1
|
Calabrese G, Molzahn C, Mayor T. Protein interaction networks in neurodegenerative diseases: from physiological function to aggregation. J Biol Chem 2022; 298:102062. [PMID: 35623389 PMCID: PMC9234719 DOI: 10.1016/j.jbc.2022.102062] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/26/2022] [Accepted: 05/18/2022] [Indexed: 11/25/2022] Open
Abstract
The accumulation of protein inclusions is linked to many neurodegenerative diseases that typically develop in older individuals, due to a combination of genetic and environmental factors. In rare familial neurodegenerative disorders, genes encoding for aggregation-prone proteins are often mutated. While the underlying mechanism leading to these diseases still remains to be fully elucidated, efforts in the past 20 years revealed a vast network of protein–protein interactions that play a major role in regulating the aggregation of key proteins associated with neurodegeneration. Misfolded proteins that can oligomerize and form insoluble aggregates associate with molecular chaperones and other elements of the proteolytic machineries that are the frontline workers attempting to protect the cells by promoting clearance and preventing aggregation. Proteins that are normally bound to aggregation-prone proteins can become sequestered and mislocalized in protein inclusions, leading to their loss of function. In contrast, mutations, posttranslational modifications, or misfolding of aggregation-prone proteins can lead to gain of function by inducing novel or altered protein interactions, which in turn can impact numerous essential cellular processes and organelles, such as vesicle trafficking and the mitochondria. This review examines our current knowledge of protein–protein interactions involving several key aggregation-prone proteins that are associated with Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, or amyotrophic lateral sclerosis. We aim to provide an overview of the protein interaction networks that play a central role in driving or mitigating inclusion formation, while highlighting some of the key proteomic studies that helped to uncover the extent of these networks.
Collapse
Affiliation(s)
- Gaetano Calabrese
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada.
| | - Cristen Molzahn
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada
| | - Thibault Mayor
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada.
| |
Collapse
|
2
|
Rahman MM, Westermark GT, Zetterberg H, Härd T, Sandgren M. Protofibrillar and Fibrillar Amyloid-β Binding Proteins in Cerebrospinal Fluid. J Alzheimers Dis 2019; 66:1053-1064. [PMID: 30372682 DOI: 10.3233/jad-180596] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Aggregation and deposition of misfolded amyloid-β (Aβ) peptide in the brain is central to Alzheimer's disease (AD). Oligomeric, protofibrillar, and fibrillar forms of Aβ are believed to be neurotoxic and cause neurodegeneration in AD, but the toxicity mechanisms are not well understood and may involve Aβ-interacting molecular partners. In a previous study, we identified potential Aβ42 protofibrillar-binding proteins in serum and cerebrospinal fluid (CSF) using an engineered version of Aβ42 (Aβ42CC) that forms protofibrils, but not fibrils. Here we studied binding of proteins to Aβ42 fibrils in AD and non-AD CSF and compared these with protofibrillar Aβ42CC-binding partners. Aβ42 fibrils sequestered 2.4-fold more proteins than Aβ42CC protofibrils. Proteins with selective binding to fibrillar aggregates with low nanomolar affinity were identified. We also found that protofibrillar and fibrillar Aβ-binding proteins represent distinct functional categories. Aβ42CC protofibrils triggered interactions with proteins involved in catalytic activities, like transferases and oxidoreductases, while Aβ42 fibrils were more likely involved in binding to proteoglycans, growth factors and neuron-associated proteins, e.g., neurexin-1, -2, and -3. Interestingly, 10 brain-enriched proteins were identified among the fibril-binding proteins, while protofibril-extracted proteins had more general expression patterns. Both types of Aβ aggregates bound several extracellular proteins. Additionally, we list a set of CSF proteins that might have potential to discriminate between AD and non-AD CSF samples. The results may be of relevance both for biomarker studies and for studies of Aβ-related toxicity mechanisms.
Collapse
Affiliation(s)
- M Mahafuzur Rahman
- Department of Molecular Sciences, Swedish University of Agricultural Sciences (SLU), Uppsala BioCenter, Uppsala, Sweden
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Torleif Härd
- Department of Molecular Sciences, Swedish University of Agricultural Sciences (SLU), Uppsala BioCenter, Uppsala, Sweden
| | - Mats Sandgren
- Department of Molecular Sciences, Swedish University of Agricultural Sciences (SLU), Uppsala BioCenter, Uppsala, Sweden
| |
Collapse
|
3
|
Andrew RJ, Fisher K, Heesom KJ, Kellett KAB, Hooper NM. Quantitative interaction proteomics reveals differences in the interactomes of amyloid precursor protein isoforms. J Neurochem 2019; 149:399-412. [PMID: 30664241 DOI: 10.1111/jnc.14666] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/21/2018] [Accepted: 01/16/2019] [Indexed: 02/04/2023]
Abstract
The generation of the amyloid-β (Aβ) peptides from the amyloid precursor protein (APP) through sequential proteolysis by β- and γ-secretases is a key pathological event in the initiation and propagation of Alzheimer's disease. Aβ and the transcriptionally active APP intracellular domain are generated preferentially from the APP695 isoform compared to the longer APP751 isoform. As the Aβ and amyloid precursor protein intracellular domain produced from cleavage of APP695 and APP751 are identical we hypothesised that the two isoforms have differences within their interactomes which mediate the differential processing of the two isoforms. To investigate this, we applied a proteomics-based approach to identify differences in the interactomes of the APP695 and APP751 isoforms. Using stable isotope labelling of amino acids in cell culture and quantitative proteomics, we compared the interactomes of APP695 and APP751 expressed in human SH-SY5Y cells. Through this approach, we identified enrichment of proteins involved in mitochondrial function, the nuclear pore and nuclear transport specifically in the APP695 interactome. Further interrogation of the APP interactome and subsequent experimental validation (co-immunoprecipitation and siRNA knockdown) revealed GAP43 as a specific modulator of APP751 proteolysis, altering Aβ generation. Our data indicate that interrogation of the APP interactome can be exploited to identify proteins which influence APP proteolysis and Aβ production in an isoform dependent-manner. Cover Image for this issue: doi: 10.1111/jnc.14504.
Collapse
Affiliation(s)
- Robert J Andrew
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Kate Fisher
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Kate J Heesom
- Proteomics Facility, Faculty of Medical and Veterinary Sciences, University of Bristol, Bristol, UK
| | - Katherine A B Kellett
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Nigel M Hooper
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Salza R, Lethias C, Ricard-Blum S. The Multimerization State of the Amyloid-β42 Amyloid Peptide Governs its Interaction Network with the Extracellular Matrix. J Alzheimers Dis 2018; 56:991-1005. [PMID: 28106549 DOI: 10.3233/jad-160751] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The goals of this work were i) to identify the interactions of amyloid-β (Aβ)42 under monomeric, oligomeric, and fibrillar forms with the extracellular matrix (ECM) and receptors, ii) to determine the influence of Aβ42 supramolecular organization on these interactions, and iii) to identify the molecular functions, biological processes, and pathways targeted by Aβ42 in the ECM. The ECM and cell surface partners of Aβ42 and its supramolecular forms were identified with protein and glycosaminoglycan (GAG) arrays (81 molecules in triplicate) probed by surface plasmon resonance imaging. The number of partners of Aβ42 increased upon its multimerization, ranging from 4 for the peptide up to 53 for the fibrillar aggregates. The peptide interacted only with ECM proteins but their percentage among Aβ42 partners decreased upon multimerization. Aβ42 and its supramolecular forms recognized different molecular features on their partners, and the partners of Aβ42 fibrillar forms were enriched in laminin IV-A, N-terminal, and EGF-like domains. Aβ42 oligomerization triggered interactions with receptors, whereas Aβ42 fibrillogenesis promoted binding to GAGs, proteoglycans, enzymes, and growth factors and the ability to interact with perineuronal nets. Fibril aggregation bind to further membrane proteins including tumor endothelial marker-8, syndecan-4, and discoidin-domain receptor-2. The partners of the Aβ42 supramolecular forms are enriched in proteins contributing to cell growth and/or maintenance, involved in integrin cell surface interactions and expressed in kidney cancer, preadipocytes, and dentin. In conclusion, the supramolecular assembly of Aβ42 governs its ability to interact in vitro with ECM proteins, remodeling and crosslinking ECM enzymes, proteoglycans, and receptors.
Collapse
Affiliation(s)
- Romain Salza
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS - Université Lyon 1, Villeurbanne cedex, France
| | - Claire Lethias
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique (LBTI), UMR 5305 CNRS - Université Lyon 1, Lyon, Cedex 07, France
| | - Sylvie Ricard-Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS - Université Lyon 1, Villeurbanne cedex, France
| |
Collapse
|
5
|
Recabarren-Leiva D, Alarcón M. New insights into the gene expression associated to amyotrophic lateral sclerosis. Life Sci 2018; 193:110-123. [DOI: 10.1016/j.lfs.2017.12.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/01/2017] [Accepted: 12/10/2017] [Indexed: 12/11/2022]
|
6
|
Sosa LJ, Cáceres A, Dupraz S, Oksdath M, Quiroga S, Lorenzo A. The physiological role of the amyloid precursor protein as an adhesion molecule in the developing nervous system. J Neurochem 2017; 143:11-29. [PMID: 28677143 DOI: 10.1111/jnc.14122] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
The amyloid precursor protein (APP) is a type I transmembrane glycoprotein better known for its participation in the physiopathology of Alzheimer disease as the source of the beta amyloid fragment. However, the physiological functions of the full length protein and its proteolytic fragments have remained elusive. APP was first described as a cell-surface receptor; nevertheless, increasing evidence highlighted APP as a cell adhesion molecule. In this review, we will focus on the current knowledge of the physiological role of APP as a cell adhesion molecule and its involvement in key events of neuronal development, such as migration, neurite outgrowth, growth cone pathfinding, and synaptogenesis. Finally, since APP is over-expressed in Down syndrome individuals because of the extra copy of chromosome 21, in the last section of the review, we discuss the potential contribution of APP to the neuronal and synaptic defects described in this genetic condition. Read the Editorial Highlight for this article on page 9. Cover Image for this issue: doi. 10.1111/jnc.13817.
Collapse
Affiliation(s)
- Lucas J Sosa
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Cáceres
- Laboratorio Neurobiología, Instituto Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto Universitario Ciencias Biomédicas Córdoba, Córdoba, Argentina
| | - Sebastián Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenarative Diseases, Bonn, Germany
| | - Mariana Oksdath
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Lorenzo
- Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
7
|
Abstract
Alzheimer's disease (AD) is the most common form of adult neurode-generation and is characterised by progressive loss of cognitive function leading to death. The neuropathological hallmarks include extracellular amyloid plaque accumulation in affected regions of the brain, formation of intraneuronal neurofibrillary tangles, chronic neuroinflammation, oxidative stress, and abnormal biometal homeostasis. Of the latter, major changes in copper (Cu) levels and localisation have been identified in AD brain, with accumulation of Cu in amyloid deposits, together with deficiency of Cu in some brain regions. The amyloid precursor protein (APP) and the amyloid beta (Aβ) peptide both have Cu binding sites, and interaction with Cu can lead to potentially neurotoxic outcomes through generation of reactive oxygen species. In addition, AD patients have systemic changes to Cu metabolism, and altered Cu may also affect neuroinflammatory outcomes in AD. Although we still have much to learn about Cu homeostasis in AD patients and its role in disease aetiopathology, therapeutic approaches for regulating Cu levels and interactions with Cu-binding proteins in the brain are currently being developed. This review will examine how Cu is associated with pathological changes in the AD brain and how these may be targeted for therapeutic intervention.
Collapse
|
8
|
Andrew RJ, Kellett KAB, Thinakaran G, Hooper NM. A Greek Tragedy: The Growing Complexity of Alzheimer Amyloid Precursor Protein Proteolysis. J Biol Chem 2016; 291:19235-44. [PMID: 27474742 DOI: 10.1074/jbc.r116.746032] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Proteolysis of the amyloid precursor protein (APP) liberates various fragments including the proposed initiator of Alzheimer disease-associated dysfunctions, amyloid-β. However, recent evidence suggests that the accepted view of APP proteolysis by the canonical α-, β-, and γ-secretases is simplistic, with the discovery of a number of novel APP secretases (including δ- and η-secretases, alternative β-secretases) and additional metabolites, some of which may also cause synaptic dysfunction. Furthermore, various proteins have been identified that interact with APP and modulate its cleavage by the secretases. Here, we give an overview of the increasingly complex picture of APP proteolysis.
Collapse
Affiliation(s)
- Robert J Andrew
- From the Departments of Neurobiology, Neurology, and Pathology, The University of Chicago, Chicago, Illinois 60637 and
| | - Katherine A B Kellett
- the Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Gopal Thinakaran
- From the Departments of Neurobiology, Neurology, and Pathology, The University of Chicago, Chicago, Illinois 60637 and
| | - Nigel M Hooper
- the Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
9
|
Long PA, Larsen BT, Evans JM, Olson TM. Exome Sequencing Identifies Pathogenic and Modifier Mutations in a Child With Sporadic Dilated Cardiomyopathy. J Am Heart Assoc 2015; 4:JAHA.115.002443. [PMID: 26656454 PMCID: PMC4845292 DOI: 10.1161/jaha.115.002443] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Idiopathic dilated cardiomyopathy (DCM) is typically diagnosed in adulthood, yet familial cases exhibit variable age‐dependent penetrance and a subset of patients develop sporadic DCM in childhood. We sought to discover the molecular basis of sporadic DCM in an 11‐year‐old female with severe heart failure necessitating cardiac transplantation. Methods and Results Parental echocardiograms excluded asymptomatic DCM. Whole exome sequencing was performed on the family trio and filtered for rare, deleterious, recessive, and de novo variants. Of the 8 candidate genes identified, only 2 had a role in cardiac physiology. A de novo missense mutation in TNNT2 was identified, previously reported and functionally validated in familial DCM with markedly variable penetrance. Additionally, recessive compound heterozygous truncating mutations were identified in XIRP2, a member of the ancient Xin gene family, which governs intercalated disc (ICD) maturation. Histomorphological analysis of explanted heart tissue revealed misregistration, mislocalization, and shortening of ICDs, findings similar to Xirp2−/− mice. Conclusions The synergistic effects of TNNT2 and XIRP2 mutations, resulting in perturbed sarcomeric force generation and transmission, respectively, would account for an early‐onset heart failure phenotype. Whereas the importance of Xin proteins in cardiac development has been well established in animal models, this study implicates XIRP2 as a novel modifier gene in the pathogenesis of DCM.
Collapse
Affiliation(s)
- Pamela A Long
- Mayo Graduate School, Molecular Pharmacology and Experimental Therapeutics Track, Mayo Clinic, Rochester, MN (P.A.L.) Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, MN (P.A.L., T.M.O.)
| | - Brandon T Larsen
- Department of Pathology, University of Arizona Medical Center, Tucson, AZ (B.T.L.)
| | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN (J.M.E.)
| | - Timothy M Olson
- Cardiovascular Genetics Research Laboratory, Mayo Clinic, Rochester, MN (P.A.L., T.M.O.) Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN (T.M.O.) Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN (T.M.O.)
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW The axon plays a central role in both the injury and repair phases after stroke. This review highlights emerging principles in the study of axonal injury in stroke and the role of the axon in neural repair after stroke. RECENT FINDINGS Ischemic stroke produces a rapid and significant loss of axons in the acute phase. This early loss of axons results from a primary ischemic injury that triggers a wave of calcium signaling, activating proteolytic mechanisms and downstream signaling cascades. A second progressive phase of axonal injury occurs during the subacute period and damages axons that survive the initial ischemic insult but go on to experience a delayed axonal degeneration driven in part by changes in axoglial contact and axonal energy metabolism. Recovery from stroke is dependent on axonal sprouting and reconnection that occurs during a third degenerative/regenerative phase. Despite this central role played by the axon, comparatively little is understood about the molecular pathways that contribute to early and subacute axonal degeneration after stroke. Recent advances in axonal neurobiology and signaling suggest new targets that hold promise as potential molecular therapeutics including axonal calcium signaling, axoglial energy metabolism and cell adhesion as well as retrograde axonal mitogen-activated protein kinase pathways. These novel pathways must be modeled appropriately as the type and severity of axonal injury vary by stroke subtype. SUMMARY Stroke-induced injury to axons occurs in three distinct phases each with a unique molecular underpinning. A wealth of new data about the molecular organization and molecular signaling within axons is available but not yet robustly applied to the study of axonal injury after stroke. Identifying the spatiotemporal patterning of molecular pathways within the axon that contribute to injury and repair may offer new therapeutic strategies for the treatment of stroke.
Collapse
|
11
|
van der Kant R, Goldstein LSB. Cellular functions of the amyloid precursor protein from development to dementia. Dev Cell 2015; 32:502-15. [PMID: 25710536 DOI: 10.1016/j.devcel.2015.01.022] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Amyloid precursor protein (APP) is a key player in Alzheimer's disease (AD). The Aβ fragments of APP are the major constituent of AD-associated amyloid plaques, and mutations or duplications of the gene coding for APP can cause familial AD. Here we review the roles of APP in neuronal development, signaling, intracellular transport, and other aspects of neuronal homeostasis. We suggest that APP acts as a signaling nexus that transduces information about a range of extracellular conditions, including neuronal damage, to induction of intracellular signaling events. Subtle disruptions of APP signaling functions may be major contributors to AD-causing neuronal dysfunction.
Collapse
Affiliation(s)
- Rik van der Kant
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA 92093, USA.
| | - Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
12
|
Corsetti V, Florenzano F, Atlante A, Bobba A, Ciotti MT, Natale F, Della Valle F, Borreca A, Manca A, Meli G, Ferraina C, Feligioni M, D'Aguanno S, Bussani R, Ammassari-Teule M, Nicolin V, Calissano P, Amadoro G. NH2-truncated human tau induces deregulated mitophagy in neurons by aberrant recruitment of Parkin and UCHL-1: implications in Alzheimer's disease. Hum Mol Genet 2015; 24:3058-81. [PMID: 25687137 DOI: 10.1093/hmg/ddv059] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/10/2015] [Indexed: 01/26/2023] Open
Abstract
Disarrangement in functions and quality control of mitochondria at synapses are early events in Alzheimer's disease (AD) pathobiology. We reported that a 20-22 kDa NH2-tau fragment mapping between 26 and 230 amino acids of the longest human tau isoform (aka NH2htau): (i) is detectable in cellular and animal AD models, as well in synaptic mitochondria and cerebrospinal fluids (CSF) from human AD subjects; (ii) is neurotoxic in primary hippocampal neurons; (iii) compromises the mitochondrial biology both directly, by inhibiting the ANT-1-dependent ADP/ATP exchange, and indirectly, by impairing their selective autophagic clearance (mitophagy). Here, we show that the extensive Parkin-dependent turnover of mitochondria occurring in NH2htau-expressing post-mitotic neurons plays a pro-death role and that UCHL-1, the cytosolic Ubiquitin-C-terminal hydrolase L1 which directs the physiological remodeling of synapses by controlling ubiquitin homeostasis, critically contributes to mitochondrial and synaptic failure in this in vitro AD model. Pharmacological or genetic suppression of improper mitophagy, either by inhibition of mitochondrial targeting to autophagosomes or by shRNA-mediated silencing of Parkin or UCHL-1 gene expression, restores synaptic and mitochondrial content providing partial but significant protection against the NH2htau-induced neuronal death. Moreover, in mitochondria from human AD synapses, the endogenous NH2htau is stably associated with Parkin and with UCHL-1. Taken together, our studies show a causative link between the excessive mitochondrial turnover and the NH2htau-induced in vitro neuronal death, suggesting that pathogenetic tau truncation may contribute to synaptic deterioration in AD by aberrant recruitment of Parkin and UCHL-1 to mitochondria making them more prone to detrimental autophagic clearance.
Collapse
Affiliation(s)
- V Corsetti
- Institute of Translational Pharmacology (IFT) - National Research Council (CNR), Via Fosso del Cavaliere 100-00133, Rome, Italy
| | - F Florenzano
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - A Atlante
- Institute of Biomembranes and Bioenergetics (IBBE)-CNR, Via Amendola 165/A, 70126 Bari, Italy
| | - A Bobba
- Institute of Biomembranes and Bioenergetics (IBBE)-CNR, Via Amendola 165/A, 70126 Bari, Italy
| | - M T Ciotti
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - F Natale
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - F Della Valle
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - A Borreca
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - A Manca
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - G Meli
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - C Ferraina
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - M Feligioni
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - S D'Aguanno
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - R Bussani
- UCO Pathological Anatomy and Histopathology Unit, Cattinara Hospital Strada di Fiume 447, 34149 Trieste, Italy and
| | - M Ammassari-Teule
- Institute of Cellular Biology and Neuroscience (IBCN)-CNR, IRCSS Santa Lucia Foundation Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - V Nicolin
- Department of Medical, Surgical and Health Science, University of Trieste, Strada di Fiume 449, 34149 Trieste, Italy
| | - P Calissano
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| | - G Amadoro
- Institute of Translational Pharmacology (IFT) - National Research Council (CNR), Via Fosso del Cavaliere 100-00133, Rome, Italy European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64-65, 00143 Rome, Italy
| |
Collapse
|
13
|
Del Prete D, Lombino F, Liu X, D'Adamio L. APP is cleaved by Bace1 in pre-synaptic vesicles and establishes a pre-synaptic interactome, via its intracellular domain, with molecular complexes that regulate pre-synaptic vesicles functions. PLoS One 2014; 9:e108576. [PMID: 25247712 PMCID: PMC4172690 DOI: 10.1371/journal.pone.0108576] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/31/2014] [Indexed: 12/21/2022] Open
Abstract
Amyloid Precursor Protein (APP) is a type I membrane protein that undergoes extensive processing by secretases, including BACE1. Although mutations in APP and genes that regulate processing of APP, such as PSENs and BRI2/ITM2B, cause dementias, the normal function of APP in synaptic transmission, synaptic plasticity and memory formation is poorly understood. To grasp the biochemical mechanisms underlying the function of APP in the central nervous system, it is important to first define the sub-cellular localization of APP in synapses and the synaptic interactome of APP. Using biochemical and electron microscopy approaches, we have found that APP is localized in pre-synaptic vesicles, where it is processed by Bace1. By means of a proteomic approach, we have characterized the synaptic interactome of the APP intracellular domain. We focused on this region of APP because in vivo data underline the central functional and pathological role of the intracellular domain of APP. Consistent with the expression of APP in pre-synaptic vesicles, the synaptic APP intracellular domain interactome is predominantly constituted by pre-synaptic, rather than post-synaptic, proteins. This pre-synaptic interactome of the APP intracellular domain includes proteins expressed on pre-synaptic vesicles such as the vesicular SNARE Vamp2/Vamp1 and the Ca2+ sensors Synaptotagmin-1/Synaptotagmin-2, and non-vesicular pre-synaptic proteins that regulate exocytosis, endocytosis and recycling of pre-synaptic vesicles, such as target-membrane-SNAREs (Syntaxin-1b, Syntaxin-1a, Snap25 and Snap47), Munc-18, Nsf, α/β/γ-Snaps and complexin. These data are consistent with a functional role for APP, via its carboxyl-terminal domain, in exocytosis, endocytosis and/or recycling of pre-synaptic vesicles.
Collapse
Affiliation(s)
- Dolores Del Prete
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Franco Lombino
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Luciano D'Adamio
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
14
|
Laßek M, Weingarten J, Volknandt W. The synaptic proteome. Cell Tissue Res 2014; 359:255-65. [PMID: 25038742 DOI: 10.1007/s00441-014-1943-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/04/2014] [Indexed: 11/29/2022]
Abstract
Synapses are focal hot spots for signal transduction and plasticity in the brain. A synapse comprises an axon terminus, the presynapse, the synaptic cleft containing extracellular matrix proteins as well as adhesion molecules, and the postsynaptic density as target structure for chemical signaling. The proteomes of the presynaptic and postsynaptic active zones control neurotransmitter release and perception. These tasks demand short- and long-term structural and functional dynamics of the synapse mediated by its proteinaceous inventory. This review addresses subcellular fractionation protocols and the related proteomic approaches to the various synaptic subcompartments with an emphasis on the presynaptic active zone (PAZ). Furthermore, it discusses major constituents of the PAZ including the amyloid precursor protein family members. Numerous proteins regulating the rearrangement of the cytoskeleton are indicative of the functional and structural dynamics of the pre- and postsynapse. The identification of protein candidates of the synapse provides the basis for further analyzing the interaction of synaptic proteins with their targets, and the effect of their deletion opens novel insights into the functional role of these proteins in neuronal communication. The knowledge of the molecular interactome is also a prerequisite for understanding numerous neurodegenerative diseases.
Collapse
Affiliation(s)
- Melanie Laßek
- Molecular and Cellular Neurobiology, Goethe University, Frankfurt, Germany
| | | | | |
Collapse
|
15
|
Zaręba-Kozioł M, Szwajda A, Dadlez M, Wysłouch-Cieszyńska A, Lalowski M. Global analysis of S-nitrosylation sites in the wild type (APP) transgenic mouse brain-clues for synaptic pathology. Mol Cell Proteomics 2014; 13:2288-305. [PMID: 24895380 DOI: 10.1074/mcp.m113.036079] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by an early synaptic loss, which strongly correlates with the severity of dementia. The pathogenesis and causes of characteristic AD symptoms are not fully understood. Defects in various cellular cascades were suggested, including the imbalance in production of reactive oxygen and nitrogen species. Alterations in S-nitrosylation of several proteins were previously demonstrated in various AD animal models and patients. In this work, using combined biotin-switch affinity/nano-LC-MS/MS and bioinformatic approaches we profiled endogenous S-nitrosylation of brain synaptosomal proteins from wild type and transgenic mice overexpressing mutated human Amyloid Precursor Protein (hAPP). Our data suggest involvement of S-nitrosylation in the regulation of 138 synaptic proteins, including MAGUK, CamkII, or synaptotagmins. Thirty-eight proteins were differentially S-nitrosylated in hAPP mice only. Ninety-five S-nitrosylated peptides were identified for the first time (40% of total, including 33 peptides exclusively in hAPP synaptosomes). We verified differential S-nitrosylation of 10 (26% of all identified) synaptosomal proteins from hAPP mice, by Western blotting with specific antibodies. Functional enrichment analysis linked S-nitrosylated proteins to various cellular pathways, including: glycolysis, gluconeogenesis, calcium homeostasis, ion, and vesicle transport, suggesting a basic role of this post-translational modification in the regulation of synapses. The linkage of SNO-proteins to axonal guidance and other processes related to APP metabolism exclusively in the hAPP brain, implicates S-nitrosylation in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Monika Zaręba-Kozioł
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Michał Dadlez
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Maciej Lalowski
- ¶Biomedicum Helsinki, Institute of Biomedicine, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Unit, University of Helsinki, Finland; ‖Folkhälsan Institute of Genetics, Helsinki, Finland
| |
Collapse
|
16
|
Guffanti A, Simchovitz A, Soreq H. Emerging bioinformatics approaches for analysis of NGS-derived coding and non-coding RNAs in neurodegenerative diseases. Front Cell Neurosci 2014; 8:89. [PMID: 24723850 PMCID: PMC3973899 DOI: 10.3389/fncel.2014.00089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/10/2014] [Indexed: 02/01/2023] Open
Abstract
Neurodegenerative diseases in general and specifically late-onset Alzheimer’s disease (LOAD) involve a genetically complex and largely obscure ensemble of causative and risk factors accompanied by complex feedback responses. The advent of “high-throughput” transcriptome investigation technologies such as microarray and deep sequencing is increasingly being combined with sophisticated statistical and bioinformatics analysis methods complemented by knowledge-based approaches such as Bayesian Networks or network and graph analyses. Together, such “integrative” studies are beginning to identify co-regulated gene networks linked with biological pathways and potentially modulating disease predisposition, outcome, and progression. Specifically, bioinformatics analyses of integrated microarray and genotyping data in cases and controls reveal changes in gene expression of both protein-coding and small and long regulatory RNAs; highlight relevant quantitative transcriptional differences between LOAD and non-demented control brains and demonstrate reconfiguration of functionally meaningful molecular interaction structures in LOAD. These may be measured as changes in connectivity in “hub nodes” of relevant gene networks (Zhang etal., 2013). We illustrate here the open analytical questions in the transcriptome investigation of neurodegenerative disease studies, proposing “ad hoc” strategies for the evaluation of differential gene expression and hints for a simple analysis of the non-coding RNA (ncRNA) part of such datasets. We then survey the emerging role of long ncRNAs (lncRNAs) in the healthy and diseased brain transcriptome and describe the main current methods for computational modeling of gene networks. We propose accessible modular and pathway-oriented methods and guidelines for bioinformatics investigations of whole transcriptome next generation sequencing datasets. We finally present methods and databases for functional interpretations of lncRNAs and propose a simple heuristic approach to visualize and represent physical and functional interactions of the coding and non-coding components of the transcriptome. Integrating in a functional and integrated vision coding and ncRNA analyses is of utmost importance for current and future analyses of neurodegenerative transcriptomes.
Collapse
Affiliation(s)
- Alessandro Guffanti
- Laboratory of Molecular Neuroscience, Department of Biological Chemistry, The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem Jerusalem, Israel ; Bioinformatics, Genomnia srl Milano, Italy
| | - Alon Simchovitz
- Laboratory of Molecular Neuroscience, Department of Biological Chemistry, The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem Jerusalem, Israel
| | - Hermona Soreq
- Laboratory of Molecular Neuroscience, Department of Biological Chemistry, The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem Jerusalem, Israel
| |
Collapse
|
17
|
Could dysregulation of UPS be a common underlying mechanism for cancer and neurodegeneration? Lessons from UCHL1. Cell Biochem Biophys 2014; 67:45-53. [PMID: 23695785 DOI: 10.1007/s12013-013-9631-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ubiquitin proteasome system (UPS) determines the timing and extent of protein turnover in cells, and it is one of the most strictly controlled cellular mechanisms. Lack of proper control over UPS is attributed to both cancer and to neurodegenerative diseases, yet in different context and direction. Cancerous cells have altered cellular metabolisms, uncontrolled cellular division, and increased proteasome activity. The specialized function prevent neurons from undergoing cellular division but allow them to extend an axon over long distances, establish connections, and to form stable neuronal circuitries. Neurons heavily depend on the proper function of the proteasome and the UPS for their proper function. Reduction of UPS function in vulnerable neurons results in protein aggregation, increased ER stress, and cell death. Identification of compounds that selectively block proteasome function in distinct set of malignancies added momentum to drug discovery efforts, and deubiquitinases (DUBs) gained much attention. This review will focus on ubiquitin carboxy-terminal hydrolase L1 (UCHL1), a DUB that is attributed to both cancer and neurodegeneration. The potential of developing effective treatment strategies for two major health problems by controlling the function of UPS opens up new avenues for innovative approaches and therapeutic interventions.
Collapse
|
18
|
Mot AI, Wedd AG, Sinclair L, Brown DR, Collins SJ, Brazier MW. Metal attenuating therapies in neurodegenerative disease. Expert Rev Neurother 2014; 11:1717-45. [DOI: 10.1586/ern.11.170] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
19
|
Laßek M, Weingarten J, Einsfelder U, Brendel P, Müller U, Volknandt W. Amyloid precursor proteins are constituents of the presynaptic active zone. J Neurochem 2013; 127:48-56. [PMID: 23815291 DOI: 10.1111/jnc.12358] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/21/2013] [Accepted: 06/26/2013] [Indexed: 11/27/2022]
Abstract
The amyloid precursor protein (APP) and its mammalian homologs, APLP1, APLP2, have been allocated to an organellar pool residing in the Golgi apparatus and in endosomal compartments, and in its mature form to a cell surface-localized pool. In the brain, all APPs are restricted to neurons; however, their precise localization at the plasma membrane remained enigmatic. Employing a variety of subcellular fractionation steps, we isolated two synaptic vesicle (SV) pools from rat and mouse brain, a pool consisting of synaptic vesicles only and a pool comprising SV docked to the presynaptic plasma membrane. Immunopurification of these two pools using a monoclonal antibody directed against the 12 membrane span synaptic vesicle protein2 (SV2) demonstrated unambiguously that APP, APLP1 and APLP2 are constituents of the active zone of murine brain but essentially absent from free synaptic vesicles. The specificity of immunodetection was confirmed by analyzing the respective knock-out animals. The fractionation experiments further revealed that APP is accumulated in the fraction containing docked synaptic vesicles. These data present novel insights into the subsynaptic localization of APPs and are a prerequisite for unraveling the physiological role of all mature APP proteins in synaptic physiology.
Collapse
Affiliation(s)
- Melanie Laßek
- Department of Molecular and Cellular Neurobiology, Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Cathepsin D deficiency induces cytoskeletal changes and affects cell migration pathways in the brain. Neurobiol Dis 2013; 50:107-19. [DOI: 10.1016/j.nbd.2012.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 09/21/2012] [Accepted: 10/03/2012] [Indexed: 01/04/2023] Open
|
21
|
Malyshev I. The Role of HSP70 in the Protection of: (A) The Brain in Alzheimer’s Disease and (B) The Heart in Cardiac Surgery. IMMUNITY, TUMORS AND AGING: THE ROLE OF HSP70 2013. [DOI: 10.1007/978-94-007-5943-5_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
22
|
Ma SL, Huang W, Tang NL, Lam LC. MxA Polymorphisms Are Associated with Risk and Age-at-Onset in Alzheimer Disease and Accelerated Cognitive Decline in Chinese Elders. Rejuvenation Res 2012; 15:516-22. [DOI: 10.1089/rej.2012.1328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Suk Ling Ma
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Wei Huang
- Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Medical Sciences & Peking Union Medical College, Beijing, China
| | - Nelson L.S. Tang
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Linda C.W. Lam
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
23
|
Götz J, Matamales M, Götz NN, Ittner LM, Eckert A. Alzheimer's disease models and functional genomics-How many needles are there in the haystack? Front Physiol 2012; 3:320. [PMID: 22934069 PMCID: PMC3429089 DOI: 10.3389/fphys.2012.00320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/23/2012] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) are complex human brain disorders that affect an increasing number of people worldwide. With the identification first of the proteins that aggregate in AD and FTLD brains and subsequently of pathogenic gene mutations that cause their formation in the familial cases, the foundation was laid for the generation of animal models. These recapitulate essential aspects of the human conditions; expression of mutant forms of the amyloid-β protein-encoding APP gene in mice reproduces amyloid-β (Aβ) plaque formation in AD, while that of mutant forms of the tau-encoding microtubule-associated protein tau (MAPT) gene reproduces tau-containing neurofibrillary tangle formation, a lesion that is also prevalent in FTLD-Tau. The mouse models have been complemented by those in lower species such as C. elegans or Drosophila, highlighting the crucial role for Aβ and tau in human neurodegenerative disease. In this review, we will introduce selected AD/FTLD models and discuss how they were instrumental, by identifying deregulated mRNAs, miRNAs and proteins, in dissecting pathogenic mechanisms in neurodegenerative disease. We will discuss some recent examples, which includes miRNA species that are specifically deregulated by Aβ, mitochondrial proteins that are targets of both Aβ and tau, and the nuclear splicing factor SFPQ that accumulates in the cytoplasm in a tau-dependent manner. These examples illustrate how a functional genomics approach followed by a careful validation in experimental models and human tissue leads to a deeper understanding of the pathogenesis of AD and FTLD and ultimately, may help in finding a cure.
Collapse
Affiliation(s)
- Jürgen Götz
- Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandSt Lucia, QLD, Australia
| | - Miriam Matamales
- Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandSt Lucia, QLD, Australia
| | - Naeman N. Götz
- Centre for Ageing Dementia Research, Queensland Brain Institute, The University of QueenslandSt Lucia, QLD, Australia
| | - Lars M. Ittner
- Alzheimer's and Parkinson's Disease Laboratory, Brain and Mind Research Institute, University of SydneyCamperdown, NSW, Australia
| | - Anne Eckert
- Neurobiology Laboratory, Psychiatric University Clinics Basel, University of BaselBasel, Switzerland
| |
Collapse
|
24
|
Kohli BM, Pflieger D, Mueller LN, Carbonetti G, Aebersold R, Nitsch RM, Konietzko U. Interactome of the amyloid precursor protein APP in brain reveals a protein network involved in synaptic vesicle turnover and a close association with Synaptotagmin-1. J Proteome Res 2012; 11:4075-90. [PMID: 22731840 DOI: 10.1021/pr300123g] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Knowledge of the protein networks interacting with the amyloid precursor protein (APP) in vivo can shed light on the physiological function of APP. To date, most proteins interacting with the APP intracellular domain (AICD) have been identified by Yeast Two Hybrid screens which only detect direct interaction partners. We used a proteomics-based approach by biochemically isolating tagged APP from the brains of transgenic mice and subjecting the affinity-purified complex to mass spectrometric (MS) analysis. Using two different quantitative MS approaches, we compared the protein composition of affinity-purified samples isolated from wild-type mice versus transgenic mice expressing tagged APP. This enabled us to assess truly enriched proteins in the transgenic sample and yielded an overlapping set of proteins containing the major proteins involved in synaptic vesicle endo- and exocytosis. Confocal microscopy analyses of cotransfected primary neurons showed colocalization of APP with synaptic vesicle proteins in vesicular structures throughout the neurites. We analyzed the interaction of APP with these proteins using pulldown experiments from transgenic mice or cotransfected cells followed by Western blotting. Synaptotagmin-1 (Stg1), a resident synaptic vesicle protein, was found to directly bind to APP. We fused Citrine and Cerulean to APP and the candidate proteins and measured fluorescence resonance energy transfer (FRET) in differentiated SH-SY5Y cells. Differentially tagged APPs showed clear sensitized FRET emission, in line with the described dimerization of APP. Among the candidate APP-interacting proteins, again only Stg1 was in close proximity to APP. Our results strongly argue for a function of APP in synaptic vesicle turnover in vivo. Thus, in addition to the APP cleavage product Aβ, which influences synaptic transmission at the postsynapse, APP interacts with the calcium sensor of synaptic vesicles and might thus play a role in the regulation of synaptic vesicle exocytosis.
Collapse
Affiliation(s)
- Bernhard M Kohli
- Institute of Psychiatry Research and Psychogeriatric Medicine, Faculty of Science, University Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
25
|
Goldstein LSB. Axonal transport and neurodegenerative disease: can we see the elephant? Prog Neurobiol 2012; 99:186-90. [PMID: 22484448 DOI: 10.1016/j.pneurobio.2012.03.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 03/18/2012] [Accepted: 03/20/2012] [Indexed: 01/07/2023]
Abstract
Although it is well established that axonal transport defects are part of the initiation or progression of some neurodegenerative diseases, the precise role of these defects in disease development is poorly understood. Thus, in this article, rather than enumerate the already well-reviewed evidence that there are transport deficits in disease, I will focus on a discussion of two crucial and unanswered questions about the possible role of axonal transport defects in HD and AD. (1) Are alterations in axonal transport caused by changes in the normal function of proteins mutated or altered in HD and AD and/or do such alterations in transport occur as a result of the formation of toxic aggregates of peptides or proteins? (2) Do alterations in axonal transport contribute to the causes of HD and AD or are they early, or late, secondary consequences of other cellular defects caused by disease-induction?
Collapse
Affiliation(s)
- Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine and Department of Neurosciences, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0695, USA.
| |
Collapse
|
26
|
Abstract
Alzheimer’s disease (AD) is one key medical challenge of the aging society and despite a great amount of effort and a huge collection of acquired data on molecular mechanisms that are associated with the onset and progression of this devastating disorder, no causal therapy is in sight. The two main hypotheses of AD, the amyloid cascade hypothesis and the Tau hypothesis, are still in the focus of AD research. With aging as the accepted main risk factor of the most important non familial and late onset sporadic forms of AD, it is now mandatory to discuss more intensively aspects of cellular aging and aging biochemistry and its impact on neurodegeneration. Since aging is accompanied by changes in cellular protein homeostasis and an increasing demand for protein degradation, aspects of protein folding, misfolding, refolding and, importantly, protein degradation need to be linked to AD pathogenesis. This is the purpose of this short review.
Collapse
|
27
|
Volknandt W, Karas M. Proteomic analysis of the presynaptic active zone. Exp Brain Res 2012; 217:449-61. [DOI: 10.1007/s00221-012-3031-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 02/04/2012] [Indexed: 02/06/2023]
|
28
|
Redeker V, Hughes C, Savistchenko J, Vissers JPC, Melki R. Qualitative and quantitative multiplexed proteomic analysis of complex yeast protein fractions that modulate the assembly of the yeast prion Sup35p. PLoS One 2011; 6:e23659. [PMID: 21931608 PMCID: PMC3172207 DOI: 10.1371/journal.pone.0023659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 07/22/2011] [Indexed: 01/25/2023] Open
Abstract
Background The aggregation of the baker's yeast prion Sup35p is at the origin of the transmissible [PSI+] trait. We and others have shown that molecular chaperones modulate Sup35p aggregation. However, other protein classes might be involved in [PSI+] formation. Results We designed a functional proteomic study that combines two techniques to identify modulators of Sup35p aggregation and describe the changes associated to [PSI+] formation. The first allows measuring the effect of fractionated Saccharomyces cerevisiae cytosolic extracts from [PSI+] and [psi−] yeast cells on Sup35p assembly. The second is a multiplex qualitative and quantitative comparison of protein composition of active and inactive fractions using a gel-free and label-free LC-MS approach. We identify changes in proteins involved in translation, folding, degradation, oxido-reduction and metabolic processes. Conclusion Our functional proteomic study provides the first inventory list of over 300 proteins that directly or indirectly affect Sup35p aggregation and [PSI+] formation. Our results highlight the complexity of the cellular changes accompanying [PSI+] formation and pave the way for in vitro studies aimed to document the effect of individual and/or combinations of proteins identified here, susceptible of affecting Sup35p assembly.
Collapse
Affiliation(s)
- Virginie Redeker
- Laboratoire d'Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
- * E-mail: (VR); (RM)
| | - Chris Hughes
- Waters Corporation, Atlas Park, Manchester, United Kingdom
| | - Jimmy Savistchenko
- Laboratoire d'Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
| | | | - Ronald Melki
- Laboratoire d'Enzymologie et Biochimie Structurales, Centre National de la Recherche Scientifique, Gif-sur-Yvette, France
- * E-mail: (VR); (RM)
| |
Collapse
|
29
|
Roet KCD, Bossers K, Franssen EHP, Ruitenberg MJ, Verhaagen J. A meta-analysis of microarray-based gene expression studies of olfactory bulb-derived olfactory ensheathing cells. Exp Neurol 2011; 229:10-45. [PMID: 21396936 DOI: 10.1016/j.expneurol.2011.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 12/28/2010] [Accepted: 03/02/2011] [Indexed: 12/23/2022]
Abstract
Genome wide transcriptional profiling and large scale proteomics have emerged as two powerful methods to dissect the molecular properties of specific neural tissues or cell types on a global scale. Several genome-wide transcriptional profiling and proteomics studies have been published on cultured olfactory ensheathing cells (OEC). In this article we present a meta-analysis of all five published and publicly available micro-array gene expression datasets of cultured early-passage-OB-OEC with other cell types (Schwann cells, late-passage-OB-OEC, mucosa-OEC, an OEC cell line, and acutely dissected OEC). The aim of this meta-analysis is to identify genes and molecular pathways that are found in multiple instead of one isolated study. 454 Genes were detected in at least three out of five microarray datasets. In this "Top-list", genes involved in the biological processes "growth of neurites", "blood vessel development", "migration of cells" and "immune response" were strongly overrepresented. By applying network analysis tools, molecular networks were constructed and Hub-genes were identified that may function as key genes in the above mentioned interrelated processes. We also identified 7 genes (ENTPD2, MATN2, CTSC, PTHLH, GLRX1, COL27A1 and ID2) with uniformly higher or lower expression in early-passage-OB-OEC in all five microarray comparisons. These genes have diverse but intriguing roles in neuroprotection, neurite extension and/or tissue repair. Our meta-analysis provides novel insights into the molecular basis of OB-OEC-mediated neural repair and can serve as a repository for investigators interested in the molecular biology of OEC. This article is part of a Special Issue entitled: Understanding olfactory ensheathing glia and their prospect for nervous system repair.
Collapse
Affiliation(s)
- Kasper C D Roet
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
30
|
Ojha J, Karmegam RV, Masilamoni JG, Terry AV, Cashikar AG. Behavioral defects in chaperone-deficient Alzheimer's disease model mice. PLoS One 2011; 6:e16550. [PMID: 21379584 PMCID: PMC3040748 DOI: 10.1371/journal.pone.0016550] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 12/31/2010] [Indexed: 12/03/2022] Open
Abstract
Molecular chaperones protect cells from the deleterious effects of protein misfolding and aggregation. Neurotoxicity of amyloid-beta (Aβ) aggregates and their deposition in senile plaques are hallmarks of Alzheimer's disease (AD). We observed that the overall content of αB-crystallin, a small heat shock protein molecular chaperone, decreased in AD model mice in an age-dependent manner. We hypothesized that αB-crystallin protects cells against Aβ toxicity. To test this, we crossed αB-crystallin/HspB2 deficient (CRYAB⁻/⁻HSPB2⁻/⁻) mice with AD model transgenic mice expressing mutant human amyloid precursor protein. Transgenic and non-transgenic mice in chaperone-sufficient or deficient backgrounds were examined for representative behavioral paradigms for locomotion and memory network functions: (i) spatial orientation and locomotion was monitored by open field test; (ii) sequential organization and associative learning was monitored by fear conditioning; and (iii) evoked behavioral response was tested by hot plate method. Interestingly, αB-crystallin/HspB2 deficient transgenic mice were severely impaired in locomotion compared to each genetic model separately. Our results highlight a synergistic effect of combining chaperone deficiency in a transgenic mouse model for AD underscoring an important role for chaperones in protein misfolding diseases.
Collapse
Affiliation(s)
- Juhi Ojha
- Center for Molecular Chaperones and Radiobiology, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Rajalakshmi V. Karmegam
- Center for Molecular Chaperones and Radiobiology, Medical College of Georgia, Augusta, Georgia, United States of America
| | - J. Gunasingh Masilamoni
- Center for Molecular Chaperones and Radiobiology, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Alvin V. Terry
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Anil G. Cashikar
- Center for Molecular Chaperones and Radiobiology, Medical College of Georgia, Augusta, Georgia, United States of America
| |
Collapse
|
31
|
Taurines R, Dudley E, Grassl J, Warnke A, Gerlach M, Coogan AN, Thome J. Proteomic research in psychiatry. J Psychopharmacol 2011; 25:151-96. [PMID: 20142298 DOI: 10.1177/0269881109106931] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Psychiatric disorders such as Alzheimer's disease, schizophrenia and mood disorders are severe and disabling conditions of largely unknown origin and poorly understood pathophysiology. An accurate diagnosis and treatment of these disorders is often complicated by their aetiological and clinical heterogeneity. In recent years proteomic technologies based on mass spectrometry have been increasingly used, especially in the search for diagnostic and prognostic biomarkers in neuropsychiatric disorders. Proteomics enable an automated high-throughput protein determination revealing expression levels, post-translational modifications and complex protein-interaction networks. In contrast to other methods such as molecular genetics, proteomics provide the opportunity to determine modifications at the protein level thereby possibly being more closely related to pathophysiological processes underlying the clinical phenomenology of specific psychiatric conditions. In this article we review the theoretical background of proteomics and its most commonly utilized techniques. Furthermore the current impact of proteomic research on diverse psychiatric diseases, such as Alzheimer's disease, schizophrenia, mood and anxiety disorders, drug abuse and autism, is discussed. Proteomic methods are expected to gain crucial significance in psychiatric research and neuropharmacology over the coming decade.
Collapse
Affiliation(s)
- Regina Taurines
- Academic Unit of Psychiatry, The School of Medicine, Institute of Life Science, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Morell M, de Groot NS, Vendrell J, Avilés FX, Ventura S. Linking amyloid protein aggregation and yeast survival. MOLECULAR BIOSYSTEMS 2011; 7:1121-8. [PMID: 21240401 DOI: 10.1039/c0mb00297f] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Protein aggregation and amyloid formation lie behind an increasing number of human diseases. Here we describe the application of an "aggregation reporter", in which the test protein is fused to dihydrofolate reductase, as a general method to assess the intracellular solubility of amyloid proteins in eukaryotic background. Because the aggregation state of the target protein is linked directly to yeast cells survival in the presence of methotrexate, protein solubility can be monitored in vivo without the requirement of a functional assay for the protein of interest. In addition, the approach allows the in vivo visualization of the cellular location and aggregated state of the target protein. To demonstrate the applicability of the assay in the screening of genes or compounds that modulate amyloid protein aggregation in living cells, we have used as models the Alzheimer's amyloid β peptide, polyglutamine expansions of huntingtin, α-synuclein and non-aggregating variants thereof. Moreover, the anti-aggregational properties of small molecules and the effects of the yeast protein quality control machinery have also been evaluated using this method.
Collapse
Affiliation(s)
- Montse Morell
- Departament de Bioquímica i Biologia Molecular, Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain.
| | | | | | | | | |
Collapse
|
33
|
Perreau VM, Orchard S, Adlard PA, Bellingham SA, Cappai R, Ciccotosto GD, Cowie TF, Crouch PJ, Duce JA, Evin G, Faux NG, Hill AF, Hung YH, James SA, Li QX, Mok SS, Tew DJ, White AR, Bush AI, Hermjakob H, Masters CL. A domain level interaction network of amyloid precursor protein and Abeta of Alzheimer's disease. Proteomics 2010; 10:2377-95. [PMID: 20391539 DOI: 10.1002/pmic.200900773] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The primary constituent of the amyloid plaque, beta-amyloid (Abeta), is thought to be the causal "toxic moiety" of Alzheimer's disease. However, despite much work focused on both Abeta and its parent protein, amyloid precursor protein (APP), the functional roles of APP and its cleavage products remain to be fully elucidated. Protein-protein interaction networks can provide insight into protein function, however, high-throughput data often report false positives and are in frequent disagreement with low-throughput experiments. Moreover, the complexity of the CNS is likely to be under represented in such databases. Therefore, we curated the published work characterizing both APP and Abeta to create a protein interaction network of APP and its proteolytic cleavage products, with annotation, where possible, to the level of APP binding domain and isoform. This is the first time that an interactome has been refined to domain level, essential for the interpretation of APP due to the presence of multiple isoforms and processed fragments. Gene ontology and network analysis were used to identify potentially novel functional relationships among interacting proteins.
Collapse
Affiliation(s)
- Victoria M Perreau
- Neuroproteomics and Neurogenomics Platform, National Neurosciences Facility, The University of Melbourne, Parkville, VIC, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sun F, Cavalli V. Neuroproteomics approaches to decipher neuronal regeneration and degeneration. Mol Cell Proteomics 2010; 9:963-75. [PMID: 20019051 PMCID: PMC2871427 DOI: 10.1074/mcp.r900003-mcp200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/14/2009] [Indexed: 01/02/2023] Open
Abstract
Given the complexity of brain and nerve tissues, systematic approaches are essential to understand normal physiological conditions and functional alterations in neurological diseases. Mass spectrometry-based proteomics is increasingly used in neurosciences to determine both basic and clinical differential protein expression, protein-protein interactions, and post-translational modifications. Proteomics approaches are especially useful to understand the mechanisms of nerve regeneration and degeneration because changes in axons following injury or in disease states often occur without the contribution of transcriptional events in the cell body. Indeed, the current understanding of axonal function in health and disease emphasizes the role of proteolysis, local axonal protein synthesis, and a broad range of post-translational modifications. Deciphering how axons regenerate and degenerate has thus become a postgenomics problem, which depends in part on proteomics approaches. This review focuses on recent proteomics approaches designed to uncover the mechanisms and molecules involved in neuronal regeneration and degeneration. It emerges that the principal degenerative mechanisms converge to oxidative stress, dysfunctions of axonal transport, mitochondria, chaperones, and the ubiquitin-proteasome systems. The mechanisms regulating nerve regeneration also impinge on axonal transport, cytoskeleton, and chaperones in addition to changes in signaling pathways. We also discuss the major challenges to proteomics work in the nervous system given the complex organization of the brain and nerve tissue at the anatomical, cellular, and subcellular levels.
Collapse
Affiliation(s)
- Faneng Sun
- From the Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri 63110
| | - Valeria Cavalli
- From the Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri 63110
| |
Collapse
|
35
|
Gozal YM, Duong DM, Gearing M, Cheng D, Hanfelt JJ, Funderburk C, Peng J, Lah JJ, Levey AI. Proteomics analysis reveals novel components in the detergent-insoluble subproteome in Alzheimer's disease. J Proteome Res 2010; 8:5069-79. [PMID: 19746990 DOI: 10.1021/pr900474t] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are often defined pathologically by the presence of protein aggregates. These aggregates, including amyloid plaques in Alzheimer's disease (AD), result from the abnormal accumulation and processing of proteins, and may ultimately lead to neuronal dysfunction and cell death. To date, conventional biochemical studies have revealed abundant core components in protein aggregates. However, rapidly improving proteomics technologies offer opportunities to revisit pathologic aggregate composition, and to identify less abundant but potentially important functional molecules that participate in neurodegeneration. The purpose of this study was to establish a proteomic strategy for the profiling of neurodegenerative disease tissues for disease-specific changes in protein abundance. Using high resolution liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS), we analyzed detergent-insoluble frontal cortex samples from AD and unaffected control cases. In addition, we analyzed samples from frontotemporal lobar degeneration (FTLD) cases to identify AD-specific changes not present in other neurodegenerative diseases. We used a labeling-free quantification technique to compare the abundance of identified peptides in the samples based on extracted ion current (XIC) of their corresponding ions. Of the 512 identified proteins, quantitation demonstrated significant changes in 81 AD-specific proteins. Following additional manual filtering, 11 proteins were accepted with high confidence as increased in AD compared to control and FTLD brains, including beta-amyloid, tau and apolipoprotein E, all well-established AD-linked proteins. In addition, we identified and validated the presence of serine protease 15, ankyrin B, and 14-3-3 eta in the detergent-insoluble fraction. Our results provide further evidence for the capacity of proteomics applications to identify conserved sets of disease-specific proteins in AD, to enhance our understanding of disease pathogenesis, and to deliver new candidates for the development of effective therapies for this, and other, devastating neurodegenerative disorders.
Collapse
Affiliation(s)
- Yair M Gozal
- Department of Neurology, Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Koren J, Jinwal UK, Lee DC, Jones JR, Shults CL, Johnson AG, Anderson LJ, Dickey CA. Chaperone signalling complexes in Alzheimer's disease. J Cell Mol Med 2009; 13:619-30. [PMID: 19449461 PMCID: PMC2749087 DOI: 10.1111/j.1582-4934.2008.00557.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Molecular chaperones and heat shock proteins (Hsp) have emerged as critical regulators of proteins associated with neurodegenerative disease pathologies. The very nature of the chaperone system, which is to maintain protein quality control, means that most nascent proteins come in contact with chaperone proteins. Thus, amyloid precursor protein (APP), members of the gamma-secretase complex (presenilin 1 [PS1] collectively), the microtubule-associated protein tau (MAPT) as well as a number of neuroinflammatory components are all in contact with chaperones from the moment of their production. Chaperones are often grouped together as one machine presenting abnormal or mutant proteins to the proteasome for degradation, but this is not at all the case. In fact, the chaperone family consists of more than 100 proteins in mammalian cells, and the primary role for most of these proteins is to protect clients following synthesis and during stress; only as a last resort do they facilitate protein degradation. To the best of our current knowledge, the chaperone system in eukaryotic cells revolves around the ATPase activities of Hsp70 and Hsp90, the two primary chaperone scaffolds. Other chaperones and co-chaperones manipulate the ATPase activities of Hsp70 and Hsp90, facilitating either folding of the client or its degradation. In the case of Alzheimer's disease (AD), a number of studies have recently emerged describing the impact that these chaperones have on the proteotoxic effects of tau and amyloid-β accumulation. Here, we present the current understandings of chaperone biology and examine the literature investigating these proteins in the context of AD.
Collapse
Affiliation(s)
- John Koren
- Johnnie B. Byrd Sr. Alzheimer's Center and Research Institute, Department of Molecular Medicine, University of South Florida, Tampa, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Watanabe Y, Morita E, Fukada Y, Doi K, Yasui K, Kitayama M, Nakano T, Nakashima K. Adherent monomer-misfolded SOD1. PLoS One 2008; 3:e3497. [PMID: 18946506 PMCID: PMC2567031 DOI: 10.1371/journal.pone.0003497] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 09/22/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Multiple cellular functions are compromised in amyotrophic lateral sclerosis (ALS). In familial ALS (FALS) with Cu/Zn superoxide dismutase (SOD1) mutations, the mechanisms by which the mutation in SOD1 leads to such a wide range of abnormalities remains elusive. METHODOLOGY/PRINCIPAL FINDINGS To investigate underlying cellular conditions caused by the SOD1 mutation, we explored mutant SOD1-interacting proteins in the spinal cord of symptomatic transgenic mice expressing a mutant SOD1, SOD1(Leu126delTT) with a FLAG sequence (DF mice). This gene product is structurally unable to form a functional homodimer. Tissues were obtained from both DF mice and disease-free mice expressing wild-type with FLAG SOD1 (WF mice). Both FLAG-tagged SOD1 and cross-linking proteins were enriched and subjected to a shotgun proteomic analysis. We identified 34 proteins (or protein subunits) in DF preparations, while in WF preparations, interactions were detected with only 4 proteins. CONCLUSIONS/SIGNIFICANCE These results indicate that disease-causing mutant SOD1 likely leads to inadequate protein-protein interactions. This could be an early and crucial process in the pathogenesis of FALS.
Collapse
Affiliation(s)
- Yasuhiro Watanabe
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
- * E-mail:
| | - Eri Morita
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yasuyo Fukada
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Koji Doi
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kenichi Yasui
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Michio Kitayama
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Toshiya Nakano
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Kenji Nakashima
- Department of Neurology, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
38
|
Banwait S, Galvan V, Zhang J, Gorostiza OF, Ataie M, Huang W, Crippen D, Koo EH, Bredesen DE. C-terminal cleavage of the amyloid-beta protein precursor at Asp664: a switch associated with Alzheimer's disease. J Alzheimers Dis 2008; 13:1-16. [PMID: 18334752 DOI: 10.3233/jad-2008-13101] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In addition to the proteolytic cleavages that give rise to amyloid-beta (Abeta), the amyloid-beta protein precursor (AbetaPP) is cleaved at Asp664 intracytoplasmically. This cleavage releases a cytotoxic peptide, APP-C31, removes AbetaPP-interaction motifs required for signaling and internalization, and is required for the generation of AD-like deficits in a mouse model of the disease. Although we and others had previously shown that Asp664 cleavage of AbetaPP is increased in AD brains, the distribution of the Asp664-cleaved forms of AbetaPP in non-diseased and AD brains at different ages had not been determined. Confirming previous reports, we found that Asp664-cleaved forms of AbetaPP were increased in neuronal cytoplasm and nuclei in early-stage AD brains but were absent in age-matched, non-diseased control brains and in late-stage AD brains. Remarkably, however, Asp664-cleaved AbetaPP was prominent in neuronal somata and in processes in entorhinal cortex and hippocampus of non-diseased human brains at ages <45 years. Our observations suggest that Asp664 cleavage of AbetaPP may be part of the normal proteolytic processing of AbetaPP in young (<45 years) human brain and that this cleavage is down-regulated with normal aging, but is aberrantly increased and altered in location in early AD.
Collapse
Affiliation(s)
- Surita Banwait
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hunsucker SW, Solomon B, Gawryluk J, Geiger JD, Vacano GN, Duncan MW, Patterson D. Assessment of post-mortem-induced changes to the mouse brain proteome. J Neurochem 2008; 105:725-37. [DOI: 10.1111/j.1471-4159.2007.05183.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
Bai Y, Markham K, Chen F, Weerasekera R, Watts J, Horne P, Wakutani Y, Bagshaw R, Mathews PM, Fraser PE, Westaway D, St. George-Hyslop P, Schmitt-Ulms G. The in Vivo Brain Interactome of the Amyloid Precursor Protein. Mol Cell Proteomics 2008; 7:15-34. [DOI: 10.1074/mcp.m700077-mcp200] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
41
|
Proteomics of Alzheimer's disease: Unveiling protein dysregulation in complex neuronal systems. Proteomics Clin Appl 2007; 1:1351-61. [DOI: 10.1002/prca.200700323] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Indexed: 11/07/2022]
|
42
|
Maltman DJ, Przyborski SA. Application of proteomic technology to neural stem cell science and neurology. FUTURE NEUROLOGY 2007. [DOI: 10.2217/14796708.2.3.285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
There is widespread recognition of the potential that stem cells hold for the treatment and repair of a large number of disorders affecting the human CNS. Therefore, stem cell research will go hand in hand with progress in specific areas of neuroscience. Proteomics has great potential to make important contributions to the basic understanding of neurological processes, and to deliver much needed cellular biomarkers in both of these fields. This review focuses on the importance of proteomic research in neuroscience, in particular the application of biomarker discovery in stem cells and degenerative diseases of the CNS.
Collapse
Affiliation(s)
- Daniel J Maltman
- University of Durham, School of Biological & Biomedical Science, South Road, Durham DH1 3LE, UK and, ReInnervate Limited, Old Shire Hall, Old Elvet, Durham DH1 3HP, UK
| | - Stefan A Przyborski
- University of Durham, School of Biological & Biomedical Science, South Road, Durham DH1 3LE, UK and, ReInnervate Limited, Old Shire Hall, Old Elvet, Durham DH1 3HP, UK
| |
Collapse
|
43
|
Cruz-Muros I, Afonso-Oramas D, Abreu P, Barroso-Chinea P, Rodríguez M, González MC, Hernández TG. Aging of the rat mesostriatal system: Differences between the nigrostriatal and the mesolimbic compartments. Exp Neurol 2007; 204:147-61. [PMID: 17112516 DOI: 10.1016/j.expneurol.2006.10.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Revised: 10/05/2006] [Accepted: 10/09/2006] [Indexed: 01/21/2023]
Abstract
The impairment of the mesostriatal dopaminergic system has been considered responsible for motor and affective disturbances associated with aging and a risk factor for Parkinson's disease. However, the basic mechanisms underlying this phenomenon are still unknown. Here we used biochemical, molecular and morphological techniques directed at detecting flaws in the dopamine synthesis route and signs of dopaminergic degeneration in the rat mesostriatal system during normal aging. We found two different age-related processes. One is characterized by a dopa decarboxylase decrease, and involves both the nigrostriatal and mesolimbic compartments, and is responsible for a moderate dopamine loss in the dorsal striatum, where other parameters of dopamine synthesis are not affected. The other is characterized by axonal degeneration with aggregation of phosphorylated forms of tyrosine hydroxylase (TH) and amyloid precursor protein in degenerate terminals, and alpha-synuclein in their original somata. This process is restricted to mesolimbic regions and is responsible for the decline of TH activity and l-dopa levels and the greater decrease in dopamine levels in this compartment. These findings suggest that both the nigrostriatal and the mesolimbic systems are vulnerable to aging, but in contrast to what occurs in Parkinson's disease, the mesolimbic system is more vulnerable to aging than the nigrostriatal one.
Collapse
Affiliation(s)
- Ignacio Cruz-Muros
- Department of Anatomy, Faculty of Medicine, University of La Laguna, Laguna, Tenerife, Spain
| | | | | | | | | | | | | |
Collapse
|
44
|
Sizova D, Charbaut E, Delalande F, Poirier F, High AA, Parker F, Van Dorsselaer A, Duchesne M, Diu-Hercend A. Proteomic analysis of brain tissue from an Alzheimer's disease mouse model by two-dimensional difference gel electrophoresis. Neurobiol Aging 2007; 28:357-70. [DOI: 10.1016/j.neurobiolaging.2006.01.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Revised: 01/17/2006] [Accepted: 01/31/2006] [Indexed: 11/28/2022]
|
45
|
Kimura N, Imamura O, Ono F, Terao K. Aging attenuates dynactin–dynein interaction: Down-regulation of dynein causes accumulation of endogenous tau and amyloid precursor protein in human neuroblastoma cells. J Neurosci Res 2007; 85:2909-16. [PMID: 17628503 DOI: 10.1002/jnr.21408] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Impaired axonal transport may promote pathogenesis in neurodegenerative disorders, such as Alzheimer's disease (AD). We previously showed that tau, amyloid precursor protein (APP), and intracellular amyloid beta-protein (Abeta) accumulate in the nerve-ending fraction of aged monkey brains, perhaps because of impaired axonal transport. In the present study, we assessed age-related changes of axonal transport motor proteins in aged monkey brains. Western blotting showed that kinesin, dynein, and dynactin (DYN) localizations dramatically changed with aging, and dynein level in nerve-ending fractions increased significantly. Coimmunoprecipitation analyses showed that DYN-dynein intermediate chain (DIC) interactions decreased, suggesting that age-related attenuation of this interaction may cause the impairment of dynein function. Moreover, RNAi-induced down-regulation of DIC in human neuroblastoma cells caused endogenous tau and APP to accumulate, and their subcellular localizations were also affected. Our findings suggest that aging attenuates DYN-DIC interaction, representing one of the risk factors for age-related impaired dynein function and even for accumulation of disease proteins.
Collapse
Affiliation(s)
- Nobuyuki Kimura
- Laboratory of Disease Control, Tsukuba Primate Research Center, National Institute of Biomedical Innovation, Ibaraki, Japan.
| | | | | | | |
Collapse
|
46
|
Kraemer B, Schellenberg GD. Using Caenorhabditis elegans models of neurodegenerative disease to identify neuroprotective strategies. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 77:219-46. [PMID: 17178476 DOI: 10.1016/s0074-7742(06)77007-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Brian Kraemer
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle Division, Seattle, Washington 98108, USA
| | | |
Collapse
|
47
|
He S, Wang Q, He J, Pu H, Yang W, Ji J. Proteomic analysis and comparison of the biopsy and autopsy specimen of human brain temporal lobe. Proteomics 2006; 6:4987-96. [PMID: 16912969 DOI: 10.1002/pmic.200600078] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The proteomic study on human temporal lobe can help us to understand the physiological function of CNS in normal as well as in pathological state. Proteomic tools are potent for the assessment of protein stability post mortem. In this pilot study, the human temporal lobe biopsy specimen with chronic pharmacoresistant temporal lobe epilepsy (TLE) and autopsy specimen in control were separated by 2-DE. Using MALDI-TOF-MS and MS/MS, 375 protein spots were identified which were the products of 267 genes. Six down-regulated and 23 up-regulated protein spots in the autopsy specimen were ascertained after the gel image analysis with the ImageMaster software. A number of proteins that include neurotransmitter metabolic and glycolytic enzymes, cytoprotective proteins and cytoskeleton were found decreased while the precursor of apolipoprotein A-I increased in the TLE brain. We tried several methods to prepare the protein samples and found that DNase and RNase treatment, ultracentrifugation and Amersham clean-up kit purification can improve gel separation quality. This work optimized the sample preparation method and constructed a primary protein database of human temporal lobe and found some proteins with remarkable level change probably involved in the post-mortem process and chronic pharmacoresistant TLE pathogenesis.
Collapse
Affiliation(s)
- Sizhi He
- Department of Biochemistry and Molecular Biology, College of Life Sciences, National Laboratory of Protein Engineering and Plant Genetic Engineering, Peking University, Beijing, PR China
| | | | | | | | | | | |
Collapse
|
48
|
Gozal YM, Cheng D, Duong DM, Lah JJ, Levey AI, Peng J. Merger of laser capture microdissection and mass spectrometry: a window into the amyloid plaque proteome. Methods Enzymol 2006; 412:77-93. [PMID: 17046653 DOI: 10.1016/s0076-6879(06)12006-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The occurrence of protein accumulation and aggregation in the brain is one of the pathological hallmarks of neurodegenerative diseases such as Alzheimer's disease (AD). Although it is instructive to analyze the aggregated proteins in the brain, biochemical purification and identification of these proteins have been challenging. Recent developments in laser capture microdissection (LCM) and mass spectrometry (MS) enable large-scale protein profiling of captured tissue samples. We present here the method of analyzing senile plaques from postmortem AD brains by coupling LCM and highly sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS). First, the senile plaques were stained with thioflavin-S and precisely isolated by adjusted laser beams under a microscope. Total proteins in the isolated tissues were extracted and resolved on an SDS gel. To identify all proteins in the samples, the gel was excised into multiple pieces followed by trypsin digestion. The resulting peptides were further separated by reverse-phase chromatography and analyzed by tandem mass spectrometry. A database search of acquired MS/MS spectra allowed the identification of hundreds to thousands of peptides/proteins in the original samples. Moreover, quantitative comparison of protein composites in different LCM samples could be achieved by MS strategies. For instance, the comparison between plaques and surrounding nonplaque tissues from the same specimen revealed tens of proteins specifically enriched in the plaques. Finally, the data were corroborated by independent experiments using the approach of immunohistochemistry. Taken together, the merger of LCM and MS is a powerful tool to probe the proteome of any given pathological lesions.
Collapse
Affiliation(s)
- Yair M Gozal
- Department of Human Genetics, Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, 30322, USA
| | | | | | | | | | | |
Collapse
|
49
|
Wojcik S, Engel WK, McFerrin J, Paciello O, Askanas V. AbetaPP-overexpression and proteasome inhibition increase alphaB-crystallin in cultured human muscle: relevance to inclusion-body myositis. Neuromuscul Disord 2006; 16:839-44. [PMID: 17056255 PMCID: PMC1976411 DOI: 10.1016/j.nmd.2006.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Revised: 08/04/2006] [Accepted: 08/17/2006] [Indexed: 11/24/2022]
Abstract
Amyloid-beta precursor protein (AbetaPP) and its fragment amyloid-beta (Abeta) are increased in s-IBM muscle fibers and appear to play an important role in the pathogenic cascade. alphaB-Crystallin (alphaBC) was shown immunohistochemically to be accumulated in s-IBM muscle fibers, but the stressor(s) influencing alphaBC accumulation was not identified. We now demonstrate, using our experimental IBM model based on genetic overexpression of AbetaPP into cultured normal human muscle fibers, that: (1) AbetaPP overexpression increased alphaBC 3.7-fold (p=0.025); (2) additional inhibition of proteasome with epoxomicin increased alphaBC 7-fold (p=0.002); and (3) alphaBC physically associated with AbetaPP and Abeta oligomers. We also show that in biopsied s-IBM muscle fibers, alphaBC was similarly increased 3-fold (p=0.025) and physically associated with AbetaPP and Abeta oligomers. We propose that increased AbetaPP is a stressor increasing alphaBC expression in s-IBM muscle fibers. Determining the consequences of alphaBC association with Abeta oligomers could have clinical therapeutic relevance.
Collapse
Affiliation(s)
- Slawomir Wojcik
- USC Neuromuscular Center, Department of Neurology, University of Southern California Keck School of Medicine, Good Samaritan Hospital, Los Angeles, CA 90017-1912, USA
| | | | | | | | | |
Collapse
|
50
|
Schulenborg T, Schmidt O, van Hall A, Meyer HE, Hamacher M, Marcus K. Proteomics in neurodegeneration – disease driven approaches. J Neural Transm (Vienna) 2006; 113:1055-73. [PMID: 16835692 DOI: 10.1007/s00702-006-0512-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Accepted: 04/05/2006] [Indexed: 10/24/2022]
Abstract
Proteins as a product from genetic information execute and determine how development, growth, aging and disease factors are orchestrated within the lifetime of an organism. Differential protein expression and/or modification are always context dependent i.e. they happen within a specific context of a tissue, organ, environmental situation and individual fate. Consequently, the function/dysfunction (in a certain disease) of a specific gene cannot be predicted comprehensively by its sequence only. Genetic information can only be understood when genes and proteins are analyzed in the context of the biological system and specific networks they are involved in. In regard to neurodegenerative diseases such as Alzheimer's (AD) and Parkinson's disease (PD) many proteins are known for long years to be the cause or the consequence of the pathomechanism of the respective disease. The treatment of these neurodegenerative diseases represents a major challenge for the pharmaceutical industry, whereas the understanding of their pathogenesis is still in its infancy. With the development of several powerful techniques for proteome analysis it is now possible to investigate the expression of thousands of proteins in single cells, tissues or whole organisms at the same time. These developments opened new doors in medical sciences, and identification of cellular alterations associated with e.g. neurodegeneration will result in the identification of novel diagnostic as well as therapeutic targets. In this review, general considerations and strategies of proteomics technologies, the advantages and challenges as well as the special needs for analyzing brain tissue in the context of AD and AD are described and summarized.
Collapse
Affiliation(s)
- T Schulenborg
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
| | | | | | | | | | | |
Collapse
|