarship. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois Urbana-Champaign, Champaign, Illinois. Yale University School of Medicine, New Haven, Connecticut
| | - Jodi Schneider
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois, Urbana-Champaign; Center for Informatics in Science and Scholarship. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois Urbana-Champaign, Champaign, Illinois. Yale University School of Medicine, New Haven, Connecticut
| | - Caroline J Zeiss
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois, Urbana-Champaign; Center for Informatics in Science and Scholarship. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. Lister Hill National Center for Biomedical Communications, National Library of Medicine, Bethesda, Maryland. School of Information Sciences, University of Illinois Urbana-Champaign, Champaign, Illinois. Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
16
|
Ma Q, Yang J, Li T, Milner TA, Hempstead BL. Selective reduction of striatal mature BDNF without induction of proBDNF in the zQ175 mouse model of Huntington's disease. Neurobiol Dis 2015; 82:466-477. [PMID: 26282324 PMCID: PMC4819334 DOI: 10.1016/j.nbd.2015.08.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 08/02/2015] [Accepted: 08/12/2015] [Indexed: 02/02/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by massive loss of medium spiny neurons in the striatum. However, the mechanisms by which mutant huntingtin leads to this selective neuronal death remain incompletely understood. Brain-derived neurotrophic factor (BDNF) has been shown to be neuroprotective on HD striatal neurons both in vitro and in vivo. ProBDNF, the precursor of mature BDNF (mBDNF), also can be secreted but promotes apoptosis of neurons expressing p75(NTR) and sortilin receptors. Although a reduction of total striatal BDNF protein has been reported in HD patients and mouse models, it remains unclear whether conversion of proBDNF to mBDNF is altered in HD, and whether the proBDNF receptors, p75(NTR) and sortilin are dysregulated, leading to impaired striatal neuron survival. To test these hypotheses, we generated bdnf-HA knock-in (KI) mice on the zQ175 HD background to accurately quantitate the levels of both proBDNF and mBDNF in the HD striatum. In aged zQ175 HD mice, we observed a significant loss of mBDNF and decreased TrkB activation, but no increase of proBDNF or p75(NTR) levels either in the sensorimotor cortex or the striatum. However, immunoreactivities of p75(NTR) and sortilin receptor are both increased in immature striatal oligodendrocytes, which associate with significant myelin defects in the HD striatum. Taken together, the present study indicates that diminished mature BDNF trophic signaling through the TrkB receptor, rather than an induction in proBDNF, is a main contributing factor to the vulnerability of striatal neurons in the zQ175 HD mouse model.
Collapse
Affiliation(s)
- Qian Ma
- Graduate Program of Neuroscience, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Thomas Li
- Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Barbara L Hempstead
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
17
|
Ferreira ST, Lourenco MV, Oliveira MM, De Felice FG. Soluble amyloid-β oligomers as synaptotoxins leading to cognitive impairment in Alzheimer's disease. Front Cell Neurosci 2015; 9:191. [PMID: 26074767 PMCID: PMC4443025 DOI: 10.3389/fncel.2015.00191] [Citation(s) in RCA: 247] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/30/2015] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia in the elderly, and affects millions of people worldwide. As the number of AD cases continues to increase in both developed and developing countries, finding therapies that effectively halt or reverse disease progression constitutes a major research and public health challenge. Since the identification of the amyloid-β peptide (Aβ) as the major component of the amyloid plaques that are characteristically found in AD brains, a major effort has aimed to determine whether and how Aβ leads to memory loss and cognitive impairment. A large body of evidence accumulated in the past 15 years supports a pivotal role of soluble Aβ oligomers (AβOs) in synapse failure and neuronal dysfunction in AD. Nonetheless, a number of basic questions, including the exact molecular composition of the synaptotoxic oligomers, the identity of the receptor(s) to which they bind, and the signaling pathways that ultimately lead to synapse failure, remain to be definitively answered. Here, we discuss recent advances that have illuminated our understanding of the chemical nature of the toxic species and the deleterious impact they have on synapses, and have culminated in the proposal of an Aβ oligomer hypothesis for Alzheimer’s pathogenesis. We also highlight outstanding questions and challenges in AD research that should be addressed to allow translation of research findings into effective AD therapies.
Collapse
Affiliation(s)
- Sergio T Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil ; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Mauricio M Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro Rio de Janeiro, RJ, Brazil
| |
Collapse
|
18
|
Viola KL, Klein WL. Amyloid β oligomers in Alzheimer's disease pathogenesis, treatment, and diagnosis. Acta Neuropathol 2015; 129:183-206. [PMID: 25604547 DOI: 10.1007/s00401-015-1386-3] [Citation(s) in RCA: 458] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/11/2015] [Accepted: 01/11/2015] [Indexed: 12/22/2022]
Abstract
Protein aggregation is common to dozens of diseases including prionoses, diabetes, Parkinson's and Alzheimer's. Over the past 15 years, there has been a paradigm shift in understanding the structural basis for these proteinopathies. Precedent for this shift has come from investigation of soluble Aβ oligomers (AβOs), toxins now widely regarded as instigating neuron damage leading to Alzheimer's dementia. Toxic AβOs accumulate in AD brain and constitute long-lived alternatives to the disease-defining Aβ fibrils deposited in amyloid plaques. Key experiments using fibril-free AβO solutions demonstrated that while Aβ is essential for memory loss, the fibrillar Aβ in amyloid deposits is not the agent. The AD-like cellular pathologies induced by AβOs suggest their impact provides a unifying mechanism for AD pathogenesis, explaining why early stage disease is specific for memory and accounting for major facets of AD neuropathology. Alternative ideas for triggering mechanisms are being actively investigated. Some research favors insertion of AβOs into membrane, while other evidence supports ligand-like accumulation at particular synapses. Over a dozen candidate toxin receptors have been proposed. AβO binding triggers a redistribution of critical synaptic proteins and induces hyperactivity in metabotropic and ionotropic glutamate receptors. This leads to Ca(2+) overload and instigates major facets of AD neuropathology, including tau hyperphosphorylation, insulin resistance, oxidative stress, and synapse loss. Because different species of AβOs have been identified, a remaining question is which oligomer is the major pathogenic culprit. The possibility has been raised that more than one species plays a role. Despite some key unknowns, the clinical relevance of AβOs has been established, and new studies are beginning to point to co-morbidities such as diabetes and hypercholesterolemia as etiological factors. Because pathogenic AβOs appear early in the disease, they offer appealing targets for therapeutics and diagnostics. Promising therapeutic strategies include use of CNS insulin signaling enhancers to protect against the presence of toxins and elimination of the toxins through use of highly specific AβO antibodies. An AD-dependent accumulation of AβOs in CSF suggests their potential use as biomarkers and new AβO probes are opening the door to brain imaging. Overall, current evidence indicates that Aβ oligomers provide a substantive molecular basis for the cause, treatment and diagnosis of Alzheimer's disease.
Collapse
|
19
|
EFFECT OF CURCUMIN LIPOSOMAL FORM ON ANGIOTENSIN CONVERTING ACTIVITY, CYTOKINES AND COGNITIVE CHARACTERISTICS OF THE RATS WITH ALZHEIMER’S DISEASE MODEL. BIOTECHNOLOGIA ACTA 2015. [DOI: 10.15407/biotech8.06.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
20
|
Goure WF, Krafft GA, Jerecic J, Hefti F. Targeting the proper amyloid-beta neuronal toxins: a path forward for Alzheimer's disease immunotherapeutics. ALZHEIMERS RESEARCH & THERAPY 2014; 6:42. [PMID: 25045405 PMCID: PMC4100318 DOI: 10.1186/alzrt272] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Levels of amyloid-beta monomer and deposited amyloid-beta in the Alzheimer’s
disease brain are orders of magnitude greater than soluble amyloid-beta oligomer
levels. Monomeric amyloid-beta has no known direct toxicity. Insoluble fibrillar
amyloid-beta has been proposed to be an in vivo mechanism for removal of
soluble amyloid-beta and exhibits relatively low toxicity. In contrast, soluble
amyloid-beta oligomers are widely reported to be the most toxic amyloid-beta form,
both causing acute synaptotoxicity and inducing neurodegenerative processes. None of
the amyloid-beta immunotherapies currently in clinical development selectively target
soluble amyloid-beta oligomers, and their lack of efficacy is not unexpected
considering their selectivity for monomeric or fibrillar amyloid-beta (or both)
rather than soluble amyloid-beta oligomers. Because they exhibit acute,
memory-compromising synaptic toxicity and induce chronic neurodegenerative toxicity
and because they exist at very low in vivo levels in the Alzheimer’s
disease brain, soluble amyloid-beta oligomers constitute an optimal immunotherapeutic
target that should be pursued more aggressively.
Collapse
Affiliation(s)
- William F Goure
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Grant A Krafft
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Jasna Jerecic
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Franz Hefti
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| |
Collapse
|
21
|
No association of SORT1 gene polymorphism with sporadic Alzheimer's disease in the Chinese Han population. Neuroreport 2013; 24:464-8. [PMID: 23660633 DOI: 10.1097/wnr.0b013e3283619f43] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Increasing evidence shows that sortilin (encoded by SORT1 gene), a member of the vacuolar protein sorting 10 family of sorting receptors, can modulate amyloid-β peptides (Aβ) metabolism and clearance, as well as mediate the neurotoxicity of the Aβ oligomer and proneurotrophins, thus playing diverse roles in the pathogenesis of Alzheimer's disease. To assess the association between single nucleotide polymorphism (SNP) of the SORT1 gene and sporadic Alzheimer's disease (sAD) in the Chinese Han population, a case-control study was carried out including 220 sAD patients and 245 controls. One tag SNP was selected from the entire SORT1 gene through construction of linkage disequilibrium blocks, and three SNPs located in the vicinity of SORT1 that affect its expression were also selected. The four target SNPs were genotyped using a multiplex PCR-ligase detection reaction method, yielding no significant association between them or haplotypes containing three of them, and the risk of sAD. The results of this study indicate that polymorphisms of the SORT1 gene are unlikely to confer the risk of sAD in the Chinese Han population.
Collapse
|
22
|
Disease modifying therapies for Alzheimer's disease targeting Aβ oligomers: implications for therapeutic mechanisms. BIOMED RESEARCH INTERNATIONAL 2013; 2013:984041. [PMID: 24063020 PMCID: PMC3770037 DOI: 10.1155/2013/984041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 12/23/2022]
Abstract
Several lines of evidence indicate that amyloid β (Aβ), particularly Aβ oligomers (AβOs), plays a causative role in Alzheimer's disease. However, the mechanisms underlying the action of an anti-AβO antibody to clarify the toxic action of AβOs remain elusive. Here, we showed that the anti-AβO antibody (monoclonal 72D9) can modify the Aβ aggregation pathway. We also found that 72D9 directly sequesters both extracellular and intraneuronal AβOs in a nontoxic state. Thus, therapeutic intervention targeting AβOs is a promising strategy for neuronal protection in Alzheimer's disease.
Collapse
|
23
|
Saadipour K, Yang M, Lim Y, Georgiou K, Sun Y, Keating D, Liu J, Wang YR, Gai WP, Zhong JH, Wang YJ, Zhou XF. Amyloid beta1-42
(Aβ42
) up-regulates the expression of sortilin via the p75NTR
/RhoA signaling pathway. J Neurochem 2013; 127:152-62. [DOI: 10.1111/jnc.12383] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/04/2013] [Accepted: 07/05/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Khalil Saadipour
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
- Department of Human Physiology; Centre for Neuroscience; University of Flinders; Adelaide SA Australia
| | - Miao Yang
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
| | - Yoon Lim
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
| | - Kristen Georgiou
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
| | - Ying Sun
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
| | - Damien Keating
- Department of Human Physiology; Centre for Neuroscience; University of Flinders; Adelaide SA Australia
| | - Jia Liu
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
| | - Ye-Ran Wang
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
- Department of Neurology and Center for Clinical Neuroscience; Daping Hospital; Chongqing China
| | - Wei-ping Gai
- Department of Human Physiology; Centre for Neuroscience; University of Flinders; Adelaide SA Australia
| | - Jin-hua Zhong
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience; Daping Hospital; Chongqing China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences; Faculty of Health Sciences; University of South Australia; Adelaide SA Australia
| |
Collapse
|
24
|
Xia Y, Chen BY, Sun XL, Duan L, Gao GD, Wang JJ, Yung KKL, Chen LW. Presence of proNGF-sortilin signaling complex in nigral dopamine neurons and its variation in relation to aging, lactacystin and 6-OHDA insults. Int J Mol Sci 2013; 14:14085-104. [PMID: 23880857 PMCID: PMC3742233 DOI: 10.3390/ijms140714085] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/20/2013] [Accepted: 06/25/2013] [Indexed: 12/15/2022] Open
Abstract
Growing evidence has shown that proNGF-p75NTR-sortilin signaling might be a crucial factor in neurodegeneration, but it remains unclear if it may function in nigral neurons under aging and disease. The purpose of this study is to examine and quantify proNGF and sortilin expression in the substantia nigra and dynamic changes of aging in lactacystin and 6-hydroxydopamine (6-OHDA) rat models of Parkinson’s disease using immunofluorescence, electronic microscopy, western blot and FLIVO staining methods. The expression of proNGF and sortilin was abundantly and selectively identified in tyrosine hydroxylase (TH)-containing dopamine neurons in the substantia nigra. These proNGF/TH, sortilin/TH-positive neurons were densely distributed in the ventral tier, while they were less distributed in the dorsal tier, where calbindin-D28K-containing neurons were numerously located. A correlated decrease of proNGF, sortilin and TH was also detected during animal aging process. While increase of proNGF, sortilin and cleaved (active) caspase-3 expression was found in the lactacystin model, dynamic proNGF and sortilin changes along with dopamine neuronal loss were demonstrated in the substantia nigra of both the lactacystin and 6-OHDA models. This study has thus revealed the presence of the proNGF-sortilin signaling complex in nigral dopamine neurons and its response to aging, lactacystin and 6-OHDA insults, suggesting that it might contribute to neuronal apoptosis or neurodegeneration during pathogenesis and disease progression of Parkinson’s disease; the underlying mechanism and key signaling pathways involved warrant further investigation.
Collapse
Affiliation(s)
- Yi Xia
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
- Department of Neurosurgery, Tangdou Hospital, Fourth Military Medical University, Xi’an 710038, China; E-Mail:
| | - Bei-Yu Chen
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; E-Mail:
| | - Xiao-Long Sun
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
| | - Li Duan
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdou Hospital, Fourth Military Medical University, Xi’an 710038, China; E-Mail:
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdou Hospital, Fourth Military Medical University, Xi’an 710038, China
- Authors to whom correspondence should be addressed; E-Mails: (J.-J.W.); (L.-W.C.); Tel.: +86-29-8477-6840 (L.-W.C.); Fax: +86-29-8324-6270 (L.-W.C.)
| | - Ken Kam-Lin Yung
- Department of Biology, Baptist University of Hong Kong, Hong Kong, China; E-Mail:
| | - Liang-Wei Chen
- Institute of Neurosciences, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Y.X.); (X.-L.S.); (L.D.)
- Authors to whom correspondence should be addressed; E-Mails: (J.-J.W.); (L.-W.C.); Tel.: +86-29-8477-6840 (L.-W.C.); Fax: +86-29-8324-6270 (L.-W.C.)
| |
Collapse
|
25
|
The cell biology of prion-like spread of protein aggregates: mechanisms and implication in neurodegeneration. Biochem J 2013; 452:1-17. [DOI: 10.1042/bj20121898] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The misfolding and aggregation of specific proteins is a common hallmark of many neurodegenerative disorders, including highly prevalent illnesses such as Alzheimer's and Parkinson's diseases, as well as rarer disorders such as Huntington's and prion diseases. Among these, only prion diseases are ‘infectious’. By seeding misfolding of the PrPC (normal conformer prion protein) into PrPSc (abnormal disease-specific conformation of prion protein), prions spread from the periphery of the body to the central nervous system and can also be transmitted between individuals of the same or different species. However, recent exciting data suggest that the transmissibility of misfolded proteins within the brain is a property that goes way beyond the rare prion diseases. Evidence indicates that non-prion aggregates [tau, α-syn (α-synuclein), Aβ (amyloid-β) and Htt (huntingtin) aggregates] can also move between cells and seed the misfolding of their normal conformers. These findings have enormous implications. On the one hand they question the therapeutical use of transplants, and on the other they indicate that it may be possible to bring these diseases to an early arrest by preventing cell-to-cell transmission. To better understand the prion-like spread of these protein aggregates it is essential to identify the underlying cellular and molecular factors. In the present review we analyse and discuss the evidence supporting prion-like spreading of amyloidogenic proteins, especially focusing on the cellular and molecular mechanisms and their significance.
Collapse
|