401
|
Sanchez-Juan P, Bishop MT, Kovacs GG, Calero M, Aulchenko YS, Ladogana A, Boyd A, Lewis V, Ponto C, Calero O, Poleggi A, Carracedo Á, van der Lee SJ, Ströbel T, Rivadeneira F, Hofman A, Haïk S, Combarros O, Berciano J, Uitterlinden AG, Collins SJ, Budka H, Brandel JP, Laplanche JL, Pocchiari M, Zerr I, Knight RSG, Will RG, van Duijn CM. A genome wide association study links glutamate receptor pathway to sporadic Creutzfeldt-Jakob disease risk. PLoS One 2015; 10:e0123654. [PMID: 25918841 PMCID: PMC4412535 DOI: 10.1371/journal.pone.0123654] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/23/2015] [Indexed: 02/03/2023] Open
Abstract
We performed a genome-wide association (GWA) study in 434 sporadic Creutzfeldt-Jakob disease (sCJD) patients and 1939 controls from the United Kingdom, Germany and The Netherlands. The findings were replicated in an independent sample of 1109 sCJD and 2264 controls provided by a multinational consortium. From the initial GWA analysis we selected 23 SNPs for further genotyping in 1109 sCJD cases from seven different countries. Five SNPs were significantly associated with sCJD after correction for multiple testing. Subsequently these five SNPs were genotyped in 2264 controls. The pooled analysis, including 1543 sCJD cases and 4203 controls, yielded two genome wide significant results: rs6107516 (p-value=7.62x10-9) a variant tagging the prion protein gene (PRNP); and rs6951643 (p-value=1.66x10-8) tagging the Glutamate Receptor Metabotropic 8 gene (GRM8). Next we analysed the data stratifying by country of origin combining samples from the pooled analysis with genotypes from the 1000 Genomes Project and imputed genotypes from the Rotterdam Study (Total n=12967). The meta-analysis of the results showed that rs6107516 (p-value=3.00x10-8) and rs6951643 (p-value=3.91x10-5) remained as the two most significantly associated SNPs. Rs6951643 is located in an intronic region of GRM8, a gene that was additionally tagged by a cluster of 12 SNPs within our top100 ranked results. GRM8 encodes for mGluR8, a protein which belongs to the metabotropic glutamate receptor family, recently shown to be involved in the transduction of cellular signals triggered by the prion protein. Pathway enrichment analyses performed with both Ingenuity Pathway Analysis and ALIGATOR postulates glutamate receptor signalling as one of the main pathways associated with sCJD. In summary, we have detected GRM8 as a novel, non-PRNP, genome-wide significant marker associated with heightened disease risk, providing additional evidence supporting a role of glutamate receptors in sCJD pathogenesis.
Collapse
Affiliation(s)
- Pascual Sanchez-Juan
- Neurology Department, University Hospital “Marqués de Valdecilla”. Instituto de Investigación “Marqués de Valdecilla” IDIVAL and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED). Santander, Spain
| | - Matthew T. Bishop
- The National Creutzfeldt-Jakob disease Research and Surveillance Unit, University of Edinburgh, United Kingdom
| | - Gabor G. Kovacs
- Institute of Neurology, Medical University Vienna, Vienna, Austria
| | - Miguel Calero
- Chronic Disease Programme and CIBERNED. Carlos III Institute of Health. Madrid. Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Carlos III Institute of Health, Alzheimer Center Reina Sofia Foundation, Madrid, Spain
| | - Yurii S. Aulchenko
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Anna Ladogana
- Department of Cell Biology and Neurosciences Instituto Superiore di Sanità, Roma, Italy
| | - Alison Boyd
- Department of Pathology, The University of Melbourne, Parkville, 3010, Australia
| | - Victoria Lewis
- Department of Pathology, The University of Melbourne, Parkville, 3010, Australia
| | - Claudia Ponto
- Department of Neurology, Clinical Dementia Centre, University Medical Center and German Center for Neurodegenerative Diseases (DZNE)—site Göttingen, Göttingen, Germany
| | - Olga Calero
- Chronic Disease Programme and CIBERNED. Carlos III Institute of Health. Madrid. Spain
| | - Anna Poleggi
- Department of Cell Biology and Neurosciences Instituto Superiore di Sanità, Roma, Italy
| | - Ángel Carracedo
- Fundación Pública Galega de Medicina Xenómica, CIBERER, Grupo de Medicina Xenómica-Universidad de Santiago de Compostela, Santiago de Compostela, Spain
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, KSA
| | - Sven J. van der Lee
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Thomas Ströbel
- Institute of Neurology, Medical University Vienna, Vienna, Austria
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Stéphane Haïk
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, and Inserm, U 1127, and CNRS UMR 7225, and ICM, F-75013, Paris, France; AP-HP, Hôpital de la Pitié Salpêtrière, Cellule Nationale de Référence des maladies de Creutzfeldt-Jakob, F-75013, Paris, France
| | - Onofre Combarros
- Neurology Department, University Hospital “Marqués de Valdecilla”. Instituto de Investigación “Marqués de Valdecilla” IDIVAL and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED). Santander, Spain
| | - José Berciano
- Neurology Department, University Hospital “Marqués de Valdecilla”. Instituto de Investigación “Marqués de Valdecilla” IDIVAL and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED). Santander, Spain
| | - Andre G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Steven J. Collins
- Department of Pathology, The University of Melbourne, Parkville, 3010, Australia
| | - Herbert Budka
- Institute of Neurology, Medical University Vienna, Vienna, Austria
| | - Jean-Philippe Brandel
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, and Inserm, U 1127, and CNRS UMR 7225, and ICM, F-75013, Paris, France; AP-HP, Hôpital de la Pitié Salpêtrière, Cellule Nationale de Référence des maladies de Creutzfeldt-Jakob, F-75013, Paris, France
| | - Jean Louis Laplanche
- Service de biochimie et biologie moleculaire, Laboratoire associé au CNR "ATNC", Hôpital Lariboisiére, AP-HP, Paris, France
| | - Maurizio Pocchiari
- Department of Cell Biology and Neurosciences Instituto Superiore di Sanità, Roma, Italy
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Centre, University Medical Center and German Center for Neurodegenerative Diseases (DZNE)—site Göttingen, Göttingen, Germany
| | - Richard S. G. Knight
- The National Creutzfeldt-Jakob disease Research and Surveillance Unit, University of Edinburgh, United Kingdom
| | - Robert G. Will
- The National Creutzfeldt-Jakob disease Research and Surveillance Unit, University of Edinburgh, United Kingdom
| | | |
Collapse
|
402
|
Nygaard HB, Wagner AF, Bowen GS, Good SP, MacAvoy MG, Strittmatter KA, Kaufman AC, Rosenberg BJ, Sekine-Konno T, Varma P, Chen K, Koleske AJ, Reiman EM, Strittmatter SM, van Dyck CH. A phase Ib multiple ascending dose study of the safety, tolerability, and central nervous system availability of AZD0530 (saracatinib) in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2015; 7:35. [PMID: 25874001 PMCID: PMC4396171 DOI: 10.1186/s13195-015-0119-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 03/16/2015] [Indexed: 12/31/2022]
Abstract
Introduction Despite significant progress, a disease-modifying therapy for Alzheimer’s disease (AD) has not yet been developed. Recent findings implicate soluble oligomeric amyloid beta as the most relevant protein conformation in AD pathogenesis. We recently described a signaling cascade whereby oligomeric amyloid beta binds to cellular prion protein on the neuronal cell surface, activating intracellular Fyn kinase to mediate synaptotoxicity. Fyn kinase has been implicated in AD pathophysiology both in in vitro models and in human subjects, and is a promising new therapeutic target for AD. Herein, we present a Phase Ib trial of the repurposed investigational drug AZD0530, a Src family kinase inhibitor specific for Fyn and Src kinase, for the treatment of patients with mild-to-moderate AD. Methods The study was a 4-week Phase Ib multiple ascending dose, randomized, double-blind, placebo-controlled trial of AZD0530 in AD patients with Mini-Mental State Examination (MMSE) scores ranging from 16 to 26. A total of 24 subjects were recruited in three sequential groups, with each randomized to receive oral AZD0530 at doses of 50 mg, 100 mg, 125 mg, or placebo daily for 4 weeks. The drug:placebo ratio was 3:1. Primary endpoints were safety, tolerability, and cerebrospinal fluid (CSF) penetration of AZD0530. Secondary endpoints included changes in clinical efficacy measures (Alzheimer’s Disease Assessment Scale – cognitive subscale, MMSE, Alzheimer’s Disease Cooperative Study – Activities of Daily Living Inventory, Neuropsychiatric Inventory, and Clinical Dementia Rating Scale – Sum of Boxes) and regional cerebral glucose metabolism measured by fluorodeoxyglucose positron emission tomography. Results AZD0530 was generally safe and well tolerated across doses. One subject receiving 125 mg of AZD0530 was discontinued from the study due to the development of congestive heart failure and atypical pneumonia, which were considered possibly related to the study drug. Plasma/CSF ratio of AZD0530 was 0.4. The 100 mg and 125 mg doses achieved CSF drug levels corresponding to brain levels that rescued memory deficits in transgenic mouse models. One-month treatment with AZD0530 had no significant effect on clinical efficacy measures or regional cerebral glucose metabolism. Conclusions AZD0530 is reasonably safe and well tolerated in patients with mild-to-moderate AD, achieving substantial central nervous system penetration with oral dosing at 100–125 mg. Targeting Fyn kinase may be a promising therapeutic approach in AD, and a larger Phase IIa clinical trial of AZD0530 for the treatment of patients with AD has recently launched. Trial registration ClinicalTrials.gov: NCT01864655. Registered 12 June 2014.
Collapse
Affiliation(s)
- Haakon B Nygaard
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, Connecticut USA ; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut USA ; Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Yale University School of Medicine, New Haven, Connecticut USA ; Current address: University of British Columbia, Division of Neurology, Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Allison F Wagner
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, Connecticut USA ; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut USA
| | - Garrett S Bowen
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, Connecticut USA ; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut USA
| | - Susan P Good
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, Connecticut USA ; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut USA
| | - Martha G MacAvoy
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, Connecticut USA ; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut USA
| | - Kurt A Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Yale University School of Medicine, New Haven, Connecticut USA
| | - Adam C Kaufman
- Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Yale University School of Medicine, New Haven, Connecticut USA
| | - Brian J Rosenberg
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut USA
| | - Tomoko Sekine-Konno
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut USA
| | - Pradeep Varma
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut USA
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, Arizona USA
| | - Anthony J Koleske
- Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Yale University School of Medicine, New Haven, Connecticut USA ; Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut USA
| | | | - Stephen M Strittmatter
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut USA ; Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Yale University School of Medicine, New Haven, Connecticut USA
| | - Christopher H van Dyck
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, Connecticut USA ; Department of Neurology, Yale University School of Medicine, New Haven, Connecticut USA ; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut USA
| |
Collapse
|
403
|
Posttraumatic stress disorder-like induction elevates β-amyloid levels, which directly activates corticotropin-releasing factor neurons to exacerbate stress responses. J Neurosci 2015; 35:2612-23. [PMID: 25673853 DOI: 10.1523/jneurosci.3333-14.2015] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent studies have found that those who suffer from posttraumatic stress disorder (PTSD) are more likely to experience dementia as they age, most often Alzheimer's disease (AD). These findings suggest that the symptoms of PTSD might have an exacerbating effect on AD progression. AD and PTSD might also share common susceptibility factors such that those who experience trauma-induced disease were already more likely to succumb to dementia with age. Here, we explored these two hypotheses using a mouse model of PTSD in wild-type and AD model animals. We found that expression of human familial AD mutations in amyloid precursor protein and presenilin 1 leads to sensitivity to trauma-induced PTSD-like changes in behavioral and endocrine stress responses. PTSD-like induction, in turn, chronically elevates levels of CSF β-amyloid (Aβ), exacerbating ongoing AD pathogenesis. We show that PTSD-like induction and Aβ elevation are dependent on corticotropin-releasing factor (CRF) receptor 1 signaling and an intact hypothalamic-pituitary-adrenal axis. Furthermore, we show that Aβ species can hyperexcite CRF neurons, providing a mechanism by which Aβ influences stress-related symptoms and PTSD-like phenotypes. Consistent with Aβ causing excitability of the stress circuitry, we attenuate PTSD-like phenotypes in vivo by lowering Aβ levels during PTSD-like trauma exposure. Together, these data demonstrate that exposure to PTSD-like trauma can drive AD pathogenesis, which directly perturbs CRF signaling, thereby enhancing chronic PTSD symptoms while increasing risk for AD-related dementia.
Collapse
|
404
|
Abstract
The most accredited (and fashionable) hypothesis of the pathogenesis of Alzheimer Disease (AD) sees accumulation of β-amyloid protein in the brain (in both soluble and insoluble forms) as a leading mechanism of neurotoxicity. How β-amyloid triggers the neurodegenerative disorder is at present unclear, but growing evidence suggests that a deregulation of Ca(2+) homeostasis and deficient Ca(2+) signalling may represent a fundamental pathogenic factor. Given that symptoms of AD are most likely linked to synaptic dysfunction (at the early stages) followed by neuronal loss (at later and terminal phases of the disease), the effects of β-amyloid have been mainly studied in neurones. Yet, it must be acknowledged that neuroglial cells, including astrocytes, contribute to pathological progression of most (if not all) neurological diseases. Here, we review the literature pertaining to changes in Ca(2+) signalling in astrocytes exposed to exogenous β-amyloid or in astrocytes from transgenic Alzheimer disease animals models, characterized by endogenous β-amyloidosis. Accumulated experimental data indicate deregulation of Ca(2+) homeostasis and signalling in astrocytes in AD, which should be given full pathogenetic consideration. Further studies are warranted to comprehend the role of deficient astroglial Ca(2+) signalling in the disease progression.
Collapse
|
405
|
Lourenco MV, Ferreira ST, De Felice FG. Neuronal stress signaling and eIF2α phosphorylation as molecular links between Alzheimer's disease and diabetes. Prog Neurobiol 2015; 129:37-57. [PMID: 25857551 DOI: 10.1016/j.pneurobio.2015.03.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/10/2015] [Accepted: 03/29/2015] [Indexed: 12/22/2022]
Abstract
Mounting evidence from clinical, epidemiological, neuropathology and preclinical studies indicates that mechanisms similar to those leading to peripheral metabolic deregulation in metabolic disorders, such as diabetes and obesity, take place in the brains of Alzheimer's disease (AD) patients. These include pro-inflammatory mechanisms, brain metabolic stress and neuronal insulin resistance. From a molecular and cellular perspective, recent progress has been made in unveiling novel pathways that act in an orchestrated way to cause neuronal damage and cognitive decline in AD. These pathways converge to the activation of neuronal stress-related protein kinases and excessive phosphorylation of eukaryotic translation initiation factor 2α (eIF2α-P), which plays a key role in control of protein translation, culminating in synapse dysfunction and memory loss. eIF2α-P signaling thus links multiple neuronal stress pathways to impaired neuronal function and neurodegeneration. Here, we present a critical analysis of recently discovered molecular mechanisms underlying impaired brain insulin signaling and metabolic stress, with emphasis on the role of stress kinase/eIF2α-P signaling as a hub that promotes brain and behavioral impairments in AD. Because very similar mechanisms appear to operate in peripheral metabolic deregulation in T2D and in brain defects in AD, we discuss the concept that targeting defective brain insulin signaling and neuronal stress mechanisms with anti-diabetes agents may be an attractive approach to fight memory decline in AD. We conclude by raising core questions that remain to be addressed toward the development of much needed therapeutic approaches for AD.
Collapse
Affiliation(s)
- Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| |
Collapse
|
406
|
Wang ZX, Tan L, Liu J, Yu JT. The Essential Role of Soluble Aβ Oligomers in Alzheimer's Disease. Mol Neurobiol 2015; 53:1905-1924. [PMID: 25833098 DOI: 10.1007/s12035-015-9143-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/18/2015] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by amyloid plaque and neurofibrillary tangles (NFT). With the finding that soluble nonfibrillar Aβ levels actually correlate strongly with the severity of the disease, the initial focus on amyloid plaques shifted to the contemporary concept that AD memory failure is caused by soluble Aβ oligomers. The soluble Aβ are known to be more neurotoxicthan fibrillar Aβ species. In this paper, we summarize the essential role of soluble Aβ oligomers in AD and discuss therapeutic strategies that target soluble Aβ oligomers.
Collapse
Affiliation(s)
- Zi-Xuan Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China.
| | - Jinyuan Liu
- Columbia College, Columbia University, New York, NY, USA
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China. .,Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA.
| |
Collapse
|
407
|
Raka F, Di Sebastiano AR, Kulhawy SC, Ribeiro FM, Godin CM, Caetano FA, Angers S, Ferguson SSG. Ca(2+)/calmodulin-dependent protein kinase II interacts with group I metabotropic glutamate and facilitates receptor endocytosis and ERK1/2 signaling: role of β-amyloid. Mol Brain 2015; 8:21. [PMID: 25885040 PMCID: PMC4378271 DOI: 10.1186/s13041-015-0111-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Agonist stimulation of Group I metabotropic glutamate receptors (mGluRs) initiates their coupling to the heterotrimeric G protein, Gαq/11, resulting in the activation of phospholipase C, the release of Ca(2+) from intracellular stores and the subsequent activation of protein kinase C. However, it is now recognized that mGluR5a also functions as a receptor for cellular prion protein (PrP(C)) and β-amyloid peptide (Aβ42) oligomers to facilitate intracellular signaling via the resulting protein complex. Intracellular mGluR5a signaling is also regulated by its association with a wide variety of intracellular regulation proteins. RESULTS In the present study, we utilized mass spectroscopy to identify calmodulin kinase IIα (CaMKIIα) as a protein that interacts with the second intracellular loop domain of mGluR5. We show that CaMKIIα interacts with both mGluR1a and mGluR5a in an agonist-independent manner and is co-immunoprecipitated with mGluR5a from hippocampal mouse brain. CaMKIIα positively regulates both mGluR1a and mGluR5a endocytosis, but selectively attenuates mGluR5a but not mGluR1a-stimulated ERK1/2 phosphorylation in a kinase activity-dependent manner. We also find that Aβ42 oligomers stimulate the association of CaMKIIα with mGluR5a and activate ERK1/2 in an mGluR5a-dependent manner. However, Aβ42 oligomer-stimulated ERK1/2 phosphorylation is not regulated by mGluR5a/CaMKIIα interactions suggesting that agonist and Aβ42 oligomers stabilize distinct mGluR5a activation states that are differentially regulated by CaMKIIα. The expression of both mGluR5a and PrP(C) together, but not alone resulted in the agonist-stimulated subcellular distribution of CaMKIIα into cytoplasmic puncta. CONCLUSIONS Taken together these results indicate that CaMKIIα selectively regulates mGluR1a and mGluR5a ERK1/2 signaling. As mGluR5 and CaMKIIα are involved in learning and memory and Aβ and mGluR5 are implicated in Alzheimer's disease, results of these studies could provide insight into potential pharmacological targets for treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Fitore Raka
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Andrea R Di Sebastiano
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Stephanie C Kulhawy
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Fabiola M Ribeiro
- Departamento de Bioquimica e Imunologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Christina M Godin
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Fabiana A Caetano
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| | - Stephane Angers
- Leslie Dan Faculty of Pharmacy, University of Toronto, Room 901 144 College Street, Toronto, Ontario, Canada.
| | - Stephen S G Ferguson
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, and the Department of Physiology and Pharmacology, University of Western Ontario, 100 Perth Dr. London, Ontario, N6A 5K8, Canada.
| |
Collapse
|
408
|
Hamilton A, Zamponi GW, Ferguson SSG. Glutamate receptors function as scaffolds for the regulation of β-amyloid and cellular prion protein signaling complexes. Mol Brain 2015; 8:18. [PMID: 25888324 PMCID: PMC4395978 DOI: 10.1186/s13041-015-0107-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/27/2015] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects 36 million people worldwide, but currently has no effective treatment options. One of the original hallmarks of AD are plaques comprised of beta amyloid (Aβ) and neurofibrillary tangles comprised of phosphorylated Tau protein. However, it is soluble oligomeric Aβ which is more closely correlated with cognitive decline and is therefore considered to be the neurotoxic species. Oligomeric Aβ has recently been shown to form complexes with the glycosylphosphatidylinositol (GPI)-anchored membrane protein, cellular prion protein (PrP(c)), and these complexes are believed to play an important role in the progression of AD pathogenesis. Glutamate, the major excitatory neurotransmitter is responsible for mediating learning and memory under normal physiological conditions. However, the dysregulation of glutamatergic signaling has also been implicated in a number of neurodegenerative diseases including AD. Glutamate acts via both ionotropic glutamate receptors (iGluR) and metabotropic glutamate receptors (mGluR), each of which have been implicated in AD. There is now growing evidence to suggest that mGluR5 may contribute the AD pathogenesis by acting as scaffolds for the PrP(c)/Aβ oligomer complex, enabling the propagation of neurotoxic signaling in AD. In addition, PrP(c) and Aβ oligomer signaling via NMDARs may also contribute to AD pathology. The current review overviews our current understanding of the role of PrP(c) and Aβ oligomers in regulating glutamate receptor signaling, as well as highlights the importance of understanding these signaling complexes to develop more effective therapeutic strategies to treat AD.
Collapse
Affiliation(s)
- Alison Hamilton
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, Ontario, N6A 5 K8, Canada.
- Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario, Canada.
| | - Gerald W Zamponi
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Stephen S G Ferguson
- J. Allyn Taylor Centre for Cell Biology, Robarts Research Institute, University of Western Ontario, 100 Perth Dr, London, Ontario, N6A 5 K8, Canada.
- Department of Physiology & Pharmacology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
409
|
Nasica-Labouze J, Nguyen PH, Sterpone F, Berthoumieu O, Buchete NV, Coté S, De Simone A, Doig AJ, Faller P, Garcia A, Laio A, Li MS, Melchionna S, Mousseau N, Mu Y, Paravastu A, Pasquali S, Rosenman DJ, Strodel B, Tarus B, Viles JH, Zhang T, Wang C, Derreumaux P. Amyloid β Protein and Alzheimer's Disease: When Computer Simulations Complement Experimental Studies. Chem Rev 2015; 115:3518-63. [PMID: 25789869 DOI: 10.1021/cr500638n] [Citation(s) in RCA: 499] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jessica Nasica-Labouze
- †Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | - Phuong H Nguyen
- †Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | - Fabio Sterpone
- †Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | - Olivia Berthoumieu
- ‡LCC (Laboratoire de Chimie de Coordination), CNRS, Université de Toulouse, Université Paul Sabatier (UPS), Institut National Polytechnique de Toulouse (INPT), 205 route de Narbonne, BP 44099, Toulouse F-31077 Cedex 4, France
| | | | - Sébastien Coté
- ∥Département de Physique and Groupe de recherche sur les protéines membranaires (GEPROM), Université de Montréal, C.P. 6128, succursale Centre-ville, Montréal, Québec H3C 3T5, Canada
| | - Alfonso De Simone
- ⊥Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Andrew J Doig
- #Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Peter Faller
- ‡LCC (Laboratoire de Chimie de Coordination), CNRS, Université de Toulouse, Université Paul Sabatier (UPS), Institut National Polytechnique de Toulouse (INPT), 205 route de Narbonne, BP 44099, Toulouse F-31077 Cedex 4, France
| | | | - Alessandro Laio
- ○The International School for Advanced Studies (SISSA), Via Bonomea 265, 34136 Trieste, Italy
| | - Mai Suan Li
- ◆Institute of Physics, Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland.,¶Institute for Computational Science and Technology, SBI Building, Quang Trung Software City, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City, Vietnam
| | - Simone Melchionna
- ⬠Instituto Processi Chimico-Fisici, CNR-IPCF, Consiglio Nazionale delle Ricerche, 00185 Roma, Italy
| | | | - Yuguang Mu
- ▲School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | - Anant Paravastu
- ⊕National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee, Florida 32310, United States
| | - Samuela Pasquali
- †Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | | | - Birgit Strodel
- △Institute of Complex Systems: Structural Biochemistry (ICS-6), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Bogdan Tarus
- †Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | - John H Viles
- ▼School of Biological and Chemical Sciences, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Tong Zhang
- †Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 rue Pierre et Marie Curie, 75005 Paris, France.,▲School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore
| | | | - Philippe Derreumaux
- †Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 rue Pierre et Marie Curie, 75005 Paris, France.,□Institut Universitaire de France, 75005 Paris, France
| |
Collapse
|
410
|
Martin HGS, Lassalle O, Brown JT, Manzoni OJ. Age-Dependent Long-Term Potentiation Deficits in the Prefrontal Cortex of the Fmr1 Knockout Mouse Model of Fragile X Syndrome. Cereb Cortex 2015; 26:2084-2092. [PMID: 25750254 DOI: 10.1093/cercor/bhv031] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The most common inherited monogenetic cause of intellectual disability is Fragile X syndrome (FXS). The clinical symptoms of FXS evolve with age during adulthood; however, neurophysiological data exploring this phenomenon are limited. The Fmr1 knockout (Fmr1KO) mouse models FXS, but studies in these mice of prefrontal cortex (PFC) function are underrepresented, and aging linked data are absent. We studied synaptic physiology and activity-dependent synaptic plasticity in the medial PFC of Fmr1KO mice from 2 to 12 months. In young adult Fmr1KO mice, NMDA receptor (NMDAR)-mediated long-term potentiation (LTP) is intact; however, in 12-month-old mice this LTP is impaired. In parallel, there was an increase in the AMPAR/NMDAR ratio and a concomitant decrease of synaptic NMDAR currents in 12-month-old Fmr1KO mice. We found that acute pharmacological blockade of mGlu5 receptor in 12-month-old Fmr1KO mice restored a normal AMPAR/NMDAR ratio and LTP. Taken together, the data reveal an age-dependent deficit in LTP in Fmr1KO mice, which may correlate to some of the complex age-related deficits in FXS.
Collapse
Affiliation(s)
- Henry G S Martin
- INSERM U901, Marseille 13009, France.,INMED, Marseille 13009, France.,Université de Aix-Marseille, UMR S901, Marseille, France
| | - Olivier Lassalle
- INSERM U901, Marseille 13009, France.,INMED, Marseille 13009, France.,Université de Aix-Marseille, UMR S901, Marseille, France
| | - Jonathan T Brown
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Olivier J Manzoni
- INSERM U901, Marseille 13009, France.,INMED, Marseille 13009, France.,Université de Aix-Marseille, UMR S901, Marseille, France
| |
Collapse
|
411
|
Puig B, Altmeppen H, Glatzel M. The GPI-anchoring of PrP: implications in sorting and pathogenesis. Prion 2015; 8:11-8. [PMID: 24509692 DOI: 10.4161/pri.27892] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cellular prion protein (PrP(C)) is an N-glycosylated GPI-anchored protein usually present in lipid rafts with numerous putative functions. When it changes its conformation to a pathological isoform (then referred to as PrP(Sc)), it is an essential part of the prion, the agent causing fatal and transmissible neurodegenerative prion diseases. There is growing evidence that toxicity and neuronal damage on the one hand and propagation/infectivity on the other hand are two distinct processes of the disease and that the GPI-anchor attachment of PrP(C) and PrP(Sc) plays an important role in protein localization and in neurotoxicity. Here we review how the signal sequence of the GPI-anchor matters in PrP(C) localization, how an altered cellular localization of PrP(C) or differences in GPI-anchor composition can affect prion infection, and we discuss through which mechanisms changes on the anchorage of PrP(C) can modify the disease process.
Collapse
|
412
|
Béland M, Roucou X. Taking advantage of physiological proteolytic processing of the prion protein for a therapeutic perspective in prion and Alzheimer diseases. Prion 2015; 8:106-10. [PMID: 24335160 DOI: 10.4161/pri.27438] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Prion and Alzheimer diseases are fatal neurodegenerative diseases caused by misfolding and aggregation of the cellular prion protein (PrP(C)) and the β-amyloid peptide, respectively. Soluble oligomeric species rather than large aggregates are now believed to be neurotoxic. PrP(C) undergoes three proteolytic cleavages as part of its natural life cycle, α-cleavage, β-cleavage, and ectodomain shedding. Recent evidences demonstrate that the resulting secreted PrP(C) molecules might represent natural inhibitors against soluble toxic species. In this mini-review, we summarize recent observations suggesting the potential benefit of using PrP(C)-derived molecules as therapeutic agents in prion and Alzheimer diseases.
Collapse
|
413
|
Slingshot-Cofilin activation mediates mitochondrial and synaptic dysfunction via Aβ ligation to β1-integrin conformers. Cell Death Differ 2015; 22:921-34. [PMID: 25698445 PMCID: PMC4423195 DOI: 10.1038/cdd.2015.5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/19/2014] [Accepted: 12/16/2014] [Indexed: 02/08/2023] Open
Abstract
The accumulation of amyloid-β protein (Aβ) is an early event associated with synaptic and mitochondrial damage in Alzheimer's disease (AD). Recent studies have implicated the filamentous actin (F-actin) severing protein, Cofilin, in synaptic remodeling, mitochondrial dysfunction, and AD pathogenesis. However, whether Cofilin is an essential component of the AD pathogenic process and how Aβ impinges its signals to Cofilin from the neuronal surface are unknown. In this study, we found that Aβ42 oligomers (Aβ42O, amyloid-β protein 1–42 oligomers) bind with high affinity to low or intermediate activation conformers of β1-integrin, resulting in the loss of surface β1-integrin and activation of Cofilin via Slingshot homology-1 (SSH1) activation. Specifically, conditional loss of β1-integrin prevented Aβ42O-induced Cofilin activation, and allosteric modulation or activation of β1-integrin significantly reduced Aβ42O binding to neurons while blocking Aβ42O-induced reactive oxygen species (ROS) production, mitochondrial dysfunction, depletion of F-actin/focal Vinculin, and apoptosis. Cofilin, in turn, was required for Aβ42O-induced loss of cell surface β1-integrin, disruption of F-actin/focal Talin–Vinculin, and depletion of F-actin-associated postsynaptic proteins. SSH1 reduction, which mitigated Cofilin activation, prevented Aβ42O-induced mitochondrial Cofilin translocation and apoptosis, while AD brain mitochondria contained significantly increased activated/oxidized Cofilin. In mechanistic support in vivo, AD mouse model (APP (amyloid precursor protein)/PS1) brains contained increased SSH1/Cofilin and decreased SSH1/14-3-3 complexes, indicative of SSH1–Cofilin activation via release of SSH1 from 14-3-3. Finally, genetic reduction in Cofilin rescued APP/Aβ-induced synaptic protein loss and gliosis in vivo as well as deficits in long-term potentiation (LTP) and contextual memory in APP/PS1 mice. These novel findings therefore implicate the essential involvement of the β1-integrin–SSH1–Cofilin pathway in mitochondrial and synaptic dysfunction in AD.
Collapse
|
414
|
Wang H. Fragile X mental retardation protein: from autism to neurodegenerative disease. Front Cell Neurosci 2015; 9:43. [PMID: 25729352 PMCID: PMC4325920 DOI: 10.3389/fncel.2015.00043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 01/28/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Hansen Wang
- Faculty of Medicine, University of Toronto Toronto, ON, Canada
| |
Collapse
|
415
|
Aβ42 oligomers selectively disrupt neuronal calcium release. Neurobiol Aging 2015; 36:877-85. [PMID: 25453559 DOI: 10.1016/j.neurobiolaging.2014.10.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/09/2014] [Accepted: 10/14/2014] [Indexed: 12/30/2022]
|
416
|
Viola KL, Klein WL. Amyloid β oligomers in Alzheimer's disease pathogenesis, treatment, and diagnosis. Acta Neuropathol 2015; 129:183-206. [PMID: 25604547 DOI: 10.1007/s00401-015-1386-3] [Citation(s) in RCA: 477] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/11/2015] [Accepted: 01/11/2015] [Indexed: 12/22/2022]
Abstract
Protein aggregation is common to dozens of diseases including prionoses, diabetes, Parkinson's and Alzheimer's. Over the past 15 years, there has been a paradigm shift in understanding the structural basis for these proteinopathies. Precedent for this shift has come from investigation of soluble Aβ oligomers (AβOs), toxins now widely regarded as instigating neuron damage leading to Alzheimer's dementia. Toxic AβOs accumulate in AD brain and constitute long-lived alternatives to the disease-defining Aβ fibrils deposited in amyloid plaques. Key experiments using fibril-free AβO solutions demonstrated that while Aβ is essential for memory loss, the fibrillar Aβ in amyloid deposits is not the agent. The AD-like cellular pathologies induced by AβOs suggest their impact provides a unifying mechanism for AD pathogenesis, explaining why early stage disease is specific for memory and accounting for major facets of AD neuropathology. Alternative ideas for triggering mechanisms are being actively investigated. Some research favors insertion of AβOs into membrane, while other evidence supports ligand-like accumulation at particular synapses. Over a dozen candidate toxin receptors have been proposed. AβO binding triggers a redistribution of critical synaptic proteins and induces hyperactivity in metabotropic and ionotropic glutamate receptors. This leads to Ca(2+) overload and instigates major facets of AD neuropathology, including tau hyperphosphorylation, insulin resistance, oxidative stress, and synapse loss. Because different species of AβOs have been identified, a remaining question is which oligomer is the major pathogenic culprit. The possibility has been raised that more than one species plays a role. Despite some key unknowns, the clinical relevance of AβOs has been established, and new studies are beginning to point to co-morbidities such as diabetes and hypercholesterolemia as etiological factors. Because pathogenic AβOs appear early in the disease, they offer appealing targets for therapeutics and diagnostics. Promising therapeutic strategies include use of CNS insulin signaling enhancers to protect against the presence of toxins and elimination of the toxins through use of highly specific AβO antibodies. An AD-dependent accumulation of AβOs in CSF suggests their potential use as biomarkers and new AβO probes are opening the door to brain imaging. Overall, current evidence indicates that Aβ oligomers provide a substantive molecular basis for the cause, treatment and diagnosis of Alzheimer's disease.
Collapse
|
417
|
Chumakov I, Nabirotchkin S, Cholet N, Milet A, Boucard A, Toulorge D, Pereira Y, Graudens E, Traoré S, Foucquier J, Guedj M, Vial E, Callizot N, Steinschneider R, Maurice T, Bertrand V, Scart-Grès C, Hajj R, Cohen D. Combining two repurposed drugs as a promising approach for Alzheimer's disease therapy. Sci Rep 2015; 5:7608. [PMID: 25566747 PMCID: PMC5378993 DOI: 10.1038/srep07608] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/19/2014] [Indexed: 02/08/2023] Open
Abstract
Alzheimer disease (AD) represents a major medical problem where mono-therapeutic interventions demonstrated only a limited efficacy so far. We explored the possibility of developing a combinational therapy that might prevent the degradation of neuronal and endothelial structures in this disease. We argued that the distorted balance between excitatory (glutamate) and inhibitory (GABA/glycine) systems constitutes a therapeutic target for such intervention. We found that a combination of two approved drugs – acamprosate and baclofen – synergistically protected neurons and endothelial structures in vitro against amyloid-beta (Aβ) oligomers. The neuroprotective effects of these drugs were mediated by modulation of targets in GABA/glycinergic and glutamatergic pathways. In vivo, the combination alleviated cognitive deficits in the acute Aβ25–35 peptide injection model and in the mouse mutant APP transgenic model. Several patterns altered in AD were also synergistically normalised. Our results open up the possibility for a promising therapeutic approach for AD by combining repurposed drugs.
Collapse
Affiliation(s)
- Ilya Chumakov
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Nathalie Cholet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aude Milet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aurélie Boucard
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Damien Toulorge
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Yannick Pereira
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Esther Graudens
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Sory Traoré
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Julie Foucquier
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Mickael Guedj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Emmanuel Vial
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | | | - Tangui Maurice
- 1] Université de Montpellier 2, 34095 Montpellier, France; Inserm, U710, 34095 Montpellier, France; EPHE, 75017 Paris, France [2] Amylgen, 2196 bd de la Lironde, 34980 Montferrier-sur-Lez, France
| | - Viviane Bertrand
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Rodolphe Hajj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Daniel Cohen
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| |
Collapse
|
418
|
Zali H, Zamanian-Azodi M, Rezaei Tavirani M, Akbar-zadeh Baghban A. Protein Drug Targets of Lavandula angustifolia on treatment of Rat Alzheimer's Disease. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2015; 14:291-302. [PMID: 25561935 PMCID: PMC4277642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Different treatment strategies of Alzheimer's disease (AD) are being studied for treating or slowing the progression of AD. Many pharmaceutically important regulation systems operate through proteins as drug targets. Here, we investigate the drug target proteins in beta-amyloid (Aβ) injected rat hippocampus treated with Lavandula angustifolia (LA) by proteomics techniques. The reported study showed that lavender extract (LE) improves the spatial performance in AD animal model by diminishing Aβ production in histopathology of hippocampus, so in this study neuroprotective proteins expressed in Aβ injected rats treated with LE were scrutinized. Rats were divided into three groups including normal, Aβ injected, and Aβ injected that was treated with LE. Protein expression profiles of hippocampus tissue were determined by two-dimensional electrophoresis (2DE) method and dysregulated proteins such as Snca, NF-L, Hspa5, Prdx2, Apoa1, and Atp5a1were identified by MALDI-TOF/TOF. KEGG pathway and gene ontology (GO) categories were used by searching DAVID Bioinformatics Resources. All detected protein spots were used to determine predictedinteractions with other proteins in STRING online database. Different isoforms of important protein, Snca that exhibited neuroprotective effects by anti-apoptotic properties were expressed. NF-L involved in the maintenance of neuronal caliber. Hspa5 likewise Prdx2 displays as anti-apoptotic protein that Prdx2 also involved in the neurotrophic effects. Apoa1 has anti-inflammatory activity and Atp5a1, produces ATP from ADP. To sum up, these proteins as potential drug targets were expressed in hippocampus in response to effective components in LA may have therapeutic properties for the treatment of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Hakimeh Zali
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mona Zamanian-Azodi
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran. ,E-mail:
| | | |
Collapse
|
419
|
Teich AF, Nicholls RE, Puzzo D, Fiorito J, Purgatorio R, Fa’ M, Arancio O. Synaptic therapy in Alzheimer's disease: a CREB-centric approach. Neurotherapeutics 2015; 12:29-41. [PMID: 25575647 PMCID: PMC4322064 DOI: 10.1007/s13311-014-0327-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Therapeutic attempts to cure Alzheimer's disease (AD) have failed, and new strategies are desperately needed. Motivated by this reality, many laboratories (including our own) have focused on synaptic dysfunction in AD because synaptic changes are highly correlated with the severity of clinical dementia. In particular, memory formation is accompanied by altered synaptic strength, and this phenomenon (and its dysfunction in AD) has been a recent focus for many laboratories. The molecule cyclic adenosine monophosphate response element-binding protein (CREB) is at a central converging point of pathways and mechanisms activated during the processes of synaptic strengthening and memory formation, as CREB phosphorylation leads to transcription of memory-associated genes. Disruption of these mechanisms in AD results in a reduction of CREB activation with accompanying memory impairment. Thus, it is likely that strategies aimed at these mechanisms will lead to future therapies for AD. In this review, we will summarize literature that investigates 5 possible therapeutic pathways for rescuing synaptic dysfunction in AD: 4 enzymatic pathways that lead to CREB phosphorylation (the cyclic adenosine monophosphate cascade, the serine/threonine kinases extracellular regulated kinases 1 and 2, the nitric oxide cascade, and the calpains), as well as histone acetyltransferases and histone deacetylases (2 enzymes that regulate the histone acetylation necessary for gene transcription).
Collapse
Affiliation(s)
- Andrew F. Teich
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Russell E. Nicholls
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Daniela Puzzo
- />Department of Bio-Medical Sciences, Section of Physiology, University of Catania, Catania, 95125 Italy
| | - Jole Fiorito
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Rosa Purgatorio
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Mauro Fa’
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Ottavio Arancio
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| |
Collapse
|
420
|
Peters C, Espinoza MP, Gallegos S, Opazo C, Aguayo LG. Alzheimer's Aβ interacts with cellular prion protein inducing neuronal membrane damage and synaptotoxicity. Neurobiol Aging 2014; 36:1369-77. [PMID: 25599875 DOI: 10.1016/j.neurobiolaging.2014.11.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 11/06/2014] [Accepted: 11/25/2014] [Indexed: 12/31/2022]
Abstract
A major feature of Alzheimer's disease is the accumulation of β-amyloid (Aβ) peptide in the brain. Recent studies have indicated that Aβ oligomers (Aβo) can interact with the cellular prion protein (PrPc). Therefore, this interaction might be driving some of Aβ toxic effects in the synaptic region. In the present study, we report that Aβo binds to PrPc in the neuronal membrane playing a role on toxic effects induced by Aβ. Phospholipase C-enzymatic cleavage of PrPc from the plasma membrane attenuated the association of Aβo to the neurons. Furthermore, an anti-PrP antibody (6D11) decreased the association of Aβo to hippocampal neurons with a concomitant reduction in Aβo and PrPc co-localization. Interestingly, this antibody blocked the increase in membrane conductance and intracellular calcium induced by Aβo. Thus, the data indicate that PrPc plays a role on the membrane perforations produced by Aβo, the increase in calcium ions and the release of synaptic vesicles that subsequently leads to synaptic failure. Future studies blocking Aβo interaction with PrPc could be important for the discovery of new therapeutic strategies for Alzheimer's disease.
Collapse
Affiliation(s)
- Christian Peters
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - María Paz Espinoza
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Scarlet Gallegos
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile
| | - Carlos Opazo
- Oxidation Biology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
421
|
The Binding Receptors of Aβ: an Alternative Therapeutic Target for Alzheimer's Disease. Mol Neurobiol 2014; 53:455-471. [PMID: 25465238 DOI: 10.1007/s12035-014-8994-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/06/2014] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, which causes the deterioration of memory and other cognitive abilities of the elderly. Previous lines of research have shown that Aβ is an essential factor in AD pathology and the soluble oligomeric species of Aβ peptide is presumed to be the drivers of synaptic impairment in AD. However, the exact mechanisms underlying Aβ-induced synapse dysfunction are still not fully understood. Recently, increasing evidence suggests that some potential receptors which bind specifically with Aβ may play important roles in inducing the toxicity of the neurons in AD pathology. These receptors include the cellular prion protein (PrPc), the α7 nicotinic acetylcholine receptor (α7nAChR), the p75 neurotrophin receptor (p75(NTR)), the beta-adrenergic receptors (β-ARs), the Eph receptors, the paired immunoglobulin-like receptor B (PirB), the PirB's human ortholog receptor (LilrB2), and the Fcγ receptor II-b (FcγRIIb). This review summarizes the characters of these prominent receptors and how the bindings of them with Aβ inhibit the LTP, decrease the number of dendritic spine, damage the neurons, and so on in AD pathogenesis. Blocking or rescuing these receptors may have significant importance for AD treatments.
Collapse
|
422
|
Kam TI, Gwon Y, Jung YK. Amyloid beta receptors responsible for neurotoxicity and cellular defects in Alzheimer's disease. Cell Mol Life Sci 2014; 71:4803-13. [PMID: 25151011 PMCID: PMC11113744 DOI: 10.1007/s00018-014-1706-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/24/2014] [Accepted: 08/13/2014] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Although a major cause of AD is the accumulation of amyloid-β (Aβ) peptide that induces neuronal loss and cognitive impairments, our understanding of its neurotoxic mechanisms is limited. Recent studies have identified putative Aβ-binding receptors that mediate Aβ neurotoxicity in cells and models of AD. Once Aβ interacts with a receptor, a toxic signal is transduced into neurons, resulting in cellular defects including endoplasmic reticulum stress and mitochondrial dysfunction. In addition, Aβ can also be internalized into neurons through unidentified Aβ receptors and induces malfunction of subcellular organelles, which explains some part of Aβ neurotoxicity. Understanding the neurotoxic signaling initiated by Aβ-receptor binding and cellular defects provide insight into new therapeutic windows for AD. In the present review, we summarize the findings on Aβ-binding receptors and the neurotoxicity of oligomeric Aβ.
Collapse
Affiliation(s)
- Tae-In Kam
- Global Research Laboratory, School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-747 Korea
| | - Youngdae Gwon
- Global Research Laboratory, School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-747 Korea
| | - Yong-Keun Jung
- Global Research Laboratory, School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-747 Korea
| |
Collapse
|
423
|
Jong YJI, Sergin I, Purgert CA, O'Malley KL. Location-dependent signaling of the group 1 metabotropic glutamate receptor mGlu5. Mol Pharmacol 2014; 86:774-85. [PMID: 25326002 PMCID: PMC4244594 DOI: 10.1124/mol.114.094763] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022] Open
Abstract
Although G protein-coupled receptors are primarily known for converting extracellular signals into intracellular responses, some receptors, such as the group 1 metabotropic glutamate receptor, mGlu5, are also localized on intracellular membranes where they can mediate both overlapping and unique signaling effects. Thus, besides "ligand bias," whereby a receptor's signaling modality can shift from G protein dependence to independence, canonical mGlu5 receptor signaling can also be influenced by "location bias" (i.e., the particular membrane and/or cell type from which it signals). Because mGlu5 receptors play important roles in both normal development and in disorders such as Fragile X syndrome, autism, epilepsy, addiction, anxiety, schizophrenia, pain, dyskinesias, and melanoma, a large number of drugs are being developed to allosterically target this receptor. Therefore, it is critical to understand how such drugs might be affecting mGlu5 receptor function on different membranes and in different brain regions. Further elucidation of the site(s) of action of these drugs may determine which signal pathways mediate therapeutic efficacy.
Collapse
Affiliation(s)
- Yuh-Jiin I Jong
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Ismail Sergin
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Carolyn A Purgert
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Karen L O'Malley
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
424
|
Izzo NJ, Staniszewski A, To L, Fa M, Teich AF, Saeed F, Wostein H, Walko T, Vaswani A, Wardius M, Syed Z, Ravenscroft J, Mozzoni K, Silky C, Rehak C, Yurko R, Finn P, Look G, Rishton G, Safferstein H, Miller M, Johanson C, Stopa E, Windisch M, Hutter-Paier B, Shamloo M, Arancio O, LeVine H, Catalano SM. Alzheimer's therapeutics targeting amyloid beta 1-42 oligomers I: Abeta 42 oligomer binding to specific neuronal receptors is displaced by drug candidates that improve cognitive deficits. PLoS One 2014; 9:e111898. [PMID: 25390368 PMCID: PMC4229098 DOI: 10.1371/journal.pone.0111898] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 10/02/2014] [Indexed: 01/09/2023] Open
Abstract
Synaptic dysfunction and loss caused by age-dependent accumulation of synaptotoxic beta amyloid (Abeta) 1-42 oligomers is proposed to underlie cognitive decline in Alzheimer's disease (AD). Alterations in membrane trafficking induced by Abeta oligomers mediates reduction in neuronal surface receptor expression that is the basis for inhibition of electrophysiological measures of synaptic plasticity and thus learning and memory. We have utilized phenotypic screens in mature, in vitro cultures of rat brain cells to identify small molecules which block or prevent the binding and effects of Abeta oligomers. Synthetic Abeta oligomers bind saturably to a single site on neuronal synapses and induce deficits in membrane trafficking in neuronal cultures with an EC50 that corresponds to its binding affinity. The therapeutic lead compounds we have found are pharmacological antagonists of Abeta oligomers, reducing the binding of Abeta oligomers to neurons in vitro, preventing spine loss in neurons and preventing and treating oligomer-induced deficits in membrane trafficking. These molecules are highly brain penetrant and prevent and restore cognitive deficits in mouse models of Alzheimer's disease. Counter-screening these compounds against a broad panel of potential CNS targets revealed they are highly potent and specific ligands of the sigma-2/PGRMC1 receptor. Brain concentrations of the compounds corresponding to greater than 80% receptor occupancy at the sigma-2/PGRMC1 receptor restore cognitive function in transgenic hAPP Swe/Ldn mice. These studies demonstrate that synthetic and human-derived Abeta oligomers act as pharmacologically-behaved ligands at neuronal receptors--i.e. they exhibit saturable binding to a target, they exert a functional effect related to their binding and their displacement by small molecule antagonists blocks their functional effect. The first-in-class small molecule receptor antagonists described here restore memory to normal in multiple AD models and sustain improvement long-term, representing a novel mechanism of action for disease-modifying Alzheimer's therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Izzo
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Agnes Staniszewski
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Lillian To
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Mauro Fa
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Andrew F. Teich
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Faisal Saeed
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harrison Wostein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Thomas Walko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Anisha Vaswani
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Meghan Wardius
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Zanobia Syed
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Jessica Ravenscroft
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Kelsie Mozzoni
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Colleen Silky
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Patricia Finn
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gilbert Rishton
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Hank Safferstein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Miles Miller
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Conrad Johanson
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Edward Stopa
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | | | | | - Mehrdad Shamloo
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harry LeVine
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Susan M. Catalano
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
425
|
Izzo NJ, Xu J, Zeng C, Kirk MJ, Mozzoni K, Silky C, Rehak C, Yurko R, Look G, Rishton G, Safferstein H, Cruchaga C, Goate A, Cahill MA, Arancio O, Mach RH, Craven R, Head E, LeVine H, Spires-Jones TL, Catalano SM. Alzheimer's therapeutics targeting amyloid beta 1-42 oligomers II: Sigma-2/PGRMC1 receptors mediate Abeta 42 oligomer binding and synaptotoxicity. PLoS One 2014; 9:e111899. [PMID: 25390692 PMCID: PMC4229119 DOI: 10.1371/journal.pone.0111899] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 10/02/2014] [Indexed: 12/18/2022] Open
Abstract
Amyloid beta (Abeta) 1-42 oligomers accumulate in brains of patients with Mild Cognitive Impairment (MCI) and disrupt synaptic plasticity processes that underlie memory formation. Synaptic binding of Abeta oligomers to several putative receptor proteins is reported to inhibit long-term potentiation, affect membrane trafficking and induce reversible spine loss in neurons, leading to impaired cognitive performance and ultimately to anterograde amnesia in the early stages of Alzheimer's disease (AD). We have identified a receptor not previously associated with AD that mediates the binding of Abeta oligomers to neurons, and describe novel therapeutic antagonists of this receptor capable of blocking Abeta toxic effects on synapses in vitro and cognitive deficits in vivo. Knockdown of sigma-2/PGRMC1 (progesterone receptor membrane component 1) protein expression in vitro using siRNA results in a highly correlated reduction in binding of exogenous Abeta oligomers to neurons of more than 90%. Expression of sigma-2/PGRMC1 is upregulated in vitro by treatment with Abeta oligomers, and is dysregulated in Alzheimer's disease patients' brain compared to age-matched, normal individuals. Specific, high affinity small molecule receptor antagonists and antibodies raised against specific regions on this receptor can displace synthetic Abeta oligomer binding to synaptic puncta in vitro and displace endogenous human AD patient oligomers from brain tissue sections in a dose-dependent manner. These receptor antagonists prevent and reverse the effects of Abeta oligomers on membrane trafficking and synapse loss in vitro and cognitive deficits in AD mouse models. These findings suggest sigma-2/PGRMC1 receptors mediate saturable oligomer binding to synaptic puncta on neurons and that brain penetrant, small molecules can displace endogenous and synthetic oligomers and improve cognitive deficits in AD models. We propose that sigma-2/PGRMC1 is a key mediator of the pathological effects of Abeta oligomers in AD and is a tractable target for small molecule disease-modifying therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Izzo
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Jinbin Xu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, United States of America
| | - Chenbo Zeng
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, United States of America
| | - Molly J. Kirk
- Departments of Neurology and Neuroscience, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Neurology, Northeastern University, Boston, Massachusetts, United States of America
| | - Kelsie Mozzoni
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Colleen Silky
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gilbert Rishton
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Hank Safferstein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, Missouri, United States of America
| | - Alison Goate
- Department of Psychiatry, Washington University, St. Louis, Missouri, United States of America
| | - Michael A. Cahill
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga New South Wales, Australia
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University New York, New York, United States of America
| | - Robert H. Mach
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, United States of America
| | - Rolf Craven
- Department of Molecular and Biological Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Elizabeth Head
- Department of Molecular and Biological Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Harry LeVine
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Tara L. Spires-Jones
- Departments of Neurology and Neuroscience, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The University of Edinburgh, Center for Cognitive and Neural Systems and Euan MacDonald Centre for Motorneurone Disease, Edinburgh, Scotland
| | - Susan M. Catalano
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
426
|
Renoux AJ, Carducci NM, Ahmady AA, Todd PK. Fragile X mental retardation protein expression in Alzheimer's disease. Front Genet 2014; 5:360. [PMID: 25452762 PMCID: PMC4233940 DOI: 10.3389/fgene.2014.00360] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/27/2014] [Indexed: 12/31/2022] Open
Abstract
The FMR1 protein product, FMRP, is an mRNA binding protein associated with translational inhibition of target transcripts. One FMRP target is the amyloid precursor protein (APP) mRNA, and APP levels are elevated in Fmr1 KO mice. Given that elevated APP protein expression can elicit Alzheimer's disease (AD) in patients and model systems, we evaluated whether FMRP expression might be altered in Alzheimer's autopsy brain samples and mouse models compared to controls. In a double transgenic mouse model of AD (APP/PS1), we found no difference in FMRP expression in aged AD model mice compared to littermate controls. FMRP expression was also similar in AD and control patient frontal cortex and cerebellum samples. Fragile X-associated tremor/ataxia syndrome (FXTAS) is an age-related neurodegenerative disorder caused by expanded CGG repeats in the 5' untranslated region of the FMR1 gene. Patients experience cognitive impairment and dementia in addition to motor symptoms. In parallel studies, we measured FMRP expression in cortex and cerebellum from three FXTAS patients and found reduced expression compared to both controls and Alzheimer's patient brains, consistent with animal models. We also find increased APP levels in cerebellar, but not cortical, samples of FXTAS patients compared to controls. Taken together, these data suggest that a decrease in FMRP expression is unlikely to be a primary contributor to Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Abigail J Renoux
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, MI, USA ; Department of Neurology, University of Michigan , Ann Arbor, MI, USA
| | | | - Arya A Ahmady
- Department of Neurology, University of Michigan , Ann Arbor, MI, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
427
|
Toward a unified therapeutics approach targeting putative amyloid-β oligomer receptors. Proc Natl Acad Sci U S A 2014; 111:13680-1. [PMID: 25205809 DOI: 10.1073/pnas.1414554111] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
428
|
Cissé M, Checler F. Eph receptors: new players in Alzheimer's disease pathogenesis. Neurobiol Dis 2014; 73:137-49. [PMID: 25193466 DOI: 10.1016/j.nbd.2014.08.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is devastating and leads to permanent losses of memory and other cognitive functions. Although recent genetic evidences strongly argue for a causative role of Aβ in AD onset and progression (Jonsson et al., 2012), its role in AD etiology remains a matter of debate. However, even if not the sole culprit or pathological trigger, genetic and anatomical evidences in conjunction with numerous pharmacological studies, suggest that Aβ peptides, at least contribute to the disease. How Aβ contributes to memory loss remains largely unknown. Soluble Aβ species referred to as Aβ oligomers have been shown to be neurotoxic and induce network failure and cognitive deficits in animal models of the disease. In recent years, several proteins were described as potential Aβ oligomers receptors, amongst which are the receptor tyrosine kinases of Eph family. These receptors together with their natural ligands referred to as ephrins have been involved in a plethora of physiological and pathological processes, including embryonic neurogenesis, learning and memory, diabetes, cancers and anxiety. Here we review recent discoveries on Eph receptors-mediated protection against Aβ oligomers neurotoxicity as well as their potential as therapeutic targets in AD pathogenesis.
Collapse
Affiliation(s)
- Moustapha Cissé
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| |
Collapse
|
429
|
Black SAG, Stys PK, Zamponi GW, Tsutsui S. Cellular prion protein and NMDA receptor modulation: protecting against excitotoxicity. Front Cell Dev Biol 2014; 2:45. [PMID: 25364752 PMCID: PMC4207032 DOI: 10.3389/fcell.2014.00045] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/09/2014] [Indexed: 12/25/2022] Open
Abstract
Although it is well established that misfolding of the cellular prion protein (PrPC) into the β-sheet-rich, aggregated scrapie conformation (PrPSc) causes a variety of transmissible spongiform encephalopathies (TSEs), the physiological roles of PrPC are still incompletely understood. There is accumulating evidence describing the roles of PrPC in neurodegeneration and neuroinflammation. Recently, we identified a functional regulation of NMDA receptors by PrPC that involves formation of a physical protein complex between these proteins. Excessive NMDA receptor activity during conditions such as ischemia mediates enhanced Ca2+ entry into cells and contributes to excitotoxic neuronal death. In addition, NMDA receptors and/or PrPC play critical roles in neuroinflammation and glial cell toxicity. Inhibition of NMDA receptor activity protects against PrPSc-induced neuronal death. Moreover, in mice lacking PrPC, infarct size is increased after focal cerebral ischemia, and absence of PrPC increases susceptibility of neurons to NMDA receptor-dependent death. Recently, PrPC was found to be a receptor for oligomeric beta-amyloid (Aβ) peptides, suggesting a role for PrPC in Alzheimer's disease (AD). Our recent findings suggest that Aβ peptides enhance NMDA receptor current by perturbing the normal copper- and PrPC-dependent regulation of these receptors. Here, we review evidence highlighting a role for PrPC in preventing NMDA receptor-mediated excitotoxicity and inflammation. There is a need for more detailed molecular characterization of PrPC-mediated regulation of NMDA receptors, such as determining which NMDA receptor subunits mediate pathogenic effects upon loss of PrPC-mediated regulation and identifying PrPC binding site(s) on the receptor. This knowledge will allow development of novel therapeutic interventions for not only TSEs, but also for AD and other neurodegenerative disorders involving dysfunction of PrPC.
Collapse
Affiliation(s)
- Stefanie A G Black
- Department of Physiology and Pharmacology, University of Calgary Calgary, AB, Canada ; Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada
| | - Peter K Stys
- Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada ; Department of Clinical Neurosciences, University of Calgary Calgary, AB, Canada
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, University of Calgary Calgary, AB, Canada ; Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada
| | - Shigeki Tsutsui
- Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada ; Department of Clinical Neurosciences, University of Calgary Calgary, AB, Canada
| |
Collapse
|
430
|
Van Dooren T, Princen K, De Witte K, Griffioen G. Derailed intraneuronal signalling drives pathogenesis in sporadic and familial Alzheimer's disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:167024. [PMID: 25243118 PMCID: PMC4160617 DOI: 10.1155/2014/167024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/31/2014] [Accepted: 08/03/2014] [Indexed: 02/01/2023]
Abstract
Although a wide variety of genetic and nongenetic Alzheimer's disease (AD) risk factors have been identified, their role in onset and/or progression of neuronal degeneration remains elusive. Systematic analysis of AD risk factors revealed that perturbations of intraneuronal signalling pathways comprise a common mechanistic denominator in both familial and sporadic AD and that such alterations lead to increases in Aβ oligomers (Aβo) formation and phosphorylation of TAU. Conversely, Aβo and TAU impact intracellular signalling directly. This feature entails binding of Aβo to membrane receptors, whereas TAU functionally interacts with downstream transducers. Accordingly, we postulate a positive feedback mechanism in which AD risk factors or genes trigger perturbations of intraneuronal signalling leading to enhanced Aβo formation and TAU phosphorylation which in turn further derange signalling. Ultimately intraneuronal signalling becomes deregulated to the extent that neuronal function and survival cannot be sustained, whereas the resulting elevated levels of amyloidogenic Aβo and phosphorylated TAU species self-polymerizes into the AD plaques and tangles, respectively.
Collapse
|
431
|
Haas LT, Kostylev MA, Strittmatter SM. Therapeutic molecules and endogenous ligands regulate the interaction between brain cellular prion protein (PrPC) and metabotropic glutamate receptor 5 (mGluR5). J Biol Chem 2014; 289:28460-77. [PMID: 25148681 DOI: 10.1074/jbc.m114.584342] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Soluble Amyloid-β oligomers (Aβo) can trigger Alzheimer disease (AD) pathophysiology by binding to cell surface cellular prion protein (PrP(C)). PrP(C) interacts physically with metabotropic glutamate receptor 5 (mGluR5), and this interaction controls the transmission of neurotoxic signals to intracellular substrates. Because the interruption of the signal transduction from PrP(C) to mGluR5 has therapeutic potential for AD, we developed assays to explore the effect of endogenous ligands, agonists/antagonists, and antibodies on the interaction between PrP(C) and mGluR5 in cell lines and mouse brain. We show that the PrP(C) segment of amino acids 91-153 mediates the interaction with mGluR5. Agonists of mGluR5 increase the mGluR5-PrP(C) interaction, whereas mGluR5 antagonists suppress protein association. Synthetic Aβo promotes the protein interaction in mouse brain and transfected HEK-293 cell membrane preparations. The interaction of PrP(C) and mGluR5 is enhanced dramatically in the brains of familial AD transgenic model mice. In brain homogenates with Aβo, the interaction of PrP(C) and mGluR5 is reversed by mGluR5-directed antagonists or antibodies directed against the PrP(C) segment of amino acids 91-153. Silent allosteric modulators of mGluR5 do not alter Glu or basal mGluR5 activity, but they disrupt the Aβo-induced interaction of mGluR5 with PrP(C). The assays described here have the potential to identify and develop new compounds that inhibit the interaction of PrP(C) and mGluR5, which plays a pivotal role in the pathogenesis of Alzheimer disease by transmitting the signal from extracellular Aβo into the cytosol.
Collapse
Affiliation(s)
- Laura T Haas
- From the Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06536 and the Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074 Tübingen, Germany
| | - Mikhail A Kostylev
- From the Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06536 and
| | - Stephen M Strittmatter
- From the Cellular Neuroscience, Neurodegeneration and Repair Program, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06536 and
| |
Collapse
|
432
|
Watt NT, Griffiths HH, Hooper NM. Lipid rafts: linking prion protein to zinc transport and amyloid-β toxicity in Alzheimer's disease. Front Cell Dev Biol 2014; 2:41. [PMID: 25364748 PMCID: PMC4206978 DOI: 10.3389/fcell.2014.00041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/05/2014] [Indexed: 01/01/2023] Open
Abstract
Dysregulation of neuronal zinc homeostasis plays a major role in many processes related to brain aging and neurodegenerative diseases, including Alzheimer's disease (AD). Yet, despite the critical role of zinc in neuronal function, the cellular mechanisms underpinning its homeostatic control are far from clear. We reported that the cellular prion protein (PrPC) is involved in the uptake of zinc into neurons. This PrPC-mediated zinc influx required the metal-binding octapeptide repeats in PrPC and the presence of the zinc permeable AMPA channel with which PrPC directly interacted. Together with the observation that PrPC is evolutionarily related to the ZIP family of zinc transporters, these studies indicate that PrPC plays a key role in neuronal zinc homeostasis. Therefore, PrPC could contribute to cognitive health and protect against age-related zinc dyshomeostasis but PrPC has also been identified as a receptor for amyloid-β oligomers which accumulate in the brains of those with AD. We propose that the different roles that PrPC has are due to its interaction with different ligands and/or co-receptors in lipid raft-based signaling/transport complexes.
Collapse
Affiliation(s)
- Nicole T Watt
- Division of Cardiovascular and Diabetes Research, Multidisciplinary Cardiovascular Research Centre, University of Leeds Leeds, UK
| | - Heledd H Griffiths
- Faculty of Medical and Human Sciences, Institute of Brain, Behaviour and Mental Health, University of Manchester Manchester, UK
| | - Nigel M Hooper
- Faculty of Medical and Human Sciences, Institute of Brain, Behaviour and Mental Health, University of Manchester Manchester, UK
| |
Collapse
|
433
|
Ribeiro FM, Hamilton A, Doria JG, Guimaraes IM, Cregan SP, Ferguson SS. Metabotropic glutamate receptor 5 as a potential therapeutic target in Huntington's disease. Expert Opin Ther Targets 2014; 18:1293-304. [PMID: 25118797 DOI: 10.1517/14728222.2014.948419] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a polyglutamine expansion in the amino-terminal region of the huntingtin (htt) protein, which underlies the loss of striatal and cortical neurons. Glutamate has been implicated in a number of neurodegenerative diseases, and several studies suggest that the metabotropic glutamate receptor 5 (mGluR5) may represent a target for the treatment of HD. AREAS COVERED The main goal of this review is to discuss the current data in the literature regarding the role of mGluR5 in HD and evaluate the potential of mGluR5 as a therapeutic target for the treatment of HD. mGluR5 is highly expressed in the brain regions affected in HD and is involved in movement control. Moreover, mGluR5 interacts with htt and mutated htt profoundly affects mGluR5 signaling. However, mGluR5 stimulation can activate both neuroprotective and neurotoxic signaling pathways, depending on the context of activation. EXPERT OPINION Although the data published so far strongly indicate that mGluR5 plays a major role in HD-associated neurodegeneration, htt aggregation and motor symptoms, it is not clear whether mGluR5 stimulation can diminish or intensify neuronal cell loss and HD progression. Thus, future experiments will be necessary to further investigate the outcome of drugs acting on mGluR5 for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabiola M Ribeiro
- Universidade Federal de Minas Gerais, Departamento de Bioquimica e Imunologia, ICB , Belo Horizonte 31270-901 , Brazil
| | | | | | | | | | | |
Collapse
|
434
|
Goure WF, Krafft GA, Jerecic J, Hefti F. Targeting the proper amyloid-beta neuronal toxins: a path forward for Alzheimer's disease immunotherapeutics. ALZHEIMERS RESEARCH & THERAPY 2014; 6:42. [PMID: 25045405 PMCID: PMC4100318 DOI: 10.1186/alzrt272] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Levels of amyloid-beta monomer and deposited amyloid-beta in the Alzheimer’s
disease brain are orders of magnitude greater than soluble amyloid-beta oligomer
levels. Monomeric amyloid-beta has no known direct toxicity. Insoluble fibrillar
amyloid-beta has been proposed to be an in vivo mechanism for removal of
soluble amyloid-beta and exhibits relatively low toxicity. In contrast, soluble
amyloid-beta oligomers are widely reported to be the most toxic amyloid-beta form,
both causing acute synaptotoxicity and inducing neurodegenerative processes. None of
the amyloid-beta immunotherapies currently in clinical development selectively target
soluble amyloid-beta oligomers, and their lack of efficacy is not unexpected
considering their selectivity for monomeric or fibrillar amyloid-beta (or both)
rather than soluble amyloid-beta oligomers. Because they exhibit acute,
memory-compromising synaptic toxicity and induce chronic neurodegenerative toxicity
and because they exist at very low in vivo levels in the Alzheimer’s
disease brain, soluble amyloid-beta oligomers constitute an optimal immunotherapeutic
target that should be pursued more aggressively.
Collapse
Affiliation(s)
- William F Goure
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Grant A Krafft
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Jasna Jerecic
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Franz Hefti
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| |
Collapse
|
435
|
The 37kDa/67kDa laminin receptor acts as a receptor for Aβ42 internalization. Sci Rep 2014; 4:5556. [PMID: 24990253 PMCID: PMC4080222 DOI: 10.1038/srep05556] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/13/2014] [Indexed: 12/19/2022] Open
Abstract
Neuronal loss is a major neuropathological hallmark of Alzheimer's disease (AD). The associations between soluble Aβ oligomers and cellular components cause this neurotoxicity. The 37 kDa/67 kDa laminin receptor (LRP/LR) has recently been implicated in Aβ pathogenesis. In this study the mechanism underlying the pathological role of LRP/LR was elucidated. Försters Resonance Energy Transfer (FRET) revealed that LRP/LR and Aβ form a biologically relevant interaction. The ability of LRP/LR to form stable associations with endogenously shed Aβ was confirmed by pull down assays and Aβ-ELISAs. Antibody blockade of this association significantly lowered Aβ42 induced apoptosis. Furthermore, antibody blockade and shRNA mediated downregulation of LRP/LR significantly hampered Aβ42 internalization. These results suggest that LRP/LR is a receptor for Aβ42 internalization, mediating its endocytosis and contributing to the cytotoxicity of the neuropeptide by facilitating intra-cellular Aβ42 accumulation. These findings recommend anti-LRP/LR specific antibodies and shRNAs as potential therapeutic tools for AD treatment.
Collapse
|
436
|
Stagi M, Klein ZA, Gould TJ, Bewersdorf J, Strittmatter SM. Lysosome size, motility and stress response regulated by fronto-temporal dementia modifier TMEM106B. Mol Cell Neurosci 2014; 61:226-40. [PMID: 25066864 PMCID: PMC4145808 DOI: 10.1016/j.mcn.2014.07.006] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 12/14/2022] Open
Abstract
Fronto-temporal lobar degeneration with TDP-43 (FTLD-TDP) is a fatal neurodegeneration. TMEM106B variants are linked to FTLD-TDP risk, and TMEM106B is lysosomal. Here, we focus on neuronal TMEM106B, and demonstrate co-localization and traffic with lysosomal LAMP-1. pH-sensitive reporters demonstrate that the TMEM106B C-terminus is lumenal. The TMEM106B N-terminus interacts with endosomal adaptors and other TMEM106 proteins. TMEM106B knockdown reduces neuronal lysosomal number and diameter by STED microscopy, and overexpression enlarges LAMP-positive structures. Reduction of TMEM106B increases axonally transported lysosomes, while TMEM106B elevation inhibits transport and yields large lysosomes in the soma. TMEM106B overexpression alters lysosomal stress signaling, causing a translocation of the mTOR-sensitive transcription factor, TFEB, to neuronal nuclei. TMEM106B loss-of-function delays TFEB translocation after Torin-1-induced stress. Enlarged TMEM106B-overexpressing lysosomes maintain organelle integrity longer after lysosomal photodamage than do control lysosomes, while small TMEM106B-knockdown lysosomes are more sensitive to illumination. Thus, neuronal TMEM106B plays a central role in regulating lysosomal size, motility and responsiveness to stress, highlighting the possible role of lysosomal biology in FTLD-TDP.
Collapse
Affiliation(s)
- Massimiliano Stagi
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA; Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Zoe A Klein
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA; Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Travis J Gould
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joerg Bewersdorf
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, CT 06536, USA; Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
437
|
Aβ induces its own prion protein N-terminal fragment (PrPN1)–mediated neutralization in amorphous aggregates. Neurobiol Aging 2014; 35:1537-48. [DOI: 10.1016/j.neurobiolaging.2014.02.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/31/2014] [Accepted: 02/01/2014] [Indexed: 01/24/2023]
|
438
|
Nava-Mesa MO, Jiménez-Díaz L, Yajeya J, Navarro-Lopez JD. GABAergic neurotransmission and new strategies of neuromodulation to compensate synaptic dysfunction in early stages of Alzheimer's disease. Front Cell Neurosci 2014; 8:167. [PMID: 24987334 PMCID: PMC4070063 DOI: 10.3389/fncel.2014.00167] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/02/2014] [Indexed: 01/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline, brain atrophy due to neuronal and synapse loss, and formation of two pathological lesions: extracellular amyloid plaques, composed largely of amyloid-beta peptide (Aβ), and neurofibrillary tangles formed by intracellular aggregates of hyperphosphorylated tau protein. Lesions mainly accumulate in brain regions that modulate cognitive functions such as the hippocampus, septum or amygdala. These brain structures have dense reciprocal glutamatergic, cholinergic, and GABAergic connections and their relationships directly affect learning and memory processes, so they have been proposed as highly susceptible regions to suffer damage by Aβ during AD course. Last findings support the emerging concept that soluble Aβ peptides, inducing an initial stage of synaptic dysfunction which probably starts 20–30 years before the clinical onset of AD, can perturb the excitatory–inhibitory balance of neural circuitries. In turn, neurotransmission imbalance will result in altered network activity that might be responsible of cognitive deficits in AD. Therefore, Aβ interactions on neurotransmission systems in memory-related brain regions such as amygdaloid complex, medial septum or hippocampus are critical in cognitive functions and appear as a pivotal target for drug design to improve learning and dysfunctions that manifest with age. Since treatments based on glutamatergic and cholinergic pharmacology in AD have shown limited success, therapies combining modulators of different neurotransmission systems including recent findings regarding the GABAergic system, emerge as a more useful tool for the treatment, and overall prevention, of this dementia. In this review, focused on inhibitory systems, we will analyze pharmacological strategies to compensate neurotransmission imbalance that might be considered as potential therapeutic interventions in AD.
Collapse
Affiliation(s)
| | - Lydia Jiménez-Díaz
- Neurophysiology and Behavior Lab, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha Ciudad Real, Spain
| | - Javier Yajeya
- Department of Physiology and Pharmacology, University of Salamanca Salamanca, Spain
| | - Juan D Navarro-Lopez
- Neurophysiology and Behavior Lab, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, University of Castilla-La Mancha Ciudad Real, Spain
| |
Collapse
|
439
|
Petrov D, Pedros I, de Lemos ML, Pallàs M, Canudas AM, Lazarowski A, Beas-Zarate C, Auladell C, Folch J, Camins A. Mavoglurant as a treatment for Parkinson's disease. Expert Opin Investig Drugs 2014; 23:1165-79. [PMID: 24960254 DOI: 10.1517/13543784.2014.931370] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION A major unresolved issue in the Parkinson's disease (PD) treatment is the development of l-DOPA-induced dyskinesias (LIDs) as a side effect of chronic L-DOPA administration. Currently, LIDs are managed in part by reducing the L-DOPA dose or by the administration of amantadine. However, this treatment is only partially effective. A potential strategy, currently under investigation, is the coadministration of metabotropic glutamate receptor 5 (mGluR5) negative allosteric modulators (NAMs) and L-DOPA; a treatment that results in the improvement of dyskinesia symptoms and that permits reductions in l-DOPA dosage frequency. AREAS COVERED The authors examine the role of mGluR5 in the pathophysiology of PD and the potential use of mGluR5 NAM as an adjuvant therapy together with a primary treatment with L-DOPA. Specifically, the authors look at the mavoglurant therapy and the evidence presented through preclinical and clinical trials. EXPERT OPINION Interaction between mGluR5 NAM and L-DOPA is an area of interest in PD research as concomitant treatment results in the improvement of LID symptoms in humans, thus enhancing the patient's quality of life. However, few months ago, Novartis decided to discontinue clinical trials of mavoglurant for the treatment of LID, due to the lack of efficacy demonstrated in trials NCT01385592 and NCT01491529, although no safety concerns were involved in this decision. Nevertheless, the potential application of mGluR5 antagonists as neuroprotective agents must be considered and further studies are warranted to better investigate their potential.
Collapse
Affiliation(s)
- Dmitry Petrov
- Universitat de Barcelona, Institut de Biomedicina (IBUB), Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Unitat de Farmacologia I Farmacognòsia, Facultat de Farmàcia , Barcelona, Avda/Joan XXIII , Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
440
|
Hirsch TZ, Hernandez-Rapp J, Martin-Lannerée S, Launay JM, Mouillet-Richard S. PrP(C) signalling in neurons: from basics to clinical challenges. Biochimie 2014; 104:2-11. [PMID: 24952348 DOI: 10.1016/j.biochi.2014.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/10/2014] [Indexed: 01/05/2023]
Abstract
The cellular prion protein PrP(C) was identified over twenty-five years ago as the normal counterpart of the scrapie prion protein PrP(Sc), itself the main if not the sole component of the infectious agent at the root of Transmissible Spongiform Encephalopathies (TSEs). PrP(C) is a ubiquitous cell surface protein, abundantly expressed in neurons, which constitute the targets of PrP(Sc)-mediated toxicity. Converging evidence have highlighted that neuronal, GPI-anchored PrP(C) is absolutely required for prion-induced neuropathogenesis, which warrants investigating into the normal function exerted by PrP(C) in a neuronal context. It is now well-established that PrP(C) can serve as a cell signalling molecule, able to mobilize transduction cascades in response to interactions with partners. This function endows PrP(C) with the capacity to participate in multiple neuronal processes, ranging from survival to synaptic plasticity. A diverse array of data have allowed to shed light on how this function is corrupted by PrP(Sc). Recently, amyloid Aβ oligomers, whose accumulation is associated with Alzheimer's disease (AD), were shown to similarly instigate toxic events by deviating PrP(C)-mediated signalling. Here, we provide an overview of the various signal transduction cascades ascribed to PrP(C) in neurons, summarize how their subversion by PrP(Sc) or Aβ oligomers contributes to TSE or AD neuropathogenesis and discuss the ensuing clinical implications.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France
| | - Julia Hernandez-Rapp
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France; Université Paris Sud 11, ED419 Biosigne, 91400 Orsay, France
| | - Séverine Martin-Lannerée
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France
| | - Jean-Marie Launay
- AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 Hôpital Lariboisière, 75010 Paris, France; Pharma Research Department, F. Hoffmann-La-Roche Ltd., CH-4070 Basel, Switzerland
| | - Sophie Mouillet-Richard
- INSERM UMR-S1124, 75006 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124, 75006 Paris, France.
| |
Collapse
|
441
|
Brain Na(+), K(+)-ATPase Activity In Aging and Disease. INTERNATIONAL JOURNAL OF BIOMEDICAL SCIENCE : IJBS 2014; 10:85-102. [PMID: 25018677 PMCID: PMC4092085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/05/2014] [Indexed: 11/15/2022]
Abstract
Na(+)/K(+) pump or sodium- and potassium-activated adenosine 5'-triphosphatase (Na(+), K(+)-ATPase), its enzymatic version, is a crucial protein responsible for the electrochemical gradient across the cell membranes. It is an ion transporter, which in addition to exchange cations, is the ligand for cardenolides. This enzyme regulates the entry of K(+) with the exit of Na(+) from cells, being the responsible for Na(+)/K(+) equilibrium maintenance through neuronal membranes. This transport system couples the hydrolysis of one molecule of ATP to exchange three sodium ions for two potassium ions, thus maintaining the normal gradient of these cations in animal cells. Oxidative metabolism is very active in brain, where large amounts of chemical energy as ATP molecules are consumed, mostly required for the maintenance of the ionic gradients that underlie resting and action potentials which are involved in nerve impulse propagation, neurotransmitter release and cation homeostasis. Protein phosphorylation is a key process in biological regulation. At nervous system level, protein phosphorylation is the major molecular mechanism through which the function of neural proteins is modulted in response to extracellular signals, including the response to neurotransmitter stimuli. It is the major mechanism of neural plasticity, including memory processing. The phosphorylation of Na(+), K(+)-ATPase catalytic subunit inhibits enzyme activity whereas the inhibition of protein kinase C restores the enzyme activity. The dephosphorylation of neuronal Na(+), K(+)-ATPase is mediated by calcineurin, a serine / threonine phosphatase. The latter enzyme is involved in a wide range of cellular responses to Ca(2+) mobilizing signals, in the regulation of neuronal excitability by controlling the activity of ion channels, in the release of neurotransmitters and hormones, as well as in synaptic plasticity and gene transcription. In the present article evidence showing Na(+), K(+)-ATPase involvement in signaling pathways, enzyme changes in diverse neurological diseases as well as during aging, have been summarized. Issues refer mainly to Na(+), K(+)-ATPase studies in ischemia, brain injury, depression and mood disorders, mania, stress, Alzheimer´s disease, learning and memory, and neuronal hyperexcitability and epilepsy.
Collapse
|
442
|
Hamilton A, Esseltine JL, DeVries RA, Cregan SP, Ferguson SSG. Metabotropic glutamate receptor 5 knockout reduces cognitive impairment and pathogenesis in a mouse model of Alzheimer's disease. Mol Brain 2014; 7:40. [PMID: 24886239 PMCID: PMC4050478 DOI: 10.1186/1756-6606-7-40] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 05/23/2014] [Indexed: 01/09/2023] Open
Abstract
Background Alzheimer’s disease (AD) pathology occurs in part as the result of excessive production of β-amyloid (Aβ). Metabotropic glutamate receptor 5 (mGluR5) is now considered a receptor for Aβ and consequently contributes to pathogenic Aβ signaling in AD. Results Genetic deletion of mGluR5 rescues the spatial learning deficits observed in APPswe/PS1ΔE9 AD mice. Moreover, both Aβ oligomer formation and Aβ plaque number are reduced in APPswe/PS1ΔE9 mice lacking mGluR5 expression. In addition to the observed increase in Aβ oligomers and plaques in APPswe/PS1ΔE9 mice, we found that both mTOR phosphorylation and fragile X mental retardation protein (FMRP) expression were increased in these mice. Genetic deletion of mGluR5 reduced Aβ oligomers, plaques, mTOR phosphorylation and FMRP expression in APPswe/PS1ΔE9 mice. Conclusions Thus, we propose that Aβ activation of mGluR5 appears to initiate a positive feedback loop resulting in increased Aβ formation and AD pathology in APPswe/PS1ΔE9 mice via mechanism that is regulated by FMRP.
Collapse
Affiliation(s)
| | | | | | | | - Stephen S G Ferguson
- The J, Allyn Taylor Centre for Cell Biology, Robarts Research Institute, The University of Western Ontario, 100 Perth Dr, London, Ontario N6A 5 K8, Canada.
| |
Collapse
|
443
|
A role for dendritic mGluR5-mediated local translation of Arc/Arg3.1 in MEF2-dependent synapse elimination. Cell Rep 2014; 7:1589-1600. [PMID: 24857654 DOI: 10.1016/j.celrep.2014.04.035] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 03/20/2014] [Accepted: 04/18/2014] [Indexed: 02/07/2023] Open
Abstract
Experience refines synaptic connectivity through neural activity-dependent regulation of transcription factors. Although activity-dependent regulation of transcription factors has been well described, it is unknown whether synaptic activity and local, dendritic regulation of the induced transcripts are necessary for mammalian synaptic plasticity in response to transcription factor activation. Neuronal depolarization activates the myocyte enhancer factor 2 (MEF2) family of transcription factors that suppresses excitatory synapse number. We report that activation of metabotropic glutamate receptor 5 (mGluR5) on the dendrites, but not cell soma, of hippocampal CA1 neurons is required for MEF2-induced functional and structural synapse elimination. We present evidence that mGluR5 is necessary for synapse elimination to stimulate dendritic translation of the MEF2 target gene Arc/Arg3.1. Activity-regulated cytoskeletal-associated protein (Arc) is required for MEF2-induced synapse elimination, where it plays an acute, cell-autonomous, and postsynaptic role. This work reveals a role for dendritic activity in local translation of specific transcripts in synapse refinement.
Collapse
|
444
|
Spires-Jones TL, Hyman BT. The intersection of amyloid beta and tau at synapses in Alzheimer's disease. Neuron 2014; 82:756-71. [PMID: 24853936 PMCID: PMC4135182 DOI: 10.1016/j.neuron.2014.05.004] [Citation(s) in RCA: 819] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2014] [Indexed: 02/07/2023]
Abstract
The collapse of neural networks important for memory and cognition, including death of neurons and degeneration of synapses, causes the debilitating dementia associated with Alzheimer's disease (AD). We suggest that synaptic changes are central to the disease process. Amyloid beta and tau form fibrillar lesions that are the classical hallmarks of AD. Recent data indicate that both molecules may have normal roles at the synapse, and that the accumulation of soluble toxic forms of the proteins at the synapse may be on the critical path to neurodegeneration. Further, the march of neurofibrillary tangles through brain circuits appears to take advantage of recently described mechanisms of transsynaptic spread of pathological forms of tau. These two key phenomena, synapse loss and the spread of pathology through the brain via synapses, make it critical to understand the physiological and pathological roles of amyloid beta and tau at the synapse.
Collapse
Affiliation(s)
- Tara L Spires-Jones
- Centre for Cognitive and Neural Systems, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ, UK; The Euan MacDonald Centre, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ, UK.
| | - Bradley T Hyman
- Massachusetts General Hospital, Harvard Medical School, Neurology, 114 16(th) Street, Charlestown, MA 02129, USA.
| |
Collapse
|
445
|
Overk CR, Cartier A, Shaked G, Rockenstein E, Ubhi K, Spencer B, Price DL, Patrick C, Desplats P, Masliah E. Hippocampal neuronal cells that accumulate α-synuclein fragments are more vulnerable to Aβ oligomer toxicity via mGluR5--implications for dementia with Lewy bodies. Mol Neurodegener 2014; 9:18. [PMID: 24885390 PMCID: PMC4041038 DOI: 10.1186/1750-1326-9-18] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 05/13/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In dementia with Lewy bodies (DLB) abnormal interactions between α-synuclein (α-syn) and beta amyloid (Aβ) result in selective degeneration of neurons in the neocortex, limbic system and striatum. However, factors rendering these neurons selectively vulnerable have not been fully investigated. The metabotropic glutamate receptor 5 (mGluR5) has been shown to be up regulated in DLB and might play a role as a mediator of the neurotoxic effects of Aβ and α-syn in vulnerable neuronal populations. In this context, the main objective of the present study was to investigate the role of mGluR5 as a mediator of the neurotoxic effects of α-syn and Aβ in the hippocampus. RESULTS We generated double transgenic mice over-expressing amyloid precursor protein (APP) and α-syn under the mThy1 cassette and investigated the relationship between α-syn cleavage, Aβ, mGluR5 and neurodegeneration in the hippocampus. We found that compared to the single tg mice, the α-syn/APP tg mice displayed greater accumulation of α-syn and mGluR5 in the CA3 region of the hippocampus compared to the CA1 and other regions. This was accompanied by loss of CA3 (but not CA1) neurons in the single and α-syn/APP tg mice and greater loss of MAP 2 and synaptophysin in the CA3 in the α-syn/APP tg. mGluR5 gene transfer using a lentiviral vector into the hippocampus CA1 region resulted in greater α-syn accumulation and neurodegeneration in the single and α-syn/APP tg mice. In contrast, silencing mGluR5 with a lenti-shRNA protected neurons in the CA3 region of tg mice. In vitro, greater toxicity was observed in primary hippocampal neuronal cultures treated with Aβ oligomers and over-expressing α-syn; this effect was attenuated by down-regulating mGluR5 with an shRNA lentiviral vector. In α-syn-expressing neuronal cells lines, Aβ oligomers promoted increased intracellular calcium levels, calpain activation and α-syn cleavage resulting in caspase-3-dependent cell death. Treatment with pharmacological mGluR5 inhibitors such as 2-Methyl-6-(phenylethynyl)pyridine (MPEP) and 3-((2-Methyl-4-thiazolyl)ethynyl)pyridine (MTEP) attenuated the toxic effects of Aβ in α-syn-expressing neuronal cells. CONCLUSIONS Together, these results support the possibility that vulnerability of hippocampal neurons to α-syn and Aβ might be mediated via mGluR5. Moreover, therapeutical interventions targeting mGluR5 might have a role in DLB.
Collapse
Affiliation(s)
- Cassia R Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Anna Cartier
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Gideon Shaked
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Kiren Ubhi
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Brian Spencer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Christina Patrick
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Paula Desplats
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
446
|
Xia D, Götz J. Premature lethality, hyperactivity, and aberrant phosphorylation in transgenic mice expressing a constitutively active form of Fyn. Front Mol Neurosci 2014; 7:40. [PMID: 24860422 PMCID: PMC4026715 DOI: 10.3389/fnmol.2014.00040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/22/2014] [Indexed: 01/10/2023] Open
Abstract
The kinase Fyn, the microtubule-associated protein tau and the peptide amyloid-β (Aβ) constitute a toxic triad in Alzheimer's disease (AD). Tau's subcellular localization is mainly regulated by phosphorylation whereas Fyn's localization is dictated by palmitoylation targeting it to the plasma membrane in a reversible manner. We have previously shown that tau is required for Fyn to be targeted to the dendritic spine. We had also shown that a truncated form of tau (Δtau) that accumulates in the cell soma is capable of trapping Fyn and preventing it from entering the spine. Here we determined that palmitoylation is required for Fyn's membrane and spine localization. We further evaluated the functional consequences of neuronal over-expression of the constitutively active Y531F mutant form of Fyn (FynCA) in transgenic mice. We found that the FynCA transgenic mice displayed a reduced weight, a massively reduced lifespan and a high level of hyperactivity. The lifespan of the FynCA mice was only slightly extended by crossing them with Δtau transgenic mice, possibly reflecting differences in expression patterns of the transgenes and high levels of transgenic FynCA compared to endogenous Fyn. Analysis of synaptosomes revealed that FynCA accumulated at high levels in the spine, resulting in increased levels of the NMDA receptor subunit NR2b phosphorylated at residue Y1472. Tau was strongly phosphorylated at the AT8 epitope S202/T205 as shown by Western blot and immunohistochemistry indicating that an increased tyrosine kinase activity of Fyn has down-stream consequences for serine/threonine-directed phosphorylation.
Collapse
Affiliation(s)
- Di Xia
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland Brisbane, QLD, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland Brisbane, QLD, Australia
| |
Collapse
|
447
|
Walsh KP, Minamide LS, Kane SJ, Shaw AE, Brown DR, Pulford B, Zabel MD, Lambeth JD, Kuhn TB, Bamburg JR. Amyloid-β and proinflammatory cytokines utilize a prion protein-dependent pathway to activate NADPH oxidase and induce cofilin-actin rods in hippocampal neurons. PLoS One 2014; 9:e95995. [PMID: 24760020 PMCID: PMC3997518 DOI: 10.1371/journal.pone.0095995] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 04/02/2014] [Indexed: 11/18/2022] Open
Abstract
Neurites of neurons under acute or chronic stress form bundles of filaments (rods) containing 1∶1 cofilin∶actin, which impair transport and synaptic function. Rods contain disulfide cross-linked cofilin and are induced by treatments resulting in oxidative stress. Rods form rapidly (5-30 min) in >80% of cultured hippocampal or cortical neurons treated with excitotoxic levels of glutamate or energy depleted (hypoxia/ischemia or mitochondrial inhibitors). In contrast, slow rod formation (50% of maximum response in ∼6 h) occurs in a subpopulation (∼20%) of hippocampal neurons upon exposure to soluble human amyloid-β dimer/trimer (Aβd/t) at subnanomolar concentrations. Here we show that proinflammatory cytokines (TNFα, IL-1β, IL-6) also induce rods at the same rate and within the same neuronal population as Aβd/t. Neurons from prion (PrP(C))-null mice form rods in response to glutamate or antimycin A, but not in response to proinflammatory cytokines or Aβd/t. Two pathways inducing rod formation were confirmed by demonstrating that NADPH-oxidase (NOX) activity is required for prion-dependent rod formation, but not for rods induced by glutamate or energy depletion. Surprisingly, overexpression of PrP(C) is by itself sufficient to induce rods in over 40% of hippocampal neurons through the NOX-dependent pathway. Persistence of PrP(C)-dependent rods requires the continuous activity of NOX. Removing inducers or inhibiting NOX activity in cells containing PrP(C)-dependent rods causes rod disappearance with a half-life of about 36 min. Cofilin-actin rods provide a mechanism for synapse loss bridging the amyloid and cytokine hypotheses for Alzheimer disease, and may explain how functionally diverse Aβ-binding membrane proteins induce synaptic dysfunction.
Collapse
Affiliation(s)
- Keifer P. Walsh
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Sarah J. Kane
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, Colorado, United States of America
| | - Alisa E. Shaw
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Bruce Pulford
- Prion Research Center, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Mark D. Zabel
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, Colorado, United States of America
- Prion Research Center, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - J. David Lambeth
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Thomas B. Kuhn
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Chemistry and Biochemistry, University of Alaska, Fairbanks, Alaska, United States of America
| | - James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
448
|
Overk CR, Masliah E. Pathogenesis of synaptic degeneration in Alzheimer's disease and Lewy body disease. Biochem Pharmacol 2014; 88:508-16. [PMID: 24462903 PMCID: PMC3973539 DOI: 10.1016/j.bcp.2014.01.015] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/10/2014] [Accepted: 01/14/2014] [Indexed: 12/15/2022]
Abstract
Considerable progress has been made in the past few years in the fight against Alzheimer's disease (AD) and Parkinson's disease (PD). Neuropathological studies in human brains and experimental in vivo and in vitro models support the notion that synapses are affected even at the earliest stages of the neurodegenerative process. The objective of this manuscript is to review some of the mechanisms of synaptic damage in AD and PD. Some lines of evidence support the notion that oligomeric neurotoxic species of amyloid β, α-synuclein, and Tau might contribute to the pathogenesis of synaptic failure at early stages of the diseases. The mechanisms leading to synaptic damage by oligomers might involve dysregulation of glutamate receptors and scaffold molecules that results in alterations in the axonal transport of synaptic vesicles and mitochondria that later on lead to dendritic and spine alterations, axonal dystrophy, and eventually neuronal loss. However, while some studies support a role of oligomers, there is an ongoing debate as to the exact nature of the toxic species. Given the efforts toward earlier clinical and preclinical diagnosis of these disorders, understanding the molecular and cellular mechanisms of synaptic degeneration is crucial toward developing specific biomarkers and new therapies targeting the synaptic apparatus of vulnerable neurons.
Collapse
Affiliation(s)
- Cassia R Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92039, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92039, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92039, USA.
| |
Collapse
|
449
|
Robinson SW, Nugent ML, Dinsdale D, Steinert JR. Prion protein facilitates synaptic vesicle release by enhancing release probability. Hum Mol Genet 2014; 23:4581-96. [PMID: 24722203 PMCID: PMC4119408 DOI: 10.1093/hmg/ddu171] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The cellular prion protein (PrP(C)) has been implicated in several neurodegenerative diseases as a result of protein misfolding. In humans, prion disease occurs typically with a sporadic origin where uncharacterized mechanisms induce spontaneous PrP(C) misfolding leading to neurotoxic PrP-scrapie formation (PrP(SC)). The consequences of misfolded PrP(C) signalling are well characterized but little is known about the physiological roles of PrP(C) and its involvement in disease. Here we investigated wild-type PrP(C) signalling in synaptic function as well as the effects of a disease-relevant mutation within PrP(C) (proline-to-leucine mutation at codon 101). Expression of wild-type PrP(C) at the Drosophila neuromuscular junction leads to enhanced synaptic responses as detected in larger miniature synaptic currents which are caused by enlarged presynaptic vesicles. The expression of the mutated PrP(C) leads to reduction of both parameters compared with wild-type PrP(C). Wild-type PrP(C) enhances synaptic release probability and quantal content but reduces the size of the ready-releasable vesicle pool. Partially, these changes are not detectable following expression of the mutant PrP(C). A behavioural test revealed that expression of either protein caused an increase in locomotor activities consistent with enhanced synaptic release and stronger muscle contractions. Both proteins were sensitive to proteinase digestion. These data uncover new functions of wild-type PrP(C) at the synapse with a disease-relevant mutation in PrP(C) leading to diminished functional phenotypes. Thus, our data present essential new information possibly related to prion pathogenesis in which a functional synaptic role of PrP(C) is compromised due to its advanced conversion into PrP(SC) thereby creating a lack-of-function scenario.
Collapse
Affiliation(s)
- Susan W Robinson
- MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| | - Marie L Nugent
- MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK Department of Genetics, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - David Dinsdale
- MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| | - Joern R Steinert
- MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| |
Collapse
|
450
|
Cochran JN, Hall AM, Roberson ED. The dendritic hypothesis for Alzheimer's disease pathophysiology. Brain Res Bull 2014; 103:18-28. [PMID: 24333192 PMCID: PMC3989444 DOI: 10.1016/j.brainresbull.2013.12.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 11/28/2013] [Accepted: 12/02/2013] [Indexed: 01/02/2023]
Abstract
Converging evidence indicates that processes occurring in and around neuronal dendrites are central to the pathogenesis of Alzheimer's disease. These data support the concept of a "dendritic hypothesis" of AD, closely related to the existing synaptic hypothesis. Here we detail dendritic neuropathology in the disease and examine how Aβ, tau, and AD genetic risk factors affect dendritic structure and function. Finally, we consider potential mechanisms by which these key drivers could affect dendritic integrity and disease progression. These dendritic mechanisms serve as a framework for therapeutic target identification and for efforts to develop disease-modifying therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- J Nicholas Cochran
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Alicia M Hall
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|